Formulations Of Brincidofovir

KABIR; Mohammed Anowrul ;   et al.

Patent Application Summary

U.S. patent application number 16/722699 was filed with the patent office on 2020-05-07 for formulations of brincidofovir. The applicant listed for this patent is Chimerix, Inc.. Invention is credited to Irma Marisa GROSSI, Mohammed Anowrul KABIR, Odin Johann NADERER.

Application Number20200138835 16/722699
Document ID /
Family ID59313330
Filed Date2020-05-07

View All Diagrams
United States Patent Application 20200138835
Kind Code A1
KABIR; Mohammed Anowrul ;   et al. May 7, 2020

FORMULATIONS OF BRINCIDOFOVIR

Abstract

Disclosed are formulations comprising brincidofovir. The formulations can be intravenous formulations. The formulations can be used in cases where a subject experiences gastrointestinal issues in response to oral administration of brincidofovir. The formulations can also be used in cases where oral administration of drug is not possible due to underlying conditions or concern around inadequate oral absorption.


Inventors: KABIR; Mohammed Anowrul; (Cary, NC) ; NADERER; Odin Johann; (Chapel Hill, NC) ; GROSSI; Irma Marisa; (Durham, NC)
Applicant:
Name City State Country Type

Chimerix, Inc.

Durham

NC

US
Family ID: 59313330
Appl. No.: 16/722699
Filed: December 20, 2019

Related U.S. Patent Documents

Application Number Filing Date Patent Number
15636393 Jun 28, 2017
16722699
62512825 May 31, 2017
62507397 May 17, 2017
62465053 Feb 28, 2017
62446213 Jan 13, 2017
62394665 Sep 14, 2016
62355844 Jun 28, 2016

Current U.S. Class: 1/1
Current CPC Class: A61K 31/675 20130101; A61K 47/26 20130101; A61K 47/18 20130101; A61P 31/22 20180101; Y02A 50/467 20180101; Y02A 50/30 20180101; A61K 9/08 20130101; A61P 31/12 20180101; A61K 9/19 20130101; A61P 31/20 20180101; A61K 47/183 20130101; A61K 9/0019 20130101; A61P 31/14 20180101
International Class: A61K 31/675 20060101 A61K031/675; A61K 9/00 20060101 A61K009/00; A61K 9/19 20060101 A61K009/19; A61K 47/18 20060101 A61K047/18; A61K 47/26 20060101 A61K047/26; A61K 9/08 20060101 A61K009/08

Claims



1. A lyophilized powder comprising: about 13-19% by weight brincidofovir; about 48%-65% by weight mannitol; and about 22-33% by weight arginine.

2. The lyophilized powder of claim 1, comprising: about 13% by weight brincidofovir; about 65% by weight mannitol; and about 22% by weight arginine.

3. The lyophilized powder of claim 1, comprising: about 19% by weight brincidofovir; about 48% by weight mannitol; and about 33% by weight arginine.

4. The lyophilized powder of claim 1, comprising: about 10 mg brincidofovir; about 50 mg mannitol; and about 17.4 mg arginine.

5. The lyophilized powder of claim 1, comprising: about 10 mg brincidofovir; about 25 mg mannitol; and about 17.4 mg arginine.

6. A method of treating a viral infection in a subject in need thereof, the method comprising: (a) dissolving the lyophilized powder of claim 1 in aqueous media to produce an aqueous pharmaceutical composition; and (b) administering to the subject an effective amount of the aqueous pharmaceutical composition.

7. The method of claim 6, wherein the viral infection is selected from polyomavirus, papillomavirus, herpes virus, adenovirus, Epstein-Barr virus, cytomegalovirus, Hepatitis B virus, Hepatitis C virus, varicella zoster virus, adenovirus, poxvirus, or a combination thereof.

8. The method of claim 6, wherein administration to the subject does not result in hemolysis.

9. The method of claim 6, wherein administration to the subject does not result in gastrointestinal toxicity.

10. The method of claim 6, wherein the aqueous media is a sugar alcohol solution, an aqueous sugar solution, Ringer's solution or an aqueous salt solution.

11. The method of claim 10, wherein the aqueous media is an aqueous sugar solution.

12. The method of claim 11, wherein the aqueous sugar solution is about 5% dextrose by weight.

13. The method of claim 11, wherein the lyophilized powder is dissolved in about 100 mL of aqueous sugar solution.

14. The method of claim 11, wherein the lyophilized powder is dissolved in about 200 mL of aqueous sugar solution.

15. The method of claim 6, wherein the aqueous pharmaceutical composition comprises: brincidofovir at a concentration of about 1.0 mg/mL; mannitol at a concentration of about 2.5 mg/mL to about 5 mg/mL; L-arginine at a concentration of about 1.74 mg/mL; and dextrose at a concentration of about 50 mg/mL; wherein the pH of the composition is about 8.0.

16. The method of claim 6, wherein the aqueous pharmaceutical composition comprises: brincidofovir at a concentration of about 1.78 mM; mannitol at a concentration of about 13.75 mM to about 27.5 mM; L-arginine at a concentration of about 10 mM; and dextrose at a concentration of about 287 mM; wherein the pH of the composition is about 8.0.

17. The method of claim 6, wherein the aqueous pharmaceutical composition comprises: brincidofovir at a concentration of about 0.5 mg/mL; mannitol at a concentration of about 1.25 mg/mL to about 2.5 mg/mL; L-arginine at a concentration of about 0.87 mg/mL; and dextrose at a concentration of about 50 mg/mL; wherein the pH of the composition is about 8.0.

18. The method of claim 6, wherein the aqueous pharmaceutical composition comprises: brincidofovir at a concentration of about 0.89 mM; mannitol at a concentration of about 6.85 mM to about 13.7 mM; L-arginine at a concentration of about 5 mM; and dextrose at a concentration of about 287 mM; wherein the pH of the composition is about 8.0.

19. The method of claim 6, wherein the aqueous pharmaceutical composition comprises: about 100 mg brincidofovir; about 250 mg to about 500 mg mannitol; about 174 mg arginine; about 5 g dextrose; and about 100 mL water; wherein the pH of the solution is about 8.0.

20. The method of claim 6, wherein the aqueous pharmaceutical composition comprises: about 200 mg brincidofovir; about 500 mg to about 1000 mg mannitol; about 348 mg arginine; about 10 g dextrose; and about 200 mL water; wherein the pH of the solution is about 8.0.

21. The method of claim 6, wherein the aqueous pharmaceutical composition comprises: about 50 mg brincidofovir; about 125 mg to about 250 mg mannitol; about 87 mg arginine; about 5 g dextrose; and about 100 mL water; wherein the pH of the solution is about 8.0.

22. The method of claim 6, wherein the aqueous pharmaceutical composition comprises: about 100 mg brincidofovir; about 250 mg to about 500 mg mannitol; about 174 mg arginine; about 10 g dextrose; and about 200 mL water; wherein the pH of the solution is about 8.0.

23. The method of claim 6, wherein the aqueous pharmaceutical composition is suitable for intravenous administration.

24. The pharmaceutical composition of claim 23, wherein the aqueous pharmaceutical composition is sterile.

25. A method of preparing the lyophilized powder of claim 1, comprising (a) preparing an aqueous solution comprising brincidofovir at a concentration of about 10 mg/mL; mannitol at a concentration of about 25-50 mg/mL; and L-arginine at a concentration of about 17.4 mg/mL; wherein the pH of the composition is about 8.0; and (b) lyophilizing the aqueous solution.
Description



CROSS-REFERENCE TO RELATED APPLICATIONS

[0001] This application is a division of U.S. patent application Ser. No. 15/636,393, filed Jun. 28, 2017, currently pending, which claims priority to, and benefit of: U.S. Provisional Patent Application No. 62/355,844, filed Jun. 28, 2016; U.S. Provisional Patent Application No. 62/394,665, filed Sep. 14, 2016; U.S. Provisional Patent Application No. 62/446,213, filed Jan. 13, 2017; U.S. Provisional Patent Application No. 62/465,053, filed Feb. 28, 2017; U.S. Provisional Patent Application No. 62/507,397, filed May 17, 2017; and U.S. Provisional Patent Application No. 62/512,825, filed May 31, 2017, the contents of each of which are incorporated by reference in their entireties.

FIELD

[0002] The present disclosure relates to pharmaceutical compositions (e.g., lyophilized and/or aqueous compositions) comprising brincidofovir and methods of use thereof.

BACKGROUND

[0003] Brincidofovir (BCV, CMX001) is an orally bioavailable, lipid acyclic nucleoside phosphonate that is converted intracellularly into the active antiviral cidofovir diphosphate (CDV-PP). Brincidofovir has broad spectrum antiviral activity against double-stranded DNA viruses. The structure of brincidofovir is shown below:

##STR00001##

SUMMARY

[0004] The present disclosure presents compositions comprising brincidofovir and methods of using the same. The compositions can be lyophilized (e.g., as a powder) for long-term storage. The lyophilized formulations can be reconstituted (e.g., in an aqueous sugar solution) as biocompatible formulations for intravenous (IV) administration (e.g., to a subject in need thereof).

[0005] In one aspect, the present disclosure provides a pharmaceutical composition, comprising: brincidofovir; a bulking agent; a buffer; and water; wherein the pH of the composition is about 8.0.

[0006] In some embodiments, the bulking agent is mannitol or sucrose. In some embodiments, the bulking agent is mannitol. In some embodiments, the buffer is sodium phosphate, L-arginine, or tromethamine. In some embodiments, the buffer is L-arginine. In some embodiments, the brincidofovir is present at a concentration of about 10.0 mg/mL. In some embodiments, the bulking agent is present at a concentration of about 2.5-9% (w/v). In some embodiments, the bulking agent is present at a concentration of about 2.5% (w/v). In some embodiments, the bulking agent is present at a concentration of about 5% (w/v). In some embodiments, the buffer is present at a concentration of about 100-200 mM. In some embodiments, the buffer is present at a concentration of about 100 mM. In some embodiments, the pH is adjusted using HCl and/or NaOH.

[0007] In some embodiments, the pharmaceutical composition comprises: brincidofovir at a concentration of about 10.0 mg/mL; mannitol at a concentration of about 25-50 mg/mL; L-arginine at a concentration of about 17.4 mg/mL; and water; wherein the pH of the composition is about 8.0.

[0008] In some embodiments, the pharmaceutical composition comprises: brincidofovir at a concentration of about 17.8 mM; mannitol at a concentration of about 137.5-275 mM; L-arginine at a concentration of about 100 mM; and water; wherein the pH of the composition is about 8.0.

[0009] In some embodiments, the liquid pharmaceutical composition is lyophilized, e.g., to remove water, forming a lyophilized powder.

[0010] In another aspect, the present disclosure provides a lyophilized powder comprising: about 13-23% by weight brincidofovir; about 48%-65% by weight mannitol; and about 22-40% by weight arginine.

[0011] In another aspect, the present disclosure provides a lyophilized powder comprising: about 13-19% by weight brincidofovir; about 48%-65% by weight mannitol; and about 22-33% by weight arginine.

[0012] In some embodiments, the lyophilized powder contains about 19% by weight brincidofovir, about 48% by weight mannitol, and about 33% by weight arginine. In some embodiments, the lyophilized powder contains about 13% by weight brincidofovir, about 65% by weight mannitol, and about 22% by weight arginine.

[0013] In some embodiments, the pH of the lyophilized powder is about 8.0.

[0014] In another aspect, the present disclosure provides an aqueous pharmaceutical composition, comprising: brincidofovir; a bulking agent; a buffer; and a sugar alcohol solution, an aqueous sugar solution, Ringer's solution or an aqueous salt (e.g., sodium chloride) solution.

[0015] In some embodiments, the aqueous pharmaceutical composition comprises: brincidofovir; mannitol; L-arginine; and dextrose.

[0016] In some embodiments, the aqueous pharmaceutical composition comprises: brincidofovir; mannitol; L-arginine; and dextrose; wherein the pH of the composition is about 8.0.

[0017] In some embodiments, the aqueous sugar solution is a solution comprising about 5% dextrose by weight. In some embodiments, the aqueous salt solution is an aqueous sodium chloride solution. In some embodiments, the concentration of the sodium chloride solution is 0.9% by weight. In some embodiments, the aqueous pharmaceutical composition further comprises additional water.

[0018] In some embodiments, the additional water is added (e.g. to adjust tonicity, concentration, or pH of the formulation).

[0019] In some embodiments, the aqueous pharmaceutical composition comprises: brincidofovir at a concentration of about 1.0 mg/mL; mannitol at a concentration of about 2.5-5 mg/mL; and L-arginine at a concentration of about 1.74 mg/mL; and dextrose at a concentration of about 50 mg/mL. In some embodiments, the pH of the composition is about 8.0, about 7.5, about 7.0, about 6.5, about 6.0, or below about 6.0.

[0020] In some embodiments, the aqueous pharmaceutical composition comprises: brincidofovir at a concentration of about 1.78 mM; mannitol at a concentration of about 13.75-27.5 mM; L-arginine at a concentration of about 10 mM; and dextrose at a concentration of about 287 mM. In some embodiments, the pH of the composition is about 8.0, about 7.5, about 7.0, about 6.5, about 6.0, or below about 6.0.

[0021] In some embodiments, the aqueous pharmaceutical composition comprises: brincidofovir at a concentration of about 0.5 mg/mL; mannitol at a concentration of about 1.25-2.5 mg/mL; and L-arginine at a concentration of about 0.87 mg/mL; and dextrose at a concentration of about 50 mg/mL. In some embodiments, the pH of the composition is about 8.0, about 7.5, about 7.0, about 6.5, about 6.0, or below about 6.0.

[0022] In some embodiments, the aqueous pharmaceutical composition comprises: brincidofovir at a concentration of about 0.89 mM; mannitol at a concentration of about 6.85-13.7 mM; L-arginine at a concentration of about 5 mM; and dextrose at a concentration of about 287 mM. In some embodiments, the pH of the composition is about 8.0, about 7.5, about 7.0, about 6.5, about 6.0, or below about 6.0.

[0023] In some embodiments, the aqueous pharmaceutical composition comprises: brincidofovir at a concentration of about 1.0 mg/mL; mannitol at a concentration of about 2.5-5 mg/mL; and L-arginine at a concentration of about 1.74 mg/mL; and dextrose at a concentration of about 50 mg/mL; wherein the pH of the composition is about 8.0.

[0024] In some embodiments, the aqueous pharmaceutical composition comprises: brincidofovir at a concentration of about 1.78 mM; mannitol at a concentration of about 13.75-27.5 mM; L-arginine at a concentration of about 10 mM; and dextrose at a concentration of about 287 mM; wherein the pH of the composition is about 8.0.

[0025] In some embodiments, the aqueous pharmaceutical composition comprises: brincidofovir at a concentration of about 0.5 mg/mL; mannitol at a concentration of about 1.25-2.5 mg/mL; and L-arginine at a concentration of about 0.87 mg/mL; and dextrose at a concentration of about 50 mg/mL; wherein the pH of the composition is about 8.0.

[0026] In some embodiments, the aqueous pharmaceutical composition comprises: brincidofovir at a concentration of about 0.89 mM; mannitol at a concentration of about 6.85-13.7 mM; L-arginine at a concentration of about 5 mM; and dextrose at a concentration of about 287 mM; wherein the pH of the composition is about 8.0.

[0027] In some embodiments, the volume of the aqueous sugar solution, aqueous sugar alcohol solution, Ringer's solution, or aqueous salt solution used to dissolve a lyophilized formulation of brincidofovir is about 100 or 200 mL. In other words, in some embodiments, a lyophilized powder comprising brincidofovir is dissolved in about 100 mL or about 200 mL of aqueous sugar solution, aqueous sugar alcohol solution, Ringer's solution, aqueous salt solution, or water. In some embodiments, the lyophilized powder is dissolved in about 100 mL, about 110 mL, about 120 mL, about 130 mL, about 140 mL, about 150 mL, about 160 mL, about 170 mL, about 180 mL, about 190 mL, or about 200 mL. For example, in some embodiments, a lyophilized powder of the disclosure can be dissolved in about 100 mL of a 5% dextrose solution in water. In some embodiments, a lyophilized powder of the disclosure can be dissolved in about 200 mL of a 5% dextrose solution in water.

[0028] In some embodiments, the aqueous pharmaceutical composition comprises: about 100 mg brincidofovir; about 250-500 mg mannitol; about 174 mg arginine; about 5 g dextrose; and about 100 mL water. In some embodiments, the pH of the composition is about 8.0, about 7.5, about 7.0, about 6.5, about 6.0, or below about 6.0.

[0029] In some embodiments, the aqueous pharmaceutical composition comprises: about 200 mg brincidofovir; about 500-1000 mg mannitol; about 348 mg arginine; about 10 g dextrose; and about 200 mL water. In some embodiments, the pH of the composition is about 8.0, about 7.5, about 7.0, about 6.5, about 6.0, or below about 6.0.

[0030] In some embodiments, the aqueous pharmaceutical composition comprises: about 50 mg brincidofovir; about 125-250 mg mannitol; about 87 mg arginine; about 5 g dextrose; and about 100 mL water. In some embodiments, the pH of the composition is about 8.0, about 7.5, about 7.0, about 6.5, about 6.0, or below about 6.0.

[0031] In some embodiments, the aqueous pharmaceutical composition comprises: about 100 mg brincidofovir; about 250-500 mg mannitol; about 174 mg arginine; about 10 g dextrose; and about 200 mL water. In some embodiments, the pH of the composition is about 8.0, about 7.5, about 7.0, about 6.5, about 6.0, or below about 6.0.

[0032] In some embodiments, the aqueous pharmaceutical composition comprises: about 100 mg brincidofovir; about 250-500 mg mannitol; about 174 mg arginine; about 5 g dextrose; and about 100 mL water; wherein the pH of the composition is about 8.0.

[0033] In some embodiments, the aqueous pharmaceutical composition comprises: about 200 mg brincidofovir; about 500-1000 mg mannitol; about 348 mg arginine; about 10 g dextrose; and about 200 mL water; wherein the pH of the composition is about 8.0.

[0034] In some embodiments, the aqueous pharmaceutical composition comprises: about 50 mg brincidofovir; about 125-250 mg mannitol; about 87 mg arginine; about 5 g dextrose; and about 100 mL water; wherein the pH of the composition is about 8.0.

[0035] In some embodiments, the aqueous pharmaceutical composition comprises: about 100 mg brincidofovir; about 250-500 mg mannitol; about 174 mg arginine; about 10 g dextrose; and about 200 mL water; wherein the pH of the composition is about 8.0.

[0036] In some embodiments, one or more of the brincidofovir, the bulking agent, and the buffer have been lyophilized before incorporation into the aqueous pharmaceutical composition described herein.

[0037] In some embodiments, the aqueous pharmaceutical composition is suitable for intravenous administration. In some embodiments, the aqueous pharmaceutical composition is sterile.

[0038] In another aspect, the present disclosure provides a sterile, aqueous pharmaceutical composition for intravenous administration, comprising: brincidofovir; a bulking agent; a buffer; and dextrose.

[0039] In another aspect, the present disclosure provides a sterile, aqueous pharmaceutical composition for intravenous administration, comprising: brincidofovir; a bulking agent; a buffer; and dextrose; wherein the pH of the composition is about 8.0.

[0040] In another aspect, the present disclosure provides an aqueous pharmaceutical composition for intravenous administration, comprising: brincidofovir at a concentration of between about 0.5 mg/mL and about 1.0 mg/mL; a bulking agent at a concentration of between about 2.5 mg/mL and about 5 mg/mL; a buffer at a concentration of between about 0.87 mg/mL and about 1.74 mg/mL; and dextrose at a concentration of about 50 mg/mL. In some embodiments, the pH of the composition is about 8.0, about 7.5, about 7.0, about 6.5, about 6.0, or below about 6.0.

[0041] In another aspect, the present disclosure provides an aqueous pharmaceutical composition for intravenous administration, comprising: brincidofovir at a concentration of between about 0.5 mg/mL and about 1.0 mg/mL; a bulking agent at a concentration of between about 2.5 mg/mL and about 5 mg/mL; a buffer at a concentration of between about 0.87 mg/mL and about 1.74 mg/mL; and dextrose at a concentration of about 50 mg/mL; wherein the pH of the composition is about 8.0.

[0042] In another aspect, the present disclosure provides a method of treating a subject with a viral infection, the method comprising: administering to the subject a pharmaceutical composition or pharmaceutical formulation as set forth herein.

[0043] In another aspect, the present disclosure provides a method of treating a subject with a viral infection, the method comprising: administering to the subject an intravenous pharmaceutical composition comprising brincidofovir; a bulking agent; a buffer; and dextrose

[0044] In another aspect, the present disclosure provides a method of treating a subject with a viral infection, the method comprising: administering to the subject an intravenous pharmaceutical composition comprising brincidofovir; a bulking agent; a buffer; and dextrose; wherein the pH of the composition is about 8.0.

[0045] In another aspect, the present disclosure provides a method of treating a subject with a viral infection, the method comprising: administering to the subject an intravenous pharmaceutical composition comprising brincidofovir at a concentration of between about 0.5 mg/mL and about 1.0 mg/mL; a bulking agent at a concentration of between about 2.5 mg/mL and about 5 mg/mL; a buffer at a concentration of between about 0.87 mg/mL and about 1.74 mg/mL; and dextrose at a concentration of about 50 mg/mL %. In some embodiments, the pH of the composition is about 8.0, about 7.5, about 7.0, about 6.5, about 6.0, or below about 6.0.

[0046] In another aspect, the present disclosure provides a method of treating a subject with a viral infection, the method comprising: administering to the subject an intravenous pharmaceutical composition comprising brincidofovir at a concentration of between about 0.5 mg/mL and about 1.0 mg/mL; a bulking agent at a concentration of between about 2.5 mg/mL and about 5 mg/mL; a buffer at a concentration of between about 0.87 mg/mL and about 1.74 mg/mL; and dextrose at a concentration of about 50 mg/mL %; wherein the pH of the composition is about 8.0.

[0047] In another aspect, the present disclosure provides an aqueous pharmaceutical formulation or composition for treatment of a viral infection, prepared by a process comprising the steps of: dissolving, in any order, an amount of brincidofovir, a bulking agent, and a buffer in water to form a first solution; lyophilizing the first solution to form a lyophilized powder; and dissolving the lyophilized powder in an aqueous sugar alcohol solution, an aqueous sugar solution, Ringer's solution or a sodium chloride solution to form the aqueous pharmaceutical formulation or composition.

[0048] In another aspect, the present disclosure provides the use of an aqueous pharmaceutical composition, comprising: brincidofovir; a bulking agent; a buffer; and an aqueous sugar alcohol solution, an aqueous sugar solution, Ringer's solution or a sodium chloride solution in the manufacture of a medicament for the treatment of a viral infection.

[0049] In another aspect, the present disclosure provides the use of an aqueous pharmaceutical composition, comprising: brincidofovir; a bulking agent; a buffer; and an aqueous sugar alcohol solution, an aqueous sugar solution, Ringer's solution or a sodium chloride solution in the treatment of a viral infection.

[0050] In some embodiments of any of the above-aspects, the viral infection to be treated is, polyomavirus, papillomavirus, herpes virus, adenovirus, Epstein-Barr virus, cytomegalovirus, Hepatitis B virus, Hepatitis C virus, varicella zoster virus, adenovirus, poxvirus, or a combination thereof.

[0051] In some embodiments, administration to the subject of the pharmaceutical compositions set forth herein does not result in hemolysis. In some embodiments, administration to the subject of the pharmaceutical compositions set forth herein does not result in gastrointestinal toxicity.

[0052] In some embodiments, the pH of any of the aqueous formulations or compositions for intravenous administration can have a pH below about 8.0 (e.g., about 8.0, about 7.5, about 7.0, about 6.5, about 6.0, about 5.5, about 5.0, about 4.5, about 4.0, or below about 4.0). In some embodiments, the pH can be higher than 8.0 (e.g., about 8.5, about 9.0, about 9.5, about 10.0, or above about 10.0).

[0053] Without wishing to be bound by theory, administration of oral brincidofovir can result in significantly higher exposure in the intestine compared with other organs; this higher exposure can lead to GI toxicity after oral administration of brincidofovir. As set forth herein, IV administration of brincidofovir can prevent over-exposure of the gut (e.g., intestine) to brincidofovir (e.g., in comparison to oral administration of BCV), with improvement in gastrointestinal (GI) tolerability and reduction in GI toxicity. In some embodiments, IV administration of brincidofovir can deliver comparable (e.g., compared with oral administration) drug exposure to blood plasma and organs such as the liver, kidney, and small intestine, even at lower doses than necessary using oral administration.

[0054] In some embodiments, IV brincidofovir can deliver the drug to organs (e.g., the brain) that can be difficult to reach by other administrative routes (e.g., oral dosage). In some embodiments, higher CNS exposure with IV brincidofovir can treat viral infections in the brain (e.g., herpes encephalitis in newborns and adults; HHV-6 encephalitis; JC virus/PML in transplant recipients or patients with multiple sclerosis).

[0055] Intravenous brincidofovir can avoid GI side effects that can be observed with oral administration of BCV and provide opportunities for treatment of DNA viruses (e.g., in patients who experience GI-side effects associated with oral administration of BCV). For example, like oral administration of brincidofovir, IV administration of brincidofovir can be used as a broad spectrum antiviral agent with limited toxicity (e.g., substantially no heme toxicity, and no kidney toxicity), and can be effective at treating, e.g., adenovirus and smallpox. Additionally, IV administration of brincidofovir can be used to treat and prevent, for example, cytomegalovirus, adenovirus, and BK or JC virus. These diseases can be treated, for instance, in hematopoietic cell transplant patients.

[0056] Additional features and advantages will be apparent to one of skill in the art and are set forth in the Detailed Description of the Invention, below.

BRIEF DESCRIPTION OF THE DRAWINGS

[0057] The skilled artisan will understand that the drawings primarily are for illustrative purposes and are not intended to limit the scope of the inventive subject matter described herein. The drawings are not necessarily to scale; in some instances, various aspects of the inventive subject matter disclosed herein may be shown exaggerated or enlarged in the drawings to facilitate an understanding of different features. In the drawings, like reference characters generally refer to like features (e.g., functionally similar and/or structurally similar elements).

[0058] The patent or application file contains at least one drawing executed in color. Copies of this patent or patent application publication with color drawing(s) will be provided by the Office upon request and payment of the necessary fee.

[0059] The above and further features will be more clearly appreciated from the following detailed description when taken in conjunction with the accompanying drawings.

[0060] FIG. 1 is a graph showing shows product temperature profiles as recorded by product probes as set forth in Example 4.

[0061] FIG. 2 is a pair of graphs illustrating the evaluation of shelf life in an accelerated stability study.

[0062] FIG. 3 is a graph showing the time course of blood and plasma concentrations of radioactivity for male Sprague Dawley (SD) and Long-Evans (LE) rats following a single 2-h intravenous infusion (IV) of [.sup.14C]brincidofovir at a target dose of 15 mg/kg or 2 mg/kg and after a single oral (P0) dose at 15 mg/kg as set forth in Example 9.

[0063] FIG. 4 is a plot of tissue concentration versus time for the small intestine for [.sup.14C]brincidofovir dosed intravenously and orally as set forth in Example 9.

[0064] FIG. 5 is a plot of tissue concentration versus time for the kidney cortex for [.sup.14C]brincidofovir dosed intravenously and orally as set forth in Example 9.

[0065] FIG. 6 is a plot of the plasma concentration of brincidofovir after doses of 100 mg orally, 10 mg IV, and 25 mg IV.

[0066] FIG. 7A is a histogram of rat intestine after oral administration of brincidofovir as set forth in Example 10.

[0067] FIG. 7B is a histogram of rat intestine after IV administration of brincidofovir as set forth in Example 10.

[0068] FIG. 8 is a graph showing the mean (+/-SE) alanine aminotransferase (ALT) levels observed in a IV BCV single ascending dose (SAD) trial.

[0069] FIG. 9 is a graph illustrating mean plasma brindidofovir concentration as a function of time for subjects in cohorts 1-4, and mean plasma BCV concentration as a function of time for subjects administered BCV orally.

[0070] FIG. 10 is a graph illustrating median plasma cidofovir concentration as a function of time following IV and Oral BCV Doses.

DETAILED DESCRIPTION OF THE DISCLOSURE

[0071] The present disclosure is related to pharmaceutical compositions (e.g., lyophilized or aqueous compositions) comprising brincidofovir and methods of use thereof. The disclosure also relates to pharmaceutical formulations comprising a reconstituted lyophilized pharmaceutical composition described herein. The compositions and formulations can be for intravenous administration. In some embodiments, IV administration of the compositions or formulations of the present disclosure do not result in hemolysis. The formulations can also be used in cases where oral administration of drug is not possible or substantially impossible, for example due to underlying conditions or because of inadequate oral absorption.

[0072] The present disclosure relates to at least three types of compositions of brincidofovir. First, the present disclosure relates to bulk formulations of brincidofovir (e.g., liquid formulations of the disclosure or pre-lyophilization formulations of the disclosure) that can be lyophilized prior to storage. The lyophilized formulations (e.g., powders) can be stored for extended periods of time. Additionally, the lyophilized formulations can be reconstituted (e.g. by dissolution in a sugar solution) prior to (e.g., immediately prior to) administration to a patient, e.g., a patient in need of treatment. Accordingly, the present disclosure teaches pre-lyophilization formulations of brincidofovir, lyophilized powders, and re-constituted formulations (e.g., pharmaceutical formulations) for administration.

Formulations of the Disclosure

Pre-Lyophilization Formulations

[0073] In some embodiments, brincidofovir is dissolved in water prior to lyophilization (e.g., freeze-drying). The brincidofovir can be co-dissolved with, for instance, a bulking agent and a buffer. The dissolution of the components may occur separately or concurrently, and may occur in any order.

[0074] In some embodiments, pre-lyophilization compositions of the disclosure comprise a buffer. In some embodiments, pre-lyophilization compositions of the disclosure comprise a bulking agent. In some embodiments, pre-lyophilization formulations of the disclosure comprise a buffer and a bulking agent. In some embodiments, the concentration of brincidofovir is 10 mg/mL. In some embodiments, the pH of the solution is adjusted with HCl and/or NaOH.

[0075] In some embodiments, the buffer is selected from sodium phosphate, arginine, tromethamine and pH-adjusted water. In some embodiments, the buffer is sodium phosphate, L-arginine, or tromethamine. In some embodiments, the buffer is Na-phosphate. In some embodiments, the buffer is arginine. In some embodiments, the buffer is L-arginine. In some embodiments, the buffer is tromethamine. In some embodiments, the buffer is pH-adjusted water. In some embodiments, the buffer is present at a concentration of about 100 mM to about 200 mM. In some embodiments, the buffer is present in an amount of about 25 mM, about 50 mM, about 75 mM, about 100 mM, about 125 mM, about 150 mM, about 175 mM, or about 200 mM.

[0076] In some embodiments, the bulking agent is mannitol or sucrose. In some embodiments, the buffer is sodium phosphate, arginine, or tromethamine. In some embodiments, the bulking agent is present at a concentration of about 5-9% (w/v). In some embodiments, the bulking agent is present at a concentration of about 2.5-9% (w/v). In some embodiments, the bulking agent is present at a concentration of about 2.5% (w/v), about 5% (w/v), or about 9% (w/v).

[0077] In some embodiments, the compositions of the disclosure comprise brincidofovir at a concentration of about 10.0 mg/mL, about 6.4 mg/mL, or about 3.2 mg/mL.

[0078] In some embodiments, the compositions of the disclosure comprise brincidofovir at a concentration of about 10.0 mg/mL, mannitol at a concentration of about 25-50 mg/mL, L-arginine at a concentration of about 17.4 mg/mL and water, and have pH=8.0.

[0079] In some embodiments, the compositions of the disclosure comprise brincidofovir at a concentration of about 17.8 mM, mannitol at a concentration of about 137.5-275 mM, L-arginine at a concentration of about 100 mM and water, and have pH=8.0.

[0080] In some embodiments, any of the pre-lyophilization formulations described herein can be lyophilized to remove water (e.g., for storage).

[0081] In some embodiments, pre-lyophilization formulations can be used directly as pharmaceutical formulations (i.e., can be given directly to a patient). However, in some embodiments, these formulations are lyophilized for storage and are later reconstituted (e.g., in 5% dextrose in water) for administration to a patient.

[0082] In some embodiments liquid formulations of brincidofovir are for intravenous administration after dilution into infusion vehicles (e.g., 5% dextrose).

[0083] In some embodiments, the liquid formulations of the disclosure (i.e., pre-lyophilization formulations of the disclosure) are compatible with infusion vehicles and materials (e.g., containers) used in a clinical setting. For example, the liquid formulations of the disclosure display no significant changes in appearance, pH or recovery of brincidofovir, or introduction of impurities, upon contact with materials such as sterile filters, vials, stoppers, infusion bags, or IV systems. In some embodiments, liquid formulations of the disclosure experience no significant change in pH, loss of brincidofovir, or introduction of impurities upon filtration through a syringe filter.

Lyophilized Formulations

[0084] The pre-lyophilization formulations can be lyophilized to produce lyophilized formulations (e.g., powders) of BCV, bulking agent, and buffer. The lyophilized formulations can be stable for extended periods of time and can be reconstituted prior to administration to a patient. In some embodiments, the lyophilized formulations are sterile.

[0085] In some embodiments, lyophilization comprises freezing, annealing, and drying of the lyophilized composition. In some embodiments, drying comprises a primary drying and a secondary drying. In some embodiments, freezing and annealing comprises exposing the formulation to temperatures between about 5.degree. C. and -50.degree. C. In some embodiments primary drying is conducted at a temperature of about 35.degree. C. In some embodiments secondary drying is conducted at a temperature of about 20.degree. C.

[0086] In some embodiments, freezing and/or annealing lasts for about 16 h. In some embodiments, the primary drying phase lasts for about 20 h. In further embodiments, the secondary drying takes about 22 h (e.g., 21.7 h) or about 28 h (e.g., 27.7 h).

[0087] In some embodiments, liquid formulations of the disclosure (e.g., liquid formulations comprising brincidofovir, arginine and mannitol) do not foam during the lyophilization process, in contrast to other formulations which suffered from foaming during lyophilization.

Reconstituted Pharmaceutical Compositions for IV Administration

[0088] Prior to IV administration to a patient, the lyophilized powders discussed above can be reconstituted, for example by dissolution in an aqueous solvent such as water. In some embodiments, the aqueous solvent is water containing a sugar alcohol or sugar (e.g., dextrose). In some embodiments, the aqueous solvent is 5% dextrose in water. The aqueous solvent can likewise be sterile (e.g., like the sterile lyophilized formulation) and can be suitable for administration to a patient in need thereof. In some embodiments, reconstitution of the lyophilized powders of the disclosures yields aqueous compositions of the disclosure.

[0089] As set forth below, the pharmaceutical compositions for IV administration described herein can provide therapeutically relevant blood plasma concentrations of brincidofovir using lower doses of brincidofovir than those necessary when orally administering brincidofovir. For example, IV administration of brincidofovir using formulations of the present disclosure provided blood plasma concentrations of brincidofovir in humans that had previously demonstrated anti-viral potency in cytomegalovirus prevention and adenovirus treatment. The amount of BCV used in the IV formulations (e.g., about 10 mg or about 25 mg) was about one tenth of the amount of BCV required to achieve similar blood-plasma concentrations using oral dosing.

[0090] Accordingly, in some embodiments, the present disclosure teaches the treatment of a viral infection comprising administering a subject in need thereof an IV formulation of brincidofovir as set forth herein. In some embodiments, the IV dose is less than the orally administered dose of brincidofovir (e.g., about 50% of an oral dose, about 40% of an oral dose, about 30% of an oral dose, about 20% of an oral dose, or about 10% of an oral dose) necessary to achieve a similar result (e.g., blood plasma concentration, anti-viral activity, etc.). Thus, in some embodiments, it is possible to use a smaller quantity of IV brincidofovir than used for oral administration to achieve a similar (e.g., substantially the same) clinical outcome. Additionally, because IV administration results in lower quantities of brincidofovir in the intestine, in some embodiments the present disclosure also provides for the treatment of viral infections without resulting in gastrointestinal toxicity. The present disclosure teaches the treatment of patients who cannot be administered drugs orally (e.g., patients with sensitive intestinal tracts or those unable to swallow oral medications).

[0091] In some embodiments, the compositions of the disclosure comprise brincidofovir at a concentration of about 1.0 mg/mL, mannitol at a concentration of about 2.5-5 mg/mL, L-arginine at a concentration of about 1.74 mg/mL and dextrose at a concentration of about 50 mg/mL, and have pH=8.0.

[0092] In some embodiments, the compositions of the disclosure comprise brincidofovir at a concentration of about 17.8 mM, mannitol at a concentration of about 13.75-27.5 mM, L-arginine at a concentration of about 10 mM and dextrose at a concentration of about 287 mM, and have pH=8.0.

[0093] In some embodiments, the compositions of the disclosure comprise brincidofovir at a concentration of about 0.5 mg/mL, mannitol at a concentration of about 1.25-2.5 mg/mL, L-arginine at a concentration of about 0.87 mg/mL and dextrose at a concentration of about 50 mg/mL, and have pH=8.0.

[0094] In some embodiments, the compositions of the disclosure comprise brincidofovir at a concentration of about 0.89 mM, mannitol at a concentration of about 6.85-13.7 mM, L-arginine at a concentration of about 5 mM and dextrose at a concentration of about 287 mM, and have pH=8.0.

[0095] In some embodiments, the pharmaceutical compositions of the disclosure further comprises a sugar. In some embodiments, the sugar is in an aqueous sugar solution. In some embodiments, the aqueous sugar solution is a 5% dextrose solution. In some embodiments, the volume of the aqueous sugar solution is about 100 or 200 mL.

[0096] In some embodiments, the compositions of the disclosure comprise brincidofovir in an amount of about 100 mg, mannitol in an amount between about 250 mg and about 500 mg, arginine in an amount of about 174 mg, dextrose in an amount of about 5 g, and water in an amount of about 100 mL, and have pH=8.0.

[0097] In some embodiments, the compositions of the disclosure comprise brincidofovir in an amount of about 200 mg, mannitol in an amount between about 500 mg and about 1000 mg, arginine in an amount of about 348 mg, dextrose in an amount of about 10 g, and water in an amount of about 100 mL, and have pH=8.0.

[0098] In some embodiments, the compositions of the disclosure comprise brincidofovir in an amount of about 50 mg, mannitol in an amount between about 125 mg and about 250 mg, arginine in an amount of about 87 mg, dextrose in an amount of about 5 g, and water in an amount of about 100 mL, and have pH=8.0.

[0099] In some embodiments, the compositions of the disclosure comprise brincidofovir in an amount of about 100 mg, mannitol in an amount between about 250 mg and about 500 mg, arginine in an amount of about 174 mg, dextrose in an amount of about 10 g, and water in an amount of about 200 mL, and have pH=8.0.

[0100] In some embodiments, compositions of the disclosure comprise brincidofovir at a concentration of about 0.1-1.0 mg/mL within 100-200 mL of a 5% dextrose solution. In some embodiments, the compositions comprise: 10 mg brincidofovir, 100 mM arginine, and 5% mannitol (w/v), and have pH=8.0.

[0101] In some embodiments, lyophilized compositions of the disclosure are for IV administration by infusion after dilution.

[0102] In some embodiments, the buffer is present at about 200 mM. In some embodiments, the brincidofovir has been previously lyophilized. In some embodiments, the compositions of the disclosure are lyophilized to remove the water. In some embodiments, the brincidofovir, the bulking agent, and the buffer have previously been lyophilized.

[0103] In some embodiments, the present disclosure provides an aqueous pharmaceutical composition for intravenous administration comprising brincidofovir, mannitol, and arginine. In some embodiments, the pH of the formulation is about 8.0. In some embodiments, the pH of the composition is adjusted using HCl and/or NaOH.

[0104] In some embodiments, compositions of the disclosure are suitable for reconstitution with deionized water. In some embodiments, compositions of the disclosure do not foam upon reconstitution. In some embodiments, compositions of the disclosure foam upon reconstitution and foam dissipates in less than 20 minutes, less than 10 minutes, less than 5 minutes, or less than 1 minute. In some embodiments, compositions comprising 100 mM or more of arginine display faster dissipation of foam or bubbles upon reconstitution than compositions comprising 50 mM or less of arginine.

[0105] In some embodiments, lyophilized compositions of the disclosure do not experience changes in appearance, pH or brincidofovir recovery upon dilution with an infusion vehicle (e.g., 5% dextrose), e.g., prior to IV administration.

[0106] In some embodiments, reconstitution of lyophilized compositions of the disclosure does not result in a loss of brincidofovir. In some embodiments, lyophilized formulations of the disclosure are clear and colorless after reconstitution.

Excipients and Pharmaceutically Acceptable Carriers

[0107] In addition to brincidofovir, a buffer and a bulking agent, the pharmaceutical compositions of the disclosure can contain additional pharmaceutically acceptable carriers. Such pharmaceutically acceptable carriers may include any and all solvents, diluents, or other liquid vehicle, dispersion or suspension aids, surface active agents, isotonic agents, thickening or emulsifying agents, preservatives, solid binders, lubricants and the like, as suited to the particular dosage form desired. Remington's Pharmaceutical Sciences, Sixteenth Edition, E. W. Martin (Mack Publishing Co., Easton, Pa., 1980) discloses various carriers used in formulating pharmaceutical compositions and known techniques for the preparation thereof. Except insofar as any conventional carrier medium is incompatible with the active compound (i.e., brincidofovir) such as by producing any undesirable biological effect or otherwise interacting in a deleterious manner with any other component(s) of the pharmaceutical composition, its use is contemplated to be within the scope of this disclosure. Some examples of materials which can serve as pharmaceutically acceptable carriers include, but are not limited to, sugars such as lactose, glucose and sucrose; starches such as corn starch and potato starch; cellulose and its derivatives such as sodium carboxymethyl cellulose, ethyl cellulose and cellulose acetate; powdered tragacanth; malt; gelatine; talc; excipients such as cocoa butter and suppository waxes; oils such as peanut oil, cottonseed oil; safflower oil, sesame oil; olive oil; corn oil and soybean oil; glycols; such as propylene glycol; esters such as ethyl oleate and ethyl laurate; agar; buffering agents such as magnesium hydroxide and aluminum hydroxide; alginic acid; pyrogen free water; isotonic saline; Ringer's solution; ethyl alcohol, and phosphate buffer solutions, as well as other non-toxic compatible lubricants such as sodium lauryl sulfate and magnesium stearate, as well as coloring agents, releasing agents, coating agents, sweetening, flavoring and perfuming agents, preservatives and antioxidants can also be present in the composition, according to the judgment of the formulator.

Methods of Use

[0108] In some embodiments, the present disclosure provides methods of treating a subject with a viral infection, comprising administering to the subject a composition of the disclosure. In some embodiments, a composition of the disclosure is administered intravenously to the subject.

[0109] In some embodiments, formulations of the present disclosure can comprise a volume of concentrated sterile liquid solution (e.g., for dilution) and/or lyophilized powder (e.g., for reconstitution). In some embodiments, the formulations can be stored for at least 12 months (e.g., at about 25.degree. C.). In some embodiments, the formulations can be stored upside down (e.g., for about 24 months, or for about 36 months). In some embodiments, the formulations can be stored in a refrigerator (e.g., for about 12 months). In some embodiments, the lyophilized powder is readily dissolvable (e.g., for reconstitution) in a liquid. In some embodiments, the liquid volume can be small (e.g., for parenteral administration). In some embodiments, the formulations demonstrate acceptable filter and tubing compatibility. In some embodiments, the formulations are compatible with normal resuscitation fluids including but not limited to D5W, NS, and D51/2 NS.

Patient Population

[0110] In some embodiments, administration of a composition of the disclosure does not result in hemolysis. In some embodiments, administration of a composition of the disclosure does not result in gastrointestinal toxicity.

[0111] Accordingly, in some embodiments, the present disclosure provides for the dosing of patients with brincidofovir who may experience unwanted side effects associated with oral administration of brincidofovir (e.g., gastro-intestinal side effects such as diarrhea, pain, constipation and the like). For instance, in some embodiments, IV formulations can be used for patients with symptoms such as anemia, irritable bowel syndrome, constipation, diarrhea, bowel pain, and the like. Additionally, in some embodiments the present disclosure provides for sustained and/or repeated dosing of brincidofovir to subjects in need thereof. For instance, a patient who has undergone a stem cell transplant can be administered IV formulations of the present disclosure to prevent gastro-intestinal side effects associated with oral administration (e.g., repeated oral administration) of brincidofovir.

Viral Indications

[0112] In some embodiments, the viral infection to be treated is selected from polyomavirus (including BK, John Cunningham virus (JCV), Merkel cell virus (MCV), KI polyomavirus (KIV), WU polyomavirus (WUV), Simian virus 40 (SV 40)), papillomavirus (including human papillomavirus, cottontail rabbit papillomavirus, equine papillomavirus and bovine papillomavirus), herpes virus (e.g., herpes simplex virus), adenovirus, Epstein-Barr virus (EBV), human cytomegalovirus (HCMV), Hepatitis B virus, Hepatitis C virus, varicella zoster virus (VZV) or a combination thereof.

[0113] In some embodiments, the IV formulations can be used to prevent clinically significant cytomegalovirus infection in at-risk (e.g., CMV seropositive) adult and pediatric allogenic hematopoietic stem cell transplant recipients. In some embodiments, the present formulations can be used for treatment of adenovirus infection in adult and pediatric immunocompromised hosts. In some embodiments, the formulations can be used for hematopoietic stem cell transplant or solid organ transplant patients.

Dosage Regimens

[0114] In some embodiments, the formulations (e.g., IV formulations) of the present disclosure are administered at a dose of between about 1 mg and 1000 mg BCV. For example, the formulations can be administered at a dose of between about 10 mg and 200 mg BCV. In some embodiments, the IV formulations can be administered at about 5 mg; about 10 mg; about 15 mg; about 20 mg; about 25 mg; about 30 mg; about 35 mg; about 40 mg; about 45 mg; about 50 mg; about 55 mg; about 60 mg; about 65 mg; about 70 mg; about 75 mg; about 80 mg; about 85 mg; about 90 mg; about 95 mg; about 100 about 105 mg; about 110 mg; about 115 mg; about 120 mg; about 125 mg; about 130 mg; about 135 mg; about 140 mg; about 145 mg; about 150 mg; about 155 mg; about 160 mg; about 165 mg; about 170 mg; about 175 mg; about 180 mg; about 185 mg; about 190 mg; about 195 mg; or about 200 mg BCV.

[0115] In some embodiments, IV formulations of the present disclosure can be administered to deliver between about 5 and about 50 mg; about 10 and about 50 mg; about 10 and about 40 mg; about 10 and about 30 mg; about 5 and about 25 mg; about 10 and about 25 mg; and about 15 and about 25 mg of BCV. The formulations can be administered to a human (e.g., an adult human).

[0116] In some embodiments, the formulations are safe and well-tolerated when administered intravenously at doses of between about 10 mg and 100 mg (e.g., at about 10 mg or at about 25 mg). For example, 10 mg and 25 mg doses of brincidofovir formulations of the present disclosure can give favorable tolerability profiles without adverse events. In some embodiments, doses of about 10 mg and 25 mg of the formulations described herein do not produce gastrointestinal side effects.

[0117] In some embodiments, IV formulations taught herein can be administered multiple times per day, or can be administered as single-doses. For instance, IV formulations can be administered once a day or can be administered twice a day. In some embodiments, formulations of the disclosure are administered once a week, or twice a week. In some embodiments, formulations of the disclosure are administered every other day. In some embodiments, formulations of the disclosure are administered every other week. In some embodiments, formulations of the disclosure are administered once a month, or twice a month. One of skill in the art will be able to determine an appropriate dosage regimen for a patient.

[0118] The duration of administration (i.e., for each dose) of IV brincidofovir can vary according to the needs of an individual subject or patient in need thereof and it is within the expertise of one skilled in the art (e.g., a clinician such as a nurse or doctor) to determine the appropriate amount of time that a subject is administered brincidofovir. For instance, IV administration of brincidofovir can last about 15 minutes, about 30 minutes, about 45 minutes, about 60 minutes, about 75 minutes, about 90 minutes, about 105 minutes, or about 120 minutes.

[0119] In some embodiments, the formulations of the disclosure can be administered in combination with other therapeutic agents or treatments. For example, other therapeutic agents can be cisplatin, doxorubicin, etoposide, irinotecan, topotecan, paclitaxel, docetaxel, the epothilones, tamoxifen, 5-fluorouracil, methotrexate, temozolomide, cyclophosphamide, lonafarib, tipifarnib, 4-((5-(((4-(3-chlorophenyl)-3-oxopiperazin-1-yl)methyl)-1H-imidazol-1-yl)- methyl)benzonitrile hydrochloride, (R)-1-((1H-imidazol-5-yl)methyl)-3-benzyl-4-(thiophen-2-ylsulfonyl)-2,3,4- ,5-tetrahydro-1H-benzo di azepine-7-carb onitrile, cetuximab, imatinib, interferon alfa-2b, pegylated interferon alfa-2b, aromatase combinations, gemcitabine, uracil mustard, chlormethine, ifosfamide, melphalan, chlorambucil, pipobroman, triethylenemelamine, triethylenethiophosphoramine, busulfan, carmustine, lomustine, streptozocin, dacarbazine, floxuridine, cytarabine, 6-mercaptopurine, 6-thioguanine, fludarabine phosphate, leucovorin, oxaliplatin, pentostatine, vinblastine, vincristine, vindesine, bleomycin, dactinomycin, daunorubicin, epirubicin, idarubicin, mithramycin, deoxycoformycin, mitomycin-C, L-asparaginase, teniposide 17.alpha.-ethinyl estradiol, diethyl stilbestrol, testosterone, prednisone, fluoxymesterone, dromostanolone propionate, testolactone, megestrol acetate, methylprednisolone, methyltestosterone, prednisolone, triamcinolone, chlorotrianisene, 17.alpha.-hydroxyprogesterone, aminoglutethimide, estramustine, medroxyprogesterone acetate, leuprolide acetate, flutamide, toremifene citrate, goserelin acetate, carboplatin, hydroxyurea, amsacrine, procarbazine, mitotane, mitoxantrone, levamisole, vinorelbine, anastrazole, letrozole, capecitabine, raloxifene, droloxafine, hexamethylmelamine, bevacizumab, trastuzumab, tositumomab, bortezomib, ibritumomab tiuxetan, arsenic trioxide, porfimer sodium, cetuximab, thioTEPA, altretamine, fulvestrant, exemestane, rituximab, alemtuzumab, dexamethasone, bicalutamide, chlorambucil, or valrubicin. The agents can be administered as part of a treatment regime and are not necessarily administered concurrently or on the same schedule.

Definitions

[0120] For the purposes of promoting an understanding of the embodiments described herein, reference made to preferred embodiments and specific language are used to describe the same. The terminology used herein is for the purpose of describing particular embodiments only, and is not intended to limit the scope of the present invention. As used throughout this disclosure, the singular forms "a," "an," and "the" include plural reference unless the context clearly dictates otherwise. Thus, for example, a reference to "a composition" includes a plurality of such compositions, as well as a single composition, and a reference to "a therapeutic agent" or "an active compound" is a reference to one or more therapeutic and/or pharmaceutical agents (e.g., brincidofovir) and equivalents thereof known to those skilled in the art. All percentages and ratios used herein, unless otherwise indicated, are by weight.

[0121] The term "about" is used herein to mean approximately, in the region of, roughly or around. When the term "about" is used in conjunction with a numerical range, it modifies that range by extending the boundaries above and below the numerical values set forth. In general, the term "about" is used herein to modify a numerical value above and below the stated value by a variance of 20%.

[0122] As used in the present disclosure, whether in a transitional phrase or in the body of a claim, the terms "comprise(s)" and "comprising" are to be interpreted as having an open-ended meaning. That is, the terms are to be interpreted synonymously with the phrases "having at least" or "including at least." When used in the context of a process the term "comprising" means that the process includes at least the recited steps, but may include additional steps. When used in the context of a molecule, compound, or composition, the term "comprising" means that the compound or composition includes at least the recited features or components, but may also include additional features or components.

[0123] A "pharmaceutical composition" is a formulation containing a compound of the present disclosure (e.g., brincidofovir) in a form suitable for administration to a subject. The term "pharmaceutical composition" includes preparations suitable for administration to mammals, e.g., humans. A pharmaceutical composition can be, e.g., an intravenous (IV) formulation, or an oral formulation.

[0124] The term "monotherapy" is understood to mean the use of a single drug to treat a particular disorder or disease. Monotherapy is different from combination therapy in that combination therapy includes the use of at least two drugs in combination to treat a particular disorder or disease.

[0125] As used herein, "treating," "treatment" or "treat" describes the management and care of a patient for the purpose of combating a disease, condition, or disorder and includes the administration of active compound of the present disclosure (i.e., brincidofovir), to alleviate the symptoms or complications of a disease, condition or disorder, or to eliminate the disease, condition or disorder. The term "treat" can also include treatment of a cell in vitro or an animal model.

[0126] As used herein, "compounds of the disclosure" refers to both active compounds (e.g., brincidofovir) as well as non-active compounds (e.g., bulking agents, buffers, sweeteners, and the like).

[0127] The active compound of the present disclosure (i.e., brincidofovir) may also be used to prevent a relevant disease, condition or disorder, or used to identify suitable candidates for such purposes. As used herein, "preventing," "prevent," or "protecting against" describes reducing, ameliorating or eliminating the onset of the symptoms or complications of such disease, condition or disorder.

[0128] Brincidofovir can also be used in the prophylaxis of a disease such as a viral infection. Prophylaxis is understood to mean action taken to prevent a disease. In the context of the current disclosure, prophylaxis can mean treatment with, e.g., brincidofovir.

[0129] As used herein, "therapeutically effective amount" means that amount necessary to make a clinically observed improvement in the patient. In some embodiments, the compounds of the disclosure are formulated such that they comprise an amount that would not cause one or more unwanted side effects. For instance, the term "therapeutically effective amount," can refer to an amount of any pharmaceutical agent or agents (e.g., brincidofovir) to treat, ameliorate, or prevent an identified disease or condition (e.g., a viral infection), or to exhibit a detectable therapeutic or inhibitory effect. The effect can be detected by any assay method known in the art. The precise effective amount for a subject will depend upon the subject's body weight, size, and health; the nature and extent of the condition; and the dosing schedule of therapeutics selected for administration.

[0130] Therapeutically effective amounts for a given situation can be determined by routine experimentation that is within the skill and judgment of the clinician. In a preferred aspect, the disease or condition to be treated is viral infection. Furthermore, an effective amount of a pharmaceutical agent is that which provides an objectively identifiable improvement as noted by the clinician or other qualified observer.

[0131] As used herein, the phrase "pharmaceutically acceptable" refers to those compounds, materials, compositions, carriers, and/or dosage forms which are, within the scope of sound medical judgment, suitable for use in contact with the tissues of human beings and animals without excessive toxicity, irritation, allergic response, or other problem or complication, commensurate with a reasonable benefit/risk ratio.

[0132] "Pharmaceutically acceptable excipient or carrier" means an excipient or carrier that is useful in preparing a pharmaceutical composition that is generally safe, non-toxic and neither biologically nor otherwise undesirable, and includes excipient that is acceptable for veterinary use as well as human pharmaceutical use. A "pharmaceutically acceptable excipient" as used in the specification and claims includes both one and more than one such excipient. Pharmaceutically acceptable excipients and carriers are listed above.

[0133] As used herein, a "subject" is interchangeable with a "subject in need thereof", both of which refer to a subject (e.g., a patient) having a disorder in which viral infection plays a part, or a subject having an increased risk of developing viral infection associated disease or disorder relative to the population at large. A "subject" includes a mammal. The mammal can be e.g., a human or appropriate non-human mammal, such as primate, mouse, rat, dog, cat, cow, horse, goat, camel, sheep or a pig. The subject can also be a bird or fowl. In one embodiment, the mammal is a human.

[0134] Representative "pharmaceutically acceptable salts" of brincidofovir include, e.g., water-soluble and water-insoluble salts, such as the acetate, amsonate (4,4-diaminostilbene-2,2-disulfonate), benzenesulfonate, benzonate, bicarbonate, bisulfate, bitartrate, borate, bromide, butyrate, calcium, calcium edetate, camsylate, carbonate, chloride, citrate, clavulariate, dihydrochloride, edetate, edisylate, estolate, esylate, fiunarate, gluceptate, gluconate, glutamate, glycollylarsanilate, hexafluorophosphate, hexylresorcinate, hydrabamine, hydrobromide, hydrochloride, hydroxynaphthoate, iodide, sethionate, lactate, lactobionate, laurate, magnesium, malate, maleate, mandelate, mesylate, methylbromide, methylnitrate, methylsulfate, mucate, napsylate, nitrate, N-methylglucamine ammonium salt, 3-hydroxy-2-naphthoate, oleate, oxalate, palmitate, pamoate (1,1-methene-bis-2-hydroxy-3-naphthoate, einbonate), pantothenate, phosphate/diphosphate, picrate, polygalacturonate, propionate, p-toluenesulfonate, salicylate, stearate, subacetate, succinate, sulfate, sulfosalicylate, suramate, tannate, tartrate, teoclate, tosylate, triethiodide, and valerate salts. In some embodiments, a pharmaceutically acceptable salt is a sodium salt, a potassium salt, or a calcium salt.

Brincidofovir

[0135] Brincidofovir (CMX001, BCV), a lipid conjugate of cidofovir (CDV), can be used for the prevention and treatment of infection and disease caused by double stranded DNA viruses with high unmet medical need. The present disclosure teaches IV formulations of brincidofovir not only as a way to deliver BCV to patients unable to take oral medications, but also as a way to potentially reduce or avoid adverse GI events observed with oral dosing.

[0136] As used herein, the term "brincidofovir" is understood to encompass the neutral compound below:

##STR00002##

[0137] as well as any pharmaceutically acceptable salts thereof. In some of the examples below, brincidofovir is referred to as the "test item."

Cidofovir

[0138] Cidofovir, also known as CDV, has the structure below:

##STR00003##

Brincidofovir Deamination Product

[0139] In some embodiments, brincidofovir can be deaminated under aqueous conditions (e.g., at acidic pH) to produce the corresponding uracil-derivative. The deaminated product is shown below:

##STR00004##

[0140] The present disclosure relates to aqueous and lyophilized formulations comprising brincidofovir and methods of using the same. As used herein, a "lyophilized formulation" means a formulation comprising an amount of solid lyophilized brincidofovir that is dissolved in a solvent (e.g., water, or a sugar solution). A "lyophilized formulation" can be a pharmaceutical composition of the present disclosure.

Characteristics of Pre-Lyophilized Formulations

[0141] As set forth in is set forth in Example 4, pre-lyophilization formulations comprising arginine as buffer and mannitol as bulking agent yield a white uniform solid with only minor melt-back upon lyophilization. Further, the lyophilized product obtained from a pre-lyophilization formulation comprising arginine as buffer and mannitol as bulking agent showed only minor foaming upon reconstitution, in contrast to other formulations tested. For example, formulations comprising sodium phosphate or pH-adjusted water as a buffer showed foaming upon reconstitution and generally required several minutes (e.g., more than 30 minutes) before the foam dissipated. Likewise, formulations comprising sucrose as a bulking agent also were found to exhibit foaming upon reconstitution and generally required several minutes (e.g., more than 30 minutes) before the foam dissipated.

[0142] Furthermore, as shown below in Example 4, mannitol-based aqueous formulations of the disclosure were also found to yield a more compact, uniform lyophilization cake upon lyophilization compared with other formulations. For example, sucrose based formulations exhibited at least partial cake collapses (and in some cases significant cake collapse) as well as an increased melt-back (e.g., extensive melt-back) upon lyophilization. However, mannitol-based aqueous formulations were found to exhibit less cake collapse and less melt-back.

[0143] Additionally, in some embodiments, phosphate-based buffers showed pH shifts on freezing during the lyophilization process, in contrast to formulations using arginine as a buffer. Accordingly, the compositions set forth herein were found to have desirable properties (e.g., greater stability and greater ease of use) compared with other formulations tested.

Stability and Material Compatibility of Liquid Formulations

[0144] As set forth in Example 1, when brincidofovir sample stock solutions (50 .mu.g/mL) in a 50 mM phosphate buffer at pH 6.0-8.0 containing 20% methanol were stored at 2-8.degree. C., precipitation was observed for of pH 7.0 and below. However, samples prepared at a higher pH (e.g., pH=7.5 and pH=8.0) revealed no observable change in appearance throughout the time course of the experiment (e.g., precipitation was not observed). For samples stored at 40.degree. C., no physical change in appearance was observed for samples at any pH and minimal variation in brincidofovir concentration was observed for all of the studied pH conditions at 40.degree. C.

[0145] In the study presented in Example 1, vehicles for brincidofovir formulations were prepared from 5.times. sodium phosphate buffer stocks (e.g., at pH 6.0, 6.5, 7.0, 7.5, or 8.0), various tonicifiers (e.g., 66.5 mM NaCl, 68.8 mM dextrose, 49.8 mM MgCl.sub.2, or 35.7 mM Ca D-gluconate) and various excipients (e.g., Cremophor.RTM., edetate disodium (EDTA), hydroxypropyl beta-cyclodextrin (HPbCD), polyethyleneglycol (PEG) 300, ethanol, glycerin, propylene glycol (PG), N,N-dimethylacetamide (DMA), N-methyl-2-pyrrolidone (NMP), polysorbate 80, albumin, or benzyl alcohol, or mixtures thereof), with a brincidofovir concentration of 6.4 mg/mL. Without wishing to be bound by theory, tested formulations comprising dextrose or NaCl as a tonicifier and Cremophor.RTM. and DMA as excipients, did not precipitate over the course of 12 days at a pH of 6.5, and tested formulations comprising dextrose, but no excipient, did not precipitate over the course of 12 days at a pH of 8.0, but did precipitate at lower pH values. Similarly, and also without wishing to be bound by theory, formulations comprising NaCl, PEG300, and polysorbate 80, and formulations comprising NaCl, Cremophor.RTM. and ethanol, did not precipitate at a pH of 7, but did precipitate at lower pH values of 12 days. Formulations comprising NaCl, and Cremophor.RTM. did not precipitate at a pH of 7. Without wishing to be bound by theory, the HPLC recovery data set forth in Example 1, suggests that under storage condition (2-8.degree. C.) and at c(BCV)=6.4 mg/mL, some formulations are not suited to continuously solubilize brincidofovir in a stable fashion.

[0146] As shown in Example 2, the equilibrium solubility of brincidofovir in aqueous infusion vehicles comprising 100 mM phosphate, and 69 mM dextrose was found to increase at higher pH. For example, the equilibrium solubility of brincidofovir in aqueous infusion vehicles comprising 100 mM phosphate, and 69 mM dextrose was determined as about 25 mg/mL at pH=6.9 and about >270 mg/mL at pH=8.0.

[0147] As set forth in Example 5, solubility of a liquid formulation of the disclosure in a 5% dextrose solution to yield a liquid formulation comprising 10 mg/mL brincidofovir, 100 mM arginine and 5% (w/v) mannitol, was 100% for at least 24 h after dilution of the liquid formulation. Furthermore, the resulting solution stayed clear and colorless, and no significant shifts in pH were observed.

[0148] As set forth in Example 5 and Example 3, liquid formulations of brincidofovir (e.g., formulations comprising 10 mg/mL brincidofovir, 100 mM arginine and 5% (w/v) mannitol; formulations comprising 10 mg/mL or 15 mg/mL brincidofovir and 200 nM sodium phosphate; or formulations comprising 2 mg/mL brincidofovir, and 4 mM sodium phosphate) were tested for compatibility with infusion vehicles as well as a series of materials from the manufacturing and clinical stage (e.g., sterilization filters, vials and stoppers in upright and inverted conditions, infusion bags, and infusion systems). In some instances, the formulations were diluted with 5% dextrose prior to testing. Without wishing to be bound by theory, no precipitation, pH shift or significant loss in recovery of brincidofovir were observed for any of the formulations at the end of the studies.

Hemolytic Potential of Brincidofovir

[0149] In some embodiments, it is advantageous for drugs (e.g., intravenous drugs) to not cause hemolysis (i.e., not to destroy red blood cells).

[0150] Example 7 describes the hemolytic potential of brincidofovir using immediately collected (within 4 hrs) whole human or whole rat blood using a cyanmethemoglobin method to evaluate hemolysis. The study measured hemoglobin released from red blood cells in the presence of brincidofovir to determine the extent to which brincidofovir lysed red blood cells.

[0151] Without wishing to be bound by theory, lysis of red blood cells can cause local reactions such as cellulitis, phlebitis and can impact organ function. The results presented herein demonstrate that the IV formulations of brincidofovir do not cause a hemolytic reaction when administered intravenously.

[0152] Rat and human whole blood (stabilized with K.sub.2EDTA) were obtained from 4 rats and 1 human donor. Each concentration (0.2, 0.5 and 1.6 mg/mL) of brincidofovir for the rat was mixed separately in a 1:4 ratio with rat whole blood while each concentration (0.2, 0.5 and 1.6 mg/mL) of brincidofovir for the human was mixed separately in a 1:4 ratio with human whole blood. All samples were then incubated for 20 minutes at 37.+-.1.degree. C. Following incubation, the blood samples containing the BCV, along with similarly prepared isotonic saline (negative control), 1% Saponin (positive control), 5% Dextrose (vehicle control), and untreated whole blood control samples, were then centrifuged and analyzed for hemolysis (supernatant hemoglobin concentration) using a cyanmethemoglobin method. Representative BCV samples in isotonic saline were analyzed to ensure that the BCV did not interfere with the analysis of supernatant hemoglobin concentration.

[0153] Without wishing to be bound by theory, brincidofovir demonstrated a hemolytic potential effect in rat whole blood that appeared to be concentration dependent with evidence of hemolysis observed at 1.6 mg/mL. However, brincidofovir was found to have no hemolytic potential in human whole blood up to the highest concentration tested corresponding to a final concentration of 0.2, 0.5 and 1.6 mg/mL.

[0154] Accordingly, in some embodiments, brincidofovir is safe for administration to patients, including patients with hematological diseases (e.g., anemia).

Maximum Tolerated Dose and 7-Day Dose Range-Finding (DRF) Study in Rats

[0155] Example 8 presents a maximum-tolerated Dose and 7-day dose range finding study (DRF) for BCV in rats. The results presented detail an acute range finding study in the rat. In some embodiments, this data can be used to generate information to allow one to rationally set the doses to be administered in further toxicology studies.

Phase 1

[0156] Phase 1 was designed to determine the Maximum Tolerated Dose (MTD) of brincidofovir following intravenous infusion (2-hour) to rats. Phase 1 encompassed a single escalating dose arm to assess any acute toxicity (e.g., `toxic syndrome`) that can present in general clinical signs generally within a 48+/- hour period of exposure to BCV. Without wishing to be bound by theory, the escalating dose arm can identify any overt signs or symptoms of over-dosage.

[0157] In Phase 1 of the study, all doses were well tolerated when administered via a single 2-hour IV infusion. Therefore, the maximally tolerated dose of brincidofovir was not determined. However, 15 mg/kg delivered via 2-hour IV infusion of a 0.75 mg/mL solution of brincidofovir represented the maximum feasible dose based on in vitro evidence of hemolysis at brincidofovir concentrations above 0.75 mg/mL. In other words, even at the maximum feasible dose tested, no consequential effects (e.g., on mean body weight or weight gain) were observed in rats. Further, there were no effects in ophthalmology, hematology, clinical chemistry, or urinalysis parameters. Even at higher dosage levels than can be achieved with oral administration, the rats were found not to present with overt toxicity or death.

Phase 2

[0158] Phase 2, the repeat-dose range-finding (DRF) phase (Phase II), was designed to assess the toxicity of brincidofovir and the plasma toxicokinetic profiles of brincidofovir and one of its metabolites, cidofovir. Cidofovir is known to cause nephrotoxicity when administered intravenously to animals and humans. Toxicokinetic parameters were evaluated following the first and last dose administration of two dose levels of brincidofovir (1 mg/kg and 15 mg/kg) administered via intravenous (2-hour) infusion on Days 1, 3, and 7.

[0159] In Phase 2 of the present study the animals received a short term treatment of the high (15 mg/kg) or low (1 mg/kg) BCV IV infusion dose (3 doses over the course of 9 days). Without wishing to be bound by theory, this second phase of the study was used to determine if any potential cumulative toxicity can occur with repeat dosing. Without wishing to be bound by theory, the second phase also included a toxicokinetic arm to determine the establish IV toxicokinetics following single (the first dose administered) and repeat (the last dose administered) BCV IV infusion dosing.

[0160] In Phase 2 of the study, peak concentrations for brincidofovir were generally observed at the end of the 2-hour IV infusion, and decreased rapidly. In general, mean brincidofovir C.sub.max and AUC.sub.last increased approximately proportional to the increase in dose from 1 to 15 mg/kg, though AUC was determined on few concentration values after 1 mg/kg dosing. The brincidofovir C.sub.max and AUC.sub.last values on Day 7 showed a trend toward lower values on Day 7 compared to Day 1 (Day7/Day1 accumulation ratios (AR) of 0.44 to 0.54). Further, no sex differences in brincidofovir TK parameters were observed. Without wishing to be bound by theory, exposure to metabolite cidofovir increased less than proportional to the increase in brincidofovir dose, there were no apparent sex differences in TK parameters, and no change in TK parameters after repeat administration. Under the conditions of the study, single dose administration of 1 or 15 mg/kg brincidofovir via 2-hour IV infusion on Days 1, 4 and 7 were well tolerated.

Brincidofovir: A 28-Day Intravenous Infusion Toxicity Study in Rats with a 14-Day Recovery Period

[0161] Example 10 details a repeat-dose, subchronic study with IV BCV in rats. Without wishing to be bound by theory, this study explores the cumulative biological effects (e.g., clinical, macro and micro) of brincidofovir administered over a range of doses. The effects can be qualitative, such as target organs, nature of effect or quantitative, such as plasma or tissue levels at which effects are observed. Without wishing to be bound by theory, the study can define the toxicity and the potential for recovery or progression.

[0162] As set forth in Example 10, and without wishing to be bound by theory, none of the animals in the present study experienced diarrhea. All of the animals gained their expected weight during the study. There were no GI findings in intestines at terminal necropsy. There were no in-life clinical findings at the highest administered dose of IV brincidofovir (15 mg/kg). No transaminase elevations were observed.

[0163] Without wishing to be bound by theory, it was observed after oral administration of brincidofovir, there was a significant loss of epithelium in intestinal villi. In contrast, after IV administration of brincidofovir, there were minimal single-cell effects noted. For example, FIG. 7A shows a histogram of histogram of rat intestine after oral administration of brincidofovir, and FIG. 7B shows a histogram of rat intestine after IV administration of brincidofovir. As shown in the figures, IV administration of brincidofovir is more well-tolerated in the intestine.

[0164] As set forth in Example 10, brincidofovir was administered twice weekly for 28 days via 2-hour intravenous infusion to Sprague-Dawley CD.RTM. rats (15/sex/group) at 0 (2.times. Sodium Phosphate Buffer Solution), 1, 4 or 15 mg/kg/dose. Up to 5 animals/sex/group were held for a 14-day post-dosing recovery.

[0165] Administration of brincidofovir resulted in intermittent (generally following infusion) clinical signs in some animals (primarily abnormal breathing and hunched posture) in both sexes at .gtoreq.4 mg/kg/dose and minimally decreased food consumption in males and females at .gtoreq.1 mg/kg/dose and 15 mg/kg/dose, respectively.

[0166] Brincidofovir-related findings were present in the male reproductive tract (testicular germ cell depletion, luminal cell debris in the epididymides and increased epithelial apoptosis in the seminal vesicles) and intestinal tract (single cell necrosis) at .gtoreq.4 mg/kg/dose and sebaceous glands (atrophy) at 15 mg/kg/dose. All brincidofovir-related findings completely recovered with the exception of testicular changes in males at .gtoreq.4 mg/kg/dose, which without wishing to be bound by theory would not be expected to fully recover in a 2 week period. In addition, a few animals administered 15 mg/kg/dose had decreased bone marrow cellularity and unilaterally or bilaterally soft and/or small testes and epididymides at the end of recovery.

[0167] Under the conditions of the study, based on the testicular germ cell depletion in males at .gtoreq.4 mg/kg/dose and moderate intestinal single cell necrosis in 1 female at 15 mg/kg/dose, the no-observed-adverse-effect level (NOAEL) for brincidofovir was considered to be 1 mg/kg/dose in males and 4 mg/kg/dose for in females.

[0168] Thus, in some embodiments, the study can provide support for the initiation or continued conduct of clinical studies in humans. In some embodiments, the study can also be used to establish doses for longer term studies. Without wishing to be bound by theory, this study can also guide parameters such as the number of animals to assess when conducting longer-term studies. This study also contained a toxicokinetic arm following the first and last administered dose.

Mass Balance, Pharmacokinetics and Tissue Distribution by Quantitative Whole-Body Autoradiography in Rats Following a Single Oral or Intravenous Dose of [.sup.14C]brincidofovir

[0169] Studies were performed to characterize the tissue distribution of total radioactivity in male Sprague Dawley (SD) and Long-Evans (LE) rats following administration of a single intravenous (IV, 2 h infusion) or oral (PO, gavage) dose of [.sup.14C]brincidofovir. In addition, the rate and extent of excretion (mass balance) and pharmacokinetics (PK) of total radioactivity in male Sprague Dawley rats following a single intravenous (IV, 2 h infusion) dose of [.sup.14C]brincidofovir were examined.

[0170] [.sup.14C]Brincidofovir was administered to pigmented and non-pigmented rats by 2-hour IV infusion or by oral gavage at a dose of 15 mg/kg. Tissue distribution was determined by quantitative whole body autoradiography at time points up to 35-days post-dose.

[0171] Radioactivity was well distributed with qualitative distribution patterns of the radioactivity similar after IV or oral administration. Quantitatively, tissue radioactive concentrations in small intestinal tissue after IV administration were approximately 1/10 the concentrations in small intestinal tissue after oral administration. For most other tissues, tissue radioactive exposure was generally higher after IV administration than after oral gavage administration. Peak concentrations of radioactivity in most tissues occurred at 4 to 8 h after oral administration, or at the end of the 2-h IV infusion. Tissues with highest concentrations of radioactivity were associated with organs of clearance or elimination, e.g., liver, kidney and small intestine, regardless of route of administration. The tissue to plasma ratios (T/P) in these organs were high (>30) and for kidney cortex and liver were similar between the IV and oral routes of administration. Tissues with lowest concentrations of radioactivity were brain, spinal cord, skeletal muscle, white adipose tissue and bone. Association of radioactivity in the brain and spinal cord was higher after IV administration (.about.20% of plasma concentration compared to .about.5% after oral administration). At 35 days post-dose, radioactivity was below the limit of quantification in all tissues except for bone marrow, lymph node, spleen and adrenal gland after IV administration; which were different from those tissues with residual concentrations after oral administration (kidney cortex, liver and small intestine). No evidence of specific association with melanin containing tissues (eye, uvea) was detected.

[0172] Compared to the oral route, IV administration of [.sup.14C]brincidofovir results in lower small intestinal concentrations of BCV-related radioactivity, which is consistent with lower incidence and severity of GI findings in toxicology studies after repeat IV BCV administration to rats.

[0173] In some embodiments, single cell necrosis in the small and large intestine were occasionally noted following IV BCV infusion. These minor and reversible GI findings are explained by the lower concentrations of radioactivity seen in intestinal tissue following IV infusion of [.sup.14C]brincidofovir. Further, the GI findings did not result in any dose-limiting GI toxicities (i.e., gastropathy, enteropathy or enteritis). Accordingly, the present disclosure provides IV formulations of BCV and methods of IV BCV administration that can be used to mitigate BCV-associated GI disturbances.

[0174] Distribution of radioactivity after IV [.sup.14C]brincidofovir infusion was higher in all tissues compared to oral BCV, except in the small intestine, where concentrations were approximately 1/10 of oral administration.

Excretion and Distribution Summary

[0175] Excretion after IV Administration

[0176] The primary route of elimination of radioactivity after a 2-h IV infusion of [.sup.14C]brincidofovir at 15 mg/kg in male SD rats was in urine, which accounted for an average of 51.2% of the administered dose over the 168 h collection period. An average of 42.2% of the administered dose was recovered in feces. The total recovery of radioactivity in urine and feces combined in Group 1 male rats averaged 93.5% of the dose over the 168 h collection period. Most (.about.86%) of the urinary and fecal excretion of radioactivity occurred in the first 24 h after dosing.

Distribution after IV Administration

[0177] The C.sub.max of [.sup.14C]brincidofovir total radioactivity in plasma after a 2-h IV infusion administration to male SD rats at 15 mg/kg (Group 2) was 10.3 .mu.g equiv/mL, which occurred at a T.sub.max of 2 h (i.e., end of infusion), and the concentration decreased to 0.045 .mu.g equiv/mL at 72 h post-dose. The AUC.sub.last of [.sup.14C]brincidofovir total radioactivity was 64.4 .mu.g equivh/mL, and the t.sub.1/2 was 13.0 h.

[0178] [.sup.14C]Brincidofovir-derived radioactivity was well distributed into most tissues of albino and pigmented male rats after a 2-h IV infusion at 15 mg/kg, and most tissues had concentrations that were slightly higher than plasma, in particular, excretory and alimentary canal tissues, which were much higher than plasma (tissue AUC.sub.all:plasma ratio >1.9). The C.sub.max of [.sup.14C]brincidofovir-derived radioactivity in most tissues were found at 2 h post-dose (i.e., end of infusion) when most of the tissues had concentrations that were between 1.0 and 6.0 .mu.g equiv/g.

[0179] Blood to plasma ratios generally showed that [.sup.14C]brincidofovir-derived radioactivity was similarly distributed after IV infusion to rats, ranging from 0.89-1.46 over the period from 1 to 24 hours after the start of infusion. At 72 hours, blood to plasma ratios of 1.42 and 3.06 were observed after administration of 2 mg/kg and 15 mg/kg, respectively.

[0180] The tissues of albino and pigmented male rats with the highest concentrations (>20.0 .mu.g equiv/g) at the respective T.sub.max after a 2-h IV infusion at 15 mg/kg were observed in liver, kidney cortex, small intestine, kidney medulla, and urinary bladder. The tissues with the lowest concentrations (<1.0 .mu.g equiv/g) at their respective T.sub.max were: brain, spinal cord, white adipose, bone, skeletal muscle, and eye lens.

[0181] The highest overall concentrations (>400 .mu.g equiv/g) after a 2-h IV infusion at 15 mg/kg determined by QWBA were observed in the urinary bladder contents, bile, and contents of the alimentary canal, which reflected the routes of elimination for the [.sup.14C]brincidofovir-drug-derived radioactivity after IV administration.

[0182] Tissue concentrations in most tissues decreased steadily, but at 168 h post-dose, elimination of drug related material was not complete in most tissues; however all tissues containing radioactivity had concentrations that were <1.0 .mu.g equiv/g. At 840 h post-dose (last sampled time point), radioactivity was still present in bone marrow, lymph node, spleen, and adrenal gland, but they were approaching the LLOQ in these tissues, except spleen. The t.sub.1/2 for all tissues (with sufficient data points, and acceptable r.sup.2) ranged from 14.2 h for blood (cardiac) to 378 h for spleen. The tissues with the longest t.sub.1/2 were spleen (378 h), lymph node (345 h), skin non-pigmented (310 h), and bone marrow (276 h). The t.sub.1/2 for tissues with quantifiable concentrations at 840 h ranged from 254 h (adrenal gland) to 378 h (spleen).

[0183] A similar pattern of tissue distribution was observed in rats given a 2-h IV infusion at the lower dose of 2 mg/kg. Most tissue concentrations after the 2 mg/kg IV dose compared to 15 mg/kg dose, increased approximately proportional to dose (i.e., with increase in dose of 7.5, a change in concentration of 4 to 15 fold). However, in small intestine, a greater than proportional increase in small intestine concentrations was seen with an increase in dose from 2 to 15 mg/kg.

Distribution after Oral Administration

[0184] The C.sub.max of [.sup.14C]brincidofovir total radioactivity in plasma after PO administration to male LE rats at 15 mg/kg was 1.5 .mu.g equiv/mL at a T.sub.max of 8 h, and the concentration decreased to 0.180 .mu.g equiv/mL at 24 h post-dose. The AUC.sub.last of [.sup.14C]brincidofovir total radioactivity was 21.0 .mu.g equivh/mL, and the t.sub.1/2 was not able to be determined due to insufficient data points.

[0185] Blood to plasma ratios showed that [.sup.14C]brincidofovir-derived radioactivity was similarly distributed from 1 h to 24 h (ranged between 0.76-1.13) after administration of a PO dose of [.sup.14C]brincidofovir to male LE rats.

[0186] [.sup.14C]brincidofovir-derived radioactivity was well distributed into most tissues of pigmented and non-pigmented male rats after a PO administration at 15 mg/kg, and most tissues had concentrations that were slightly higher plasma, especially for excretory and alimentary canal tissues, which were much higher (tissue AUC.sub.all:plasma ratio >4.1). The C.sub.max of [.sup.14C]brincidofovir-derived radioactivity in most tissues were found at 8 h post-dose (22 of 38 tissues) when most of the tissues had concentrations that were between 0.7 and 17.8 .mu.g equiv/g.

[0187] The tissues of pigmented male rats with the highest concentrations (>3.3 .mu.g equiv/g) at the respective T.sub.max after a PO dose at 15 mg/kg were observed in: small intestine, liver kidney cortex, stomach (gastric mucosa), kidney medulla, cecum, esophagus, large intestine. The tissues with the lowest concentrations (<1.0 .mu.g equiv/g) at their respective T.sub.max were: spinal cord, brain, bone, white adipose, and eye lens.

[0188] The highest overall concentrations (>36.0 .mu.g equiv/g) after a PO dose at 15 mg/kg determined by QWBA were observed in the urinary bladder contents, bile, and contents of the alimentary canal, which reflected the routes of elimination for the [.sup.14C]brincidofovir-drug-derived radioactivity after PO administration.

[0189] Tissue concentrations in most tissues of pigmented male rats decreased steadily, but at 168 h post-dose, elimination of drug related material was not complete in most tissues; however all tissues had concentrations that were <0.4 .mu.g equiv/g, with the exception of small intestine (5.9 .mu.g equiv/g). At 840 h post-dose (last sampled time point) radioactivity was still present in kidney cortex, liver, and small intestine, but was approaching the LLOQ. The t.sub.1/2 for plasma was approximately 5 h. The t.sub.1/2 for all tissues (with sufficient data points, and acceptable r.sup.2) ranged from 37.2 h for esophagus to 234 h for adipose (brown). The tissues with the longest t.sub.1/2 were adipose brown (234 h), spleen (115 h), pituitary gland (106 h), and lung (93 h). The only tissue with a reliable t.sub.1/2 and quantifiable concentrations at 840 h was small intestine at 98.1 h.

Comparison of Distribution to Tissues after a PO Dose and a 2-h IV Infusion

[0190] Following a 2-hour intravenous infusion or oral gavage of [.sup.14C]brincidofovir to Long-Evans and Sprague-Dawley rats, the AUC.sub.all blood to plasma ratio ranged between 1.1 and 1.4. For individual points, the blood to plasma ratio ranged from 0.64 to 1.5 with a median value of 1.00. Without wishing to be bound by theory, the similar distribution of radioactivity between blood and plasma after both routes of administration indicated minimal restriction of [.sup.14C] brincidofovir distribution from plasma into blood cells.

[0191] Most tissues of rats given a single oral dose of [.sup.14C]brincidofovir at 15 mg/kg attained maximal observed concentrations at either 4 or 8 h, and peak concentrations were generally about 10-25% of peak tissue concentrations observed in rats given a 2-h IV infusion. In small intestine, however, a 6-fold higher C.sub.max was observed after oral administration compared to IV. The oral bioavailability of radioactivity (AUC.sub.all PO/AUC.sub.all IV) was 48% (blood) to 61% (plasma). Lower tissue AUC ratios (AUC.sub.PO/AUC.sub.IV<25%) were observed in approximately 1/3 of the tissues after oral administration compared to IV, notably in central nervous system, skin, skeletal muscle and some endocrine and reproductive tissues. By both routes, tissues with highest concentrations of radioactivity were associated with excretory organs (i.e., liver, kidney, and intestine). The tissue to plasma ratios were high (>30) for each of these tissues, and in kidney cortex and liver the tissue/plasma ratios were similar between the IV and oral routes of administration. Small intestinal tissue to plasma ratio was highest after oral administration (T/P=428) as expected based on the route of administration, however, notable radioactivity was associated with small intestine (tissue to plasma ratio 32) after IV infusion. Elimination from tissues was incomplete at 840 h post oral-dose with radioactivity present in kidney cortex, liver, and small intestines after PO administration, which were different from those observed after a 2-h IV infusion at the same time point. In small intestine, the residual concentration at 168 h was considerably higher for orally dosed rats (5.9 equiv/g) than that for rats administered [.sup.14C]brincidofovir by IV infusion (tissue concentrations BQL). At 840 h, residual concentrations were much higher in vascular/lymphatic tissue systems after a 2-h IV infusion compared to the concentrations observed after a PO administration (BQL).

Distribution in Pigmented Tissues

[0192] A comparison of tissue distribution of radioactivity between non-pigmented and pigmented tissues showed that concentrations in the pigmented tissues (i.e., eye uveal tract and pigmented skin) were similar to those concentrations observed in the non-pigmented tissues, which suggested that there was no specific association of [.sup.14C]brincidofovir-drug-derived radioactivity with melanin.

[0193] Without wishing to be bound by theory, in most tissues, higher concentrations of radioactivity were observed after IV administration, which can be reflective of higher blood concentrations. One exception was the gastro-intestinal tissue. Tissues with the highest concentrations, ranked from highest to lowest, were liver, kidney cortex, small intestine, and kidney medulla for IV administration, and small intestine, liver, kidney cortex, kidney medulla, and cecum for oral administration. The tissues with residual drug after 840 hours were the adrenal glands, spleen, lymph nodes and bone marrow for IV administration, and small intestine, liver, and kidney cortex for oral administration.

[0194] Thus, in some embodiments, lower dose of BCV administered intravenously can provide similar plasma concentrations similar to those observed using oral administration at higher doses. Accordingly, the present disclosure provides treatment of a viral infection in a subject in need thereof using a lower dose of BCV than necessary with oral administration.

Pharmacokinetics of Brincidofovir IV Administration

[0195] As set forth in Example 11 below, brincidofovir was administered to healthy subjects both orally and intravenously. It was found that IV brincidofovir administration at 10 mg provided similar exposure as orally administered brincidofovir at 100 mg. Accordingly, the present disclosure teaches administration of brincidofovir intravenously. In some embodiments, no drug-related adverse events (e.g., no gastrointestinal events) are observed upon IV administration of brincidofovir. In some embodiments, no graded lab abnormalities (e.g., no hemolytic toxicity and no kidney toxicity) are observed.

EXAMPLES

[0196] The disclosure is further illustrated by the following examples which are not to be construed as limiting this disclosure in scope or spirit to the specific procedures herein described. It is to be understood that the examples are provided to illustrate certain embodiments and that no limitation to the scope of the disclosure is intended thereby. It is to be further understood that resort may be had to various other embodiments, modifications, and equivalents thereof which may suggest themselves to those skilled in the art without departing from the spirit of the present disclosure and/or scope of the appended claims.

[0197] This section provides abbreviations and definitions of terms and concepts that may be commonly used throughout. [0198] AUC Area Under the Time-Concentration Curve [0199] AUC.sub.all Area Under the Time-Concentration Curve Including all Time Points [0200] AUC.sub.inf_obs Area Under the Time-Concentration Curve Extrapolated to Infinity Observed [0201] AUC.sub.last Area Under the Time-Concentration Curve Including the last time point with data above the lower limit of quantitation [0202] BQL Below Quantification Limit [0203] .degree. C. Degrees Centigrade [0204] c(BCV) Concentration of brincidofovir in a liquid or aqueous formulation [0205] c(bulking agent) Concentration of bulking agent in liquid or aqueous formulation [0206] ca Circa (approximately) [0207] C.sub.max Maximum observed concentration [0208] cpm Counts Per Minute [0209] % CV Percent Coefficient of Variance [0210] DMA N,N-dimethylacetamide [0211] dpm Disintegrations Per Minute [0212] EDTA edetate disodium [0213] FVC Femoral Vein Cannulated [0214] g Gram [0215] h Hour [0216] HPLC High Performance Liquid Chromatography [0217] HPbCD hydroxypropyl beta-cyclodextrin [0218] IV Intravenous [0219] kg Kilogram [0220] L Liter [0221] LE Long-Evans [0222] LLOQ Lower Limit of Quantification [0223] LSC Liquid Scintillation Counting [0224] MB Mass Balance [0225] MBq Megabecquerel [0226] mCi/mmol Millicurie Per Millimole [0227] MCD MicroComputer Image Device [0228] MDC Molecular Dynamics Counts [0229] mg Milligram [0230] mSv milli Sievert(s) [0231] .mu.g equiv/g Microgram Equivalents per Gram of Tissue [0232] min Minutes [0233] mL Milliliter [0234] mm Millimeter [0235] n Number of Observations [0236] NA Not Applicable [0237] NI Not Identified [0238] NC Not Calculated [0239] NS Not Sampled [0240] NMP N-methyl-2-pyrrolidone [0241] PEG polyethyleneglycol [0242] PG propylene glycol [0243] PK Pharmacokinetic [0244] PO per os (oral dose) [0245] ROA Route of Administration [0246] SD Sprague Dawley or Standard Deviation [0247] t.sub.1/2 Terminal Elimination Half-Life [0248] T.sub.max Time of C.sub.max [0249] ULOQ Upper Limit of Quantification [0250] QWBA Quantitative Whole-Body Autoradiography

Example 1--Liquid Formulation Development

[0251] Various plain liquid formulations (i.e., pre-lyophilization formulations, or bulk formulations) were prepared and tested for the purpose of developing intravenous formulations of brincidofovir.

[0252] pH Stability of Brincidofovir:

[0253] Brincidofovir sample stock solutions (50 .mu.g/mL) were prepared in 50 mM phosphate buffer at pH 6.0-8.0 containing 20% methanol. The samples were aliquoted and stored at two temperatures (T=2-8.degree. C. and T=40.degree. C.), and monitored for pH and recovery of brincidofovir at multiple time points over the course of two weeks. Under both conditions, the samples maintained the pH level throughout the time course of the experiment.

[0254] Storage at 2-8.degree. C.:

[0255] For samples stored at 2-8.degree. C., precipitation was observed for pH 6.0 and 6.5 at t=2 days. For pH 7.0, precipitation was observed at t=4 days. Samples prepared at a higher target pH (pH=7.5 and pH=8.0) revealed no observable change in appearance throughout the time course of the experiment, whereas the samples prepared at pH 6.0, 6.5, and 7.0 showed a significant decrease in brincidofovir concentration with respect to the initial time point. The decrease may be related to the observed precipitation. Variation for pH 7.0 is observed for t>8 days. One impurity (RT=4.7 min) was detected that exceeded the set total peak area threshold that was applied for peak integration. The percent of the total peak area was below 0.4% for samples at pH 6 and 6.5, and below 0.1% for samples at higher pH.

[0256] Storage at 40.degree. C.:

[0257] For samples stored at 40.degree. C., no physical change in appearance was observed for samples at any pH. Minimal variation in brincidofovir concentration was observed for all of the studied pH conditions at 40.degree. C. While concentration of brincidofovir showed some fluctuation, no significant trends can be observed. A synoptic display of the HPLC chromatograms revealed differences in impurity formation across the varying pH-conditions over the course of the experiment. For the samples at lower pH, a peak associated with the deamination product of brincidofovir, was observed, encompassing an area % of 1.91 at pH 6.0, and 0.58 at pH 6.5. The degradants peak is not observed at neutral pH and above.

##STR00005##

Preparation and Characterization of Brincidofovir Formulations

[0258] Preparation of Vehicles and Formulations:

[0259] Vehicles for brincidofovir formulations were prepared from organic excipients, 5.times. sodium phosphate buffer stocks (pH 6.0, 6.5, 7.0, 7.5, and 8.0), and the stock solutions of tonicifiers and EDTA. Solid brincidofovir was dissolved in the vehicle yielding a concentration of c(BCV)=6.4 mg/mL. The appearance (including precipitation upon storage at 2-8.degree. C.) was evaluated, and the pH was measured. The results of the study are summarized in Table 1.

TABLE-US-00001 TABLE 1 Precipitation status and pH of vehicles and formulations. All formulations except formulations # 30-32 were prepared with a Na phosphate buffer (100 nM). Precipitation status.sup.1 Formulation composition Target Vehicle Form. Day Day Day Day # Tonicifier Excipient #1 Excipient #2 pH pH pH init. 1 2 7 12 1 NaCl, 66.5 mM Cremophor .RTM., 40% DMA, 10% 6.00 6.05 6.03 N N N N Y 2 NaCl, 66.5 mM Cremophor .RTM., 40% DMA, 10% 6.50 6.18 6.68 N N N N N 3 NaCl, 66.5 mM -- -- 7.00 6.87 6.83 Y Y Y Y Y 4 NaCl, 66.5 mM -- -- 7.50 7.33 7.23 N N N Y Y 5 NaCl, 66.5 mM -- -- 8.00 7.80 7.39 N N N Y Y 6 Dextrose, 68.8 nM Cremophor .RTM., 40% DMA, 10% 6.00 6.38 6.07 N N N N Y 7 Dextrose, 68.8 mM Cremophor .RTM., 40% DMA, 10% 6.50 7.22 6.78 N N N N N 8 Dextrose, 68.8 mM -- -- 7.00 6.88 6.83 N N N Y Y 9 Dextrose, 68.8 mM -- -- 7.50 7.35 7.21 N N N Y Y 10 Dextrose, 68.8 mM -- -- 8.00 7.89 7.45 N N N N N 11 MgCl2, 49.8 mM -- -- 7.00 6.22 6.42 Y Y Y Y Y 12 Ca D-gluconate, 35.7 mM -- -- 7.00 6.49 6.63 Y Y Y Y Y 13 NaCl, 66.5 mM EDTA, 50 .mu.M -- 7.00 6.82 6.95 Y Y Y Y Y 14 NaCl, 66.5 mM HPbCD, 1.8% -- 7.00 6.86 6.93 N N N Y Y 15 NaCl, 66.5 mM HPbCD, 6.4% -- 7.00 6.92 6.85 N N N Y Y 16 NaCl, 66.5 mM PEG 300, 10% -- 7.00 7.29 6.99 Y Y Y Y Y 17 NaCl, 66.5 mM polysorbate 80, 2% -- 7.00 6.82 6.87 N N N N Y 18 NaCl, 66.5 mM PEG 300, 10% polysorbate 80, 2% 6.00 6.28 6.20 Y Y Y Y Y 19 NaCl, 66.5 mM PEG 300, 10% polysorbate 80, 2% 7.00 7.31 6.98 N N N N N 20 NaCl, 66.5 mM Ethanol, 10% -- 7.00 7.35 7.02 N N N Y Y 21 NaCl, 66.5 mM Cremophor .RTM., 10% -- 7.00 6.93 6.87 N N N N N 22 NaCl, 66.5 mM Cremophor .RTM., 10% Ethanol, 10% 6.00 6.19 6.03 N N N Y Y 23 NaCl, 66.5 mM Cremophor .RTM., 4% Ethanol, 10% 7.00 7.30 7.00 N N N N N 24 NaCl, 66.5 mM Cremophor .RTM., 10% Ethanol, 10% 7.00 7.51 7.13 N N N N N 25 NaCl, 66.5 mM Glycerin, 10% -- 7.00 7.02 7.06 N N N Y Y 26 NaCl, 66.5 mM PG, 10% -- 7.00 7.36 7.08 Y Y Y Y Y 27 NaCl, 66.5 mM DMA, 10% -- 7.00 7.63 7.20 Y Y Y Y Y 28 NaCl, 66.5 mM NMP, 1% -- 7.00 7.13 7.07 N N N Y Y 29 NaCl, 66.5 mM Albumin, 5% -- 7.00 7.24 6.95 N N N Y Y 30 -- Cremophor .RTM., 50% Ethanol, 50% N/A N/A N/A Y Y Y Y Y 31 -- Cremophor .RTM., 50% Ethanol, 44%; DMA N/A N/A N/A Y Y Y Y Y 32 -- Cremophor .RTM., 50% Ethanol, 30%; N/A N/A N/A Y Y Y Y Y polysorbate 80, 8%; Benzyl alcohol, 2% .sup.1Formulations were prepared at ambient temperature and kept at ambient conditions until day 1 time point. On day 1, the hemolysis assay was carried out after which the samples were stored at 2-8.degree. C. .sup.2N: No precipitation observed. .sup.3Y: Precipitation observed. .sup.4N/A: Not applicable or not measured.

[0260] The vehicles were clear, colorless solutions. Precipitation was observed for vehicles #11 and #12, and vehicle #29 was a clear light yellow solution. Formulations #11 and #12 inherited precipitation from their vehicles. Formulations #18, #30, #31, and #32 were observed with precipitation on day 1 after preparation. Formulations were stored at 2-8.degree. C. from day 2 onwards, and the appearance was monitored over 12 days.

[0261] Brincidofovir Recovery:

[0262] The brincidofovir formulations of Table 1 were analyzed with respect to c(BCV) on day 2 and day 7 after preparation. Formulations that showed initial precipitation were not tested on day 2. Formulations that showed precipitation up to day 7 were subjected to centrifugation and HPLC recovery analysis. The results are summarized in Table 2.

TABLE-US-00002 TABLE 2 HPLC recovery of brincidofovir formulations Formulation Supernatant analysis (day 2) analysis (day 7) Target Observed Observed c(BCV) c(BCV) Recovery c(BCV) Recovery Form. # (mg/mL) (mg/mL) (%) (mg/mL) (%) 1 6.4 8.01 125 -- -- 2 6.4 8.06 126 -- -- 3 6.4 -- -- 1.15 18 4 6.4 7.20 112. 1.17 18 5 6.4 7.41 116 7.18 112 6 6.4 11.25 176 -- -- 7 6.4 8.70 136 -- -- 8 6.4 8.15 127 0.94 15 9 6.4 8.43 132 6.89 108 10 6.4 7.96 124 -- -- 11 6.4 -- -- 0.52 8 12 6.4 -- -- 8.30 130 13 6.4 -- -- 0.88 14 14 6.4 9.78 153 1.41 22 15 6.4 10.12 158 4.58 72 16 6.4 -- -- 0.52 8 17 6.4 8.53 133 -- -- 18 6.4 -- -- 2.22 35 19 6.4 9.11 142 -- -- 20 6.4 7.66 120 1.85 29 21 6.4 7.79 122 -- -- 22 6.4 8.11 127 3.74 58 23 6.4 8.33 130 -- -- 24 6.4 8.15 127 -- -- 25 6.4 8.67 136 0.57 9 26 6.4 -- -- 1.14 18 27 6.4 -- -- 0.60 9 28 6.4 7.93 124 0.73 11 29 6.4 9.56 149 3.44 54 30 6.4 -- -- 0.70 11 31 6.4 -- -- 0.72 11 32 6.4 -- -- 1.12 18

[0263] Vehicle #11 and #12 were found to form precipitate upon preparation of the formulation (i.e., addition of brincidofovir). Formulations with a low pH formed precipitate in the presence of solubilizing excipients such as Cremophor.RTM., PEG 300, polysorbate 80, and ethanol (formulation #1, #6, #18, and #22), while formulations with the same composition, but higher pH (formulation #2, #7, #19, and #24) maintained the ability to solubilize brincidofovir (Table 1). Purely organic formulations (formulation #30, #31, and #32) showed limited solubilizing ability toward brincidofovir.

[0264] The HPLC recovery data demonstrate that under storage condition (2-8.degree. C.) and at c(BCV)=6.4 mg/mL, some formulations are not suited to continuously solubilize brincidofovir. Moreover, the data demonstrate that formulations with pH greater than or equal to about 8.0 can solubilize brincidofovir.

[0265] Further formulations (formulations #33-40 and #49-56) were prepared and tested for brincidofovir equilibrium solubility and filterability.

[0266] Formulation vehicles #33-40 and #49-56 were saturated with brincidofovir and rotated at ambient conditions. The recovery of brincidofovir in the sample supernatant was determined at different time points to establish when the solubility equilibrium was reached.

[0267] The saturated supernatants of the equilibrium solubility study were subjected to filterability assessment. For this, approximately 900 .mu.L of the saturated solutions were filtered through a syringe filter (25 mm, 0.2 .mu.m PES membrane). The ease of the filtration was recorded, the pH in the filtrate was measured, and the brincidofovir recovery was determined by HPLC analysis. In order to assess if sterile filtering introduces impurities, the HPLC chromatograms of the saturated formulation vehicles were qualitatively compared at the pre- and post-filtration stage and are shown. The composition of each of formulations #33-40 and #49-56, as well as their pH, c(BCV) at solubility equilibrium and ease of filtration are summarized in Table 3.

TABLE-US-00003 TABLE 3 Summary of brincidofovir solubility and filterability assessment. The buffer in all formulations was 100 mM Na phosphate. Observed c(BCV) Measured pH (mg/mL) BCV- Volume At Form. Composition Target saturated filtered Ease of solubility Post- # Tonicifier Excipient pH Vehicle filtrate (.mu.L) filtration equilibrium filtration 33 Dextrose, 68.8 mM -- 8.0 8.1 6.9 900 Easy 27 27 34 NaCl, 66.5 mM Cremophor .RTM., 10% 7.5 7.6 6.5 900 Easy 30 31 35 NaCl, 66.5 mM Cremophor .RTM., 5% 7.5 7.6 6.6 900 Easy 27 29 36 Dextrose, 68.8 mM Cremophor .RTM., 5% 7.5 7.6 6.7 900 Easy 26 26 37 NaCl, 66.5 mM Cremophor .RTM., 2% 7.5 7.6 6.7 900 Easy 25 26 38 NaCl, 66.5 mM HPbCD, 4% 7.5 7.6 6.8 600 Easy 19 16 39 NaCl, 66.5 mM SBEbCD, 7.5% 7.5 7.6 6.3 200 Easy 4 4 40 NaCl, 66.5 mM -- 7.5 7.6 6.8 900 Easy 22 22 49 Dextrose, 68.8 mM -- 7.5 7.6 6.9 1000 Easy 25 24 50 Dextrose, 68.8 mM -- 7.5 7.6 6.9 1000 Easy 24 25 51 Dextrose, 68.8 mM -- 7.5 7.6 6.9 1000 Easy 27 25 52 NaCl, 66.5 mM -- 7.5 7.6 6.8 1000 Easy 29 27 53 NaCl, 66.5 mM -- 7.5 7.6 6.8 1000 Easy 27 27 54 NaCl, 66.5 mM -- 7.5 7.6 6.8 1000 Easy 24 29 55 Dextrose, 68.8 mM -- 8.0 8.0 6.9 1000 Easy 28 31 56 Dextrose, 68.8 mM -- 8.0 8.0 6.9 1000 Easy 29 30

[0268] The equilibrium solubility of brincidofovir in the tested formulations ranged from 4 to 30 mg/mL with an average solubility of c(BCV)=25 mg/mL within the range of apparent pH=6.3-6.9 (ambient temperature). In some embodiments, the relatively high brincidofovir concentrations observed in equilibrium solubility testing led to a significant shift of the vehicle pH. Filtration of the formulations saturated with brincidofovir through the selected syringe filters did not reveal any issues regarding the ease of filtration, loss of brincidofovir, or introduction of impurities.

Example 2--Brincidofovir Formulation Testing

[0269] A formulation vehicle comprising 100 mM phosphate and 69 mM dextrose (vehicle/Formulation #57), was prepared and tested further with respect to the a) equilibrium solubility, and b) filterability.

a) Brincidofovir Solubility Assessment

[0270] Two samples of vehicle #57, were saturated with brincidofovir. One sample vehicle was saturated with brincidofovir without further pH adjustment (leading to a formulation pH of 6.9) and the pH of the second sample was re-adjusted to pH=8.0 using a sodium hydroxide solution (1 N). The pH drifted towards lower pH values upon dissolution of brincidofovir which in return changed the brincidofovir solubility until an equilibrium was reached. Consequently, the pH had to be brought up close to the target pH before more brincidofovir was added. The concentration of brincidofovir at equilibrium solubility conditions was not reached for the pH adjusted sample. After supplementing the sample with approximately 300 mg/mL of additional brincidofovir solid, the addition of solid brincidofovir was stopped.

[0271] The recovery of brincidofovir (HPLC assay) in both sample solutions/supernatants was determined at equilibrium and after 24 h. At ambient temperature, the equilibrium solubility of brincidofovir in formulation 57 was c(BCV)=25 mg/mL at an apparent pH of pH=6.9 and 26 mg/mL after 24 h. For the sample at pH=8 the observed brincidofovir concentration was 348 mg/mL after 24 h, and the equilibrium solubility was not reached with c(BCV)>270 mg/mL.

b) Filterability Assessment

[0272] The filterability of formulation #57 was tested at c(BCV)=20, 10, and 0.5 mg/mL. The testing solution at c(BCV)=20 mg/mL was adjusted to pH=8.0 using a NaOH-solution (1 N) and was applied as a stock solution to prepare the testing solutions at lower concentration by dilution with D5W. The testing solutions were filtered (5 mL filter pass) through a syringe filter (25 mm, 0.2 .mu.m PES membrane). The ease of the filtration was observed and the pH as well as brincidofovir recovery (HPLC assay) were recorded at the pre- and post-filtration stage. No significant change in pH, loss of brincidofovir, or introduction of impurities was observed (Table 4).

TABLE-US-00004 TABLE 4 Filterability assessment of brincidofovir in the formulation 57 vehicle Observed Measured pH c(BCV) (mg/mL) c(BCV) Pre Post Pre Post prepared to filtration filtration filtration filtration 20 mg/mL 8.0 8.0 23 26 10 mg/mL 8.1 8.1 13 12 0.5 mg/mL 8.0 8.1 0.8 0.9

Example 3--Formulation Studies and Assessments

[0273] A compounding process for preparation of brincidofovir formulations was developed and has been specified for the tonicifier (dextrose)-free formulation. For stability studies and testing, the following brincidofovir formulations were prepared on a scale of up to 0.5 L: [0274] 10 mg/mL brincidofovir, 200 mM sodium phosphate, pH=8.0, 69 mM dextrose [0275] 15 mg/mL brincidofovir, 200 mM sodium phosphate, pH=8.0, 69 mM dextrose [0276] 10 mg/mL brincidofovir, 200 mM sodium phosphate, pH=8.0 (tonicifier-free) Tonicifier-Containing Formulation (w/Dextrose)

[0277] A brincidofovir formulation containing dextrose as tonicifier was applied in stability study I as well as in the infusion vehicle compatibility assessment. The outline of the corresponding formulation preparation is as follows: [0278] 1. 500 mM (2.5.times.) monobasic sodium phosphate solution and 500 mM (2.5.times.) dibasic sodium phosphate solution were prepared by dissolving the corresponding sodium phosphate species in DI water using a volumetric flask. [0279] 2. The 500 mM (2.5.times.) monobasic and dibasic sodium phosphate solutions were mixed under pH-control to prepare a 500 mM (2.5.times.) sodium phosphate buffer solution at pH=8.0. [0280] 3. A vehicle stock solution of 400 mM (2.times.) sodium phosphate, 138 mM (2.times.) dextrose at pH=8.0 was prepared by dissolving solid dextrose in the 500 mM (2.5.times.) sodium phosphate solution at pH=8.0 and Q.S. with water. [0281] 4. Brincidofovir was dissolved in the vehicle stock solution (400 mM (2.times.) sodium phosphate, 138 mM (2.times.) dextrose solution, pH=8.0) at 2.times. of the desired final concentration using a volumetric flask. Under pH control a sodium hydroxide solution was added to adjust the solution to pH=8.0. DI water was used to Q.S. yielding a formulation of the desired brincidofovir concentration containing 200 mM (1.times.) sodium phosphate, 69 mM (1.times.) dextrose, pH=8.0.

Tonicifier Free Formulation (w/o Dextrose)

[0282] A brincidofovir formulation lacking dextrose was applied in stability study II, as well as in the adsorption assessment, and the majority of experiments associated with material compatibility assessments. The tonicifier-free formulation was prepared as follows: [0283] 1. A 400 mM (2.times.) monobasic sodium phosphate solution and a 400 mM (2.times.) dibasic sodium phosphate solution were prepared by dissolving the corresponding sodium phosphate species in water (volumetric flaks). [0284] 2. The 400 mM (2.times.) monobasic and dibasic sodium phosphate solutions were mixed under pH-control to prepare a 400 mM (2.times.) sodium phosphate buffer solution at pH=8.0 (vehicle solution). [0285] 3. Brincidofovir was dissolved in the vehicle solution (400 mM (2.times.) sodium phosphate solution at pH 8.0) at 2.times. of the final concentration using a 500 mL volumetric flask. A 1 N sodium hydroxide solution was added at a volume of V.sub.NaOH [mL]=1.65.times.M.sub.BCV [g]. Q.S. with DI water yielded the formulation at the desired brincidofovir concentration in 200 mM sodium phosphate buffer at pH=8.0. [0286] 4. For preparation of lower brincidofovir concentrations the prepared formulation was diluted with the corresponding vehicle to the desired strength.

Stability Study I: Brincidofovir Formulation Containing Tonicifier (Dextrose)

[0287] Design and Setup:

[0288] In this study, the chemical and physical stability of formulation 57 was evaluated. Testing of the chemical degradation was performed in an accelerated stability study setup applying heat as an accelerator/stressor. An Arrhenius analysis was carried out providing an API half-life and a degradation rate at lower temperatures. Physical stability was evaluated at the desired storage temperature (2-8.degree. C.); a benchmark/reference condition was likewise included in the study. The setup of the study is specified below:

Testing Solutions:

[0289] 10 mg/mL brincidofovir, 200 mM sodium phosphate, 69 mM dextrose, pH=8.0, [0290] 15 mg/mL brincidofovir, 200 mM sodium phosphate, 69 mM dextrose, pH=8.0, [0291] Vehicle control 1: 200 mM sodium phosphate, 69 mM dextrose, pH=8.0 (only at 75.degree. C.), [0292] Vehicle control 2: 200 mM sodium phosphate, pH=8.0 (only at 75.degree. C.).

Stability Station Temperatures:

[0292] [0293] Accelerated stability study: 50.degree. C., 60.degree. C., 65.degree. C., 70.degree. C., 75.degree. C., [0294] Physical degradation and benchmark stability study: 2-8.degree. C., 25.degree. C.

Time Points:

[0294] [0295] Accelerated stability study: initial, 2, 6, 9, 14 days, [0296] Physical degradation and benchmark stability study: initial, 14 and 28 days, [0297] Vehicle control: initial, 1, 5, 8, and 14 days.

Tests:

[0297] [0298] Brincidofovir formulation: appearance, pH, c(BCV) recovery (HPLC), purity (HPLC), and liquid particle counting (physical degradation and benchmark stability study), [0299] Vehicle control: appearance and pH.

[0300] The stability testing results (accelerated conditions), i.e., the changes of pH and trends of brincidofovir recovery over the course of the stability study are summarized in Table 8 and Table 9 for the formulations at 10 mg/mL and 15 mg/mL respectively. The accelerated stability study with strong biphasic rate trends did not allow for an Arrhenius analysis and calculation of a brincidofovir half-life or extrapolation to degradation rates at typical storage conditions. The test results on the vehicle controls in the accelerated stability study (75.degree. C. station only) are summarized in Table 5.

TABLE-US-00005 TABLE 5 Accelerated stability study results of the 10 mg/mL brincidofovir formulation (Study I) Sample conditions Station Mea- Observed BCV BCV temp. Time Appear- sured c(BCV).sup.2 recovery.sup.2,3 purity (.degree. C.) point ance.sup.1 pH (mg/mL) (%) (%) 50 0 Clear, 8.1 10.1 100 99 Yellow (0) 2 Clear, 7.8 9.0 89 91 Yellow (0) 6 Clear, 7.5 9.0 89 96 Yellow (0) 9 Clear, 7.4 8.9 88 96 Yellow (1) 14 Clear, 7.4 8.8 87 95 Yellow (1) 60 0 Clear, 8.1 10.1 100 99 Yellow (0) 2 Clear, 7.5 9.0 89 98 Yellow (0) 6 Clear, 7.3 9.2 91 96 Yellow (2) 9 Clear, 7.4 9.2 91 96 Yellow (2) 14 Clear, 7.3 9.1 90 96 Yellow (2) 65 0 Clear, 8.1 10.1 100 99 Yellow (0) 2 Clear, 7.5 9.5 94 99 Yellow (1) 6 Clear, 7.3 9.4 93 97 Yellow (3) 9 Clear, 7.3 9.3 92 96 Yellow (3) 14 Clear, 7.2 9.0 89 95 Yellow (4) 70 0 Clear, 8.1 10.1 100 99 Yellow (0) 2 Clear, 7.4 9.3 92 97 Yellow (4) 6 Clear, 7.2 9.3 92 96 Yellow (5) 9 Clear, 7.1 9.2 91 95 Yellow (6) 14 Clear, 7.0 8.7 86 93 Yellow (7) 75 0 Clear, 8.1 10.1 100 99 Yellow (0) 2 Clear, 7.3 9.4 93 98 Yellow (6) 6 Clear, 6.9 9.1 90 93 Yellow (8) 9 Clear, 6.9 8.7 86 90 Yellow (8) 14 Clear, 6.8 7.9 78 84 Yellow (9) .sup.1The yellow color of the solution was expressed on a scale of 0 to 10: 0, colorless; 5, mustard; 10, amber. .sup.2Average value of three (3) replicates. .sup.3BCV recovery = c(BCV).sub.Day n/c(BCV).sub.Day 0 .times. 100%.

TABLE-US-00006 TABLE 6 Accelerated stability study results of the 15 mg/mL brincidofovir formulation (Study I) Sample conditions Station Time Mea- Observed BCV BCV temp. point Appear- sured c(BCV).sup.2 recovery.sup.2,3 purity (.degree. C.) (day) ance.sup.1 pH (mg/mL) (%) (%) 50 0 Clear, 8.1 15.0 100 99 Yellow (0) 2 Clear, 7.8 13.6 90 98 Yellow (0) 6 Clear, 7.5 13.6 90 97 Yellow (0) 9 Clear, 7.5 13.3 88 98 Yellow (2) 14 Clear, 7.4 13.7 91 97 Yellow (2) 60 0 Clear, 8.1 15.0 100 99 Yellow (0) 2 Clear, 7.6 13.8 92 98 Yellow (0) 6 Clear, 7.3 13.8 92 97 Yellow (2) 9 Clear, 7.4 13.7 91 97 Yellow (2) 14 Clear, 7.3 13.8 92 96 Yellow (3) 65 0 Clear, 8.1 15.0 100 99 Yellow (0) 2 Clear, 7.5 14.0 93 99 Yellow (1) 6 Clear, 7.3 14.0 93 97 Yellow (3) 9 Clear, 7.3 13.8 92 96 Yellow (4) 14 Clear, 7.2 13.1 87 95 Yellow (5) 70 0 Clear, 8.1 15.0 100 99 Yellow (0) 2 Clear, 7.4 13.9 93 98 Yellow (3) 6 Clear, 7.2 13.9 92 96 Yellow (5) 9 Clear, 7.2 13.8 91 95 Yellow (5) 14 Clear, 7.1 13.1 87 93 Yellow (7) 75 0 Clear, 8.1 15.0 100 99 Yellow (0) 2 Clear, 7.3 14.1 94 98 Yellow (5) 6 Clear, 6.9 13.7 90 92 Yellow (8) 9 Clear, 6.9 12.9 86 89 Yellow (9) 14 Clear, 6.7 11.7 78 83 Yellow (10) .sup.1The yellow color of the solution was expressed on a scale of 0 to 10: 0, colorless; 5, mustard; 10, amber. .sup.2Average value of three (3) replicates. .sup.3BCV recovery = c(BCV)Day n/c(BCV)Day 0 .times. 100%. It was defined as 100% at the initial time point.

TABLE-US-00007 TABLE 7 Accelerated stability study: Results of vehicle controls (Study I) Sample conditions Station Time Measured Sample temp. (.degree. C.) point (day) Appearance.sup.1 pH Vehicle 75 0 Clear, Yellow (0) 8.1 control 1 1 Clear, Yellow (5) 7.3 (dextrose- 5 Clear, Yellow (6) 7.1 added) 8 Clear, Yellow (7) 6.9 14 Clear, Yellow (9) 6.9 Vehicle 75 0 Clear, Yellow (0) 8.1 control 2 1 Clear, Yellow (0) 8.0 (dextrose- 5 Clear, Yellow (0) 8.0 free) 8 Clear, Yellow (0) 8.0 14 Clear, Yellow (0) 8.0 .sup.1The yellow color of the solution was expressed on a scale of 0 to 10: 0, colorless; 5, mustard; 10, amber.

[0301] The study showed degradation of brincidofovir of 16% (50.degree. C.) to 22% (75.degree. C.) over the course of two weeks. While some loss in brincidofovir recovery (assay loss) was observed, the purity of brincidofovir remained at 95% and 93% respectively for temperatures up to 70.degree. C. At the 75.degree. C. station, up to 16% impurities were observed at both starting concentrations. Degradation at higher temperatures was accompanied by a strong pH shift towards lower pH values and coloration of the solutions. The pH shifts and the degradation rates were higher for the initial time point, and after 2 days the degradation and pH shifting slowed down significantly (bi-phasic trends not following a first-order decomposition).

Physical Degradation and Benchmark Stability Study

[0302] The results of the physical/benchmark stability for both tested formulations are summarized in Table 11 and Table 12, respectively. The purities of the formulations at different temperatures were determined by comparing chromatograms obtained at different temperatures and time points during the study. Degradation between 6% (2-8.degree. C.) and 8% (25.degree. C.) was observed over one month while the purity of brincidofovir stagnated at 99% indicating a brincidofovir mass deficit.

TABLE-US-00008 TABLE 8 Physical degradation and benchmark stability study results of the 10 mg/mL brincidofovir formulation (Study I) Sample conditions Station Time Mea- Observed BCV BCV temp. point Appear- sured c(BCV).sup.1 recovery.sup.1,3 purity (.degree. C.) (day) ance pH (mg/mL) (%) (%) 2-8 0 Clear, 8.1 10.1 100 99 Colorless 14 Clear, 8.0 9.6 95 100 Colorless 28 Clear, 8.1 9.5 94 99 Colorless 25 0 Clear, 8.1 10.1 100 99 Colorless 14 Clear, 7.9 9.3 92 99 Colorless 28 Clear, 8.0 9.3 92 99 Colorless .sup.1Average value of three (3) replicates. .sup.2Integration region: 2.00-13.00 min. .sup.3BCV recovery = c(BCV)Day n/c(BCV)Day 0 .times. 100%. It was defined as 100% at the initial time point.

TABLE-US-00009 TABLE 9 Physical degradation and benchmark stability study results of the 15 mg/mL brincidofovir formulation (Study I) Sample conditions Station Time Mea- Observed BCV BCV temp. point Appear- sured c(BCV).sup.1 recovery.sup.1,3 purity (.degree. C.) (day) ance pH (mg/mL) (%) (%) 2-8 0 Clear, 8.1 15.0 100 99 Colorless 14 Clear, 8.0 14.3 95 100 Colorless 28 Clear, 8.1 14.6 97 100 Colorless 25 0 Clear, 8.1 15.0 100 99 Colorless 14 Clear, 7.9 13.8 92 99 Colorless 28 Clear, 8.0 13.9 93 99 Colorless .sup.1Average value of three (3) replicates. .sup.2Integration region: 2.00-13.00 min. .sup.3BCV.sub.recovery = c(BCV).sub.Day n/c(BCV).sub.Day 0 .times. 100%. It was defined as 100% at the initial time point.

[0303] Light Obstruction Measurement:

[0304] To prepare samples for liquid particle counting, under best clean conditions the formulation samples from six serum vials (1.0 mL filling) were carefully combined (avoiding foaming) into a particle-free centrifuge tube. The tube was then capped and sonicated for 1 min before the sample was subjected to liquid particle counting. Four sample strokes of 1 mL were analyzed. The drawing rate of the liquid particle counter was 10 mL/min. No significant increase in particulate matter was observed over the course of the study. The test results of the test are summarized in Table 10.

TABLE-US-00010 TABLE 10 Liquid particle counting results of the 10 and 15 mg/mL brincidofovir formulations (Study I) Stability station temperature (.degree. C.) 2-8 25 10 .mu.m 25 .mu.m 10 .mu.m 25 .mu.m Time point particle particle particle particle Formulation (day) count count count count 10 mg/mL 0 1 0 1 0 brincidofovir 14 11 1 7 0 formulation 28 4 0 5 0 15 mg/mL 0 0 0 0 0 brincidofovir 14 7 1 4 0 formulation 28 3 1 3 0

Stability Study II: Brincidofovir Formulation without Tonicifier (Dextrose)

[0305] Design and Setup:

[0306] The stability study I, designed to evaluate the chemical and physical degradation properties of the liquid brincidofovir formulation was repeated without the presence of dextrose. A color change of the solution (potential Maillard browning) and a pH-shift could be correlated with the presence of dextrose that consequently might have influenced the degradation routes and impaired the observed rates for brincidofovir degradation. The study setup for this repeated analysis was modified with regard to the tested formulation as well as stability station parameters. The exact parameters are specified below:

Testing Solutions:

[0307] 15 mg/mL brincidofovir, 200 mM sodium phosphate, pH=8.0, [0308] Vehicle control 1: 200 mM sodium phosphate, pH=8.0.

Stability Station Temperatures:

[0308] [0309] Accelerated stability study: 50.degree. C., 60.degree. C., 70.degree. C., and 75.degree. C. [0310] Physical degradation and benchmark stability study: 2-8.degree. C. and 25.degree. C.

Time Points:

[0310] [0311] Accelerated stability study: initial, 3, 6, 10, 14 days, [0312] Physical degradation and benchmark stability study: initial, 14 and 28 days.

Tests:

[0313] Brincidofovir formulation: appearance, pH, c(BCV) recovery (HPLC), purity (HPLC), and liquid particle counting (physical degradation and benchmark stability study samples only),

[0314] Vehicle control: appearance, pH, and liquid particle counting (physical degradation and benchmark stability study samples only).

[0315] Results:

[0316] The stability testing results on brincidofovir recovery and purity of the tested formulation and the vehicle control are summarized in Table 11 and Table 12, respectively.

TABLE-US-00011 TABLE 11 Accelerated stability study results of the 15 mg/mL brincidofovir formulation (Study II) Sample conditions Station Time Mea- Observed BCV BCV temp. point Appear- sured c(BCV).sup.a recovery.sup.a,b purity (.degree. C.) (day) ance pH (mg/mL) (%) (%) 50 0 Clear, 8.0 15.4 100 99 Colorless 3 Clear, 8.0 15.0 98 98 Colorless 6 Clear, 8.0 14.6 95 98 Colorless 10 Clear, 8.0 14.6 95 97 Colorless 14 Clear, 7.9 14.2 92 97 Colorless 60 0 Clear, 8.0 15.4 100 99 Colorless 3 Clear, 7.9 14.8 96 98 Colorless 6 Clear, 8.0 14.2 92 96 Colorless 10 Clear, 8.0 14.3 93 95 Colorless 14.sup.c -- -- -- -- -- 70 0 Clear, 8.0 15.4 100 99 Colorless 3 Clear, 7.9 14.6 95 96 Colorless 6 Clear, 7.9 13.8 90 94 Colorless 10 Clear, 7.9 14.0 91 92 Colorless 14 Clear, 7.9 13.2 86 90 Colorless 75 0 Clear, 8.0 15.4 100 99 Colorless 3 Clear, 7.9 14.3 93 95 Colorless 6 Clear, 7.9 13.4 87 93 Colorless 10 Clear, 7.9 13.3 86 90 Colorless 14 Clear, 7.9 12.9 84 88 Colorless .sup.aAverage value of three (3) replicates. .sup.bIntegration region: 0.00-13.00 min. .sup.cData of this time point were compromised due to oven malfunction, thus not included in the following graphic representations or calculations

TABLE-US-00012 TABLE 12 Accelerated stability study results of the vehicle control (Study II) Sample conditions Station temp. (.degree. C.) Time point (day) Appearance Measured pH n.a..sup.a 0 Clear, colorless 8.0 50 3 Clear, colorless 8.0 6 Clear, colorless 8.1 10 Clear, colorless 8.0 14 Clear, colorless 8.0 60 3 Clear, colorless 8.0 6 Clear, colorless 8.0 10 Clear, colorless 8.0 14.sup.b -- -- 70 3 Clear, colorless 8.0 6 Clear, colorless 8.1 10 Clear, colorless 8.0 14 Clear, colorless 8.1 75 3 Clear, colorless 8.0 6 Clear, colorless 8.0 10 Clear, colorless 8.0 14 Clear, colorless 8.0 .sup.aNot applicable. .sup.bData of this time point were compromised due to oven malfunction, thus not included in the following graphic representations.

[0317] It is noted that the values for brincidofovir recovery and purity do not align. This is at least partially attributed to the fact that the injection peak area of the chromatographic trace (0.00-2.1 min) was not integrated, which would could overestimate the purity value, since the non-integrated retention time area likely accommodates known degradants. Additionally, a baseline separation between brincidofovir and its deamination product could not be achieved with the existing method, which might affect recovery values (degradation) and purity values.

[0318] The obtained recovery data were applied to determine the degradation rate at the different temperatures via an Arrhenius analysis, i.e., by logarithmically plotting the brincidofovir recovery as a function of time, and determining the rate constant through a linear fit. The half-life for the dextrose-free 15 mg/mL brincidofovir formulation was so determined to 729 days and 295 days at stability station temperatures of 5.degree. C. and 25.degree. C. respectively. The Arrhenius analysis is illustrated in FIG. 2.

Physical Degradation and Benchmark Stability Study

[0319] The results of the physical/benchmark stability for the studied formulation and the vehicle control are summarized in Table 13 and Table 14, respectively.

TABLE-US-00013 TABLE 13 Physical degradation and benchmark stability study results of the 15 mg/mL brincidofovir formulation (Study II) Sample Station Time Mea- Observed BCV BCV temp. point Appear- sured c(BCV).sup.1 recovery.sup.1,3 purity (.degree. C.) (day) ance pH (mg/mL) (%) (%) 2-8 0 Clear, 8.0 15.4 100 99 Colorless 14 Clear, 8.0 14.9 97 100 Colorless 28 Clear, 8.1 15.2 99 99 Colorless 25 0 Clear, 8.0 15.4 100 99 Colorless 14 Clear, 8.0 14.9 97 99 Colorless 28 Clear, 8.0 15.2 99 99 Colorless .sup.1Average value of three (3) replicates. .sup.3BCV recovery = c(BCV).sub.Day n/c(BCV).sub.Day 0 .times. 100%. It was defined as 100% at the initial time point.

TABLE-US-00014 TABLE 14 Physical degradation and benchmark stability study results of the vehicle control (Study II) Sample conditions Station temp. (.degree. C.) time point (day) Appearance Measured pH n.a..sup.1 0 Clear, colorless 8.0 2-8 14 Clear, colorless 8.1 28 Clear, colorless 8.1 25 14 Clear, colorless 8.1 28 Clear, colorless 8.0 .sup.1Not applicable.

[0320] For the analyzes of particulate matter, the formulation sample from one serum vial (5.0 mL filling) was sonicated for 1 min before decrimping of the vial. The tonicifier-free sample was subjected to liquid particle counting as described above for the tonicifier containing formulations. The Results of the analysis are summarized in Table 15.

TABLE-US-00015 TABLE 15 Liquid particle counting results of the tonicifier-free 15 mg/mL brincidofovir formulation and vehicle control (Study II) Stability station temperature (.degree. C.) 2-8 25 Time 10 .mu.m 25 .mu.m 10 .mu.m 25 .mu.m point particle particle particle particle Solution (day) count count count count 15 mg/mL 0 0 0 0 0 brincidofovir 14 80 2 45 0 formulation 28 87 0 4 0 Vehicle 0 0 0 0 0 control 14 0 0 0 0 28 0 0 0 0

[0321] The degradation of brincidofovir in the tonicifier-free formulation at storage conditions (2-8.degree. C. and 25.degree. C.) is negligible over the course of one month. The nominal degradation of 1% is within the error of brincidofovir quantification; a mass deficit or particulate matter were not ob served.

Assessment of Brincidofovir Adsorption to Vials and Stoppers

[0322] Design and Setup:

[0323] An adsorption assessment on brincidofovir was performed to test for adsorption of brincidofovir to vials and/or stoppers. For this, different storage conditions that might have an impact (e.g. vial orientation) were assessed. Solutions of brincidofovir were studied at the formulation strength (10 mg/mL) and at a low concentration (0.2 mg/mL).

[0324] To test for adsorption, a treatment with an organic solvent (methanol) that is suited to re-solubilize adsorbed material was implemented at the different time points and compared to treatment with water or untreated samples. An observation of higher brincidofovir concentration in the samples containing methanol (after normalization for dilution) would have been indicative of an adsorption effect. Differences of an imposed volume contraction between the applied solvents are not considered. The study was conducted using solutions of 10 mg/mL and 0.2 mg/mL of brincidofovir in 200 mM sodium phosphate (pH=8.0), at temperatures of 2-8.degree. C. and 25.degree. C. Appearance, pH, brincidofovir recovery (HPLC), and purity (HPLC) of the solutions were tested initially and after 1, 2, and 7 days. Some samples were further treated by addition of methanol (1.0 mL) or water (1.0 mL). The results of the study are summarized in Table 16 and Table 17.

TABLE-US-00016 TABLE 16 Test results of the 10 mg/mL brincidofovir formulation in adsorption assessment Sample conditions Station Time Observed BCV BCV temp. Vial point Measured c(BCV).sup.a Recovery.sup.a,b purity (.degree. C.) orientation Treatment (day) Appearance pH (mg/mL) (%) (%) 2-8 Upright No 0 Clear, Colorless 8.0 10.5 100.00 99.80 treatment 1 Clear, Colorless 8.0 10.5 100.36 99.55 2 Clear, Colorless 8.0 10.5 99.57 99.67 7 Clear, Colorless 8.0 10.6 99.93 99.61 Add 0 Clear, Colorless 8.3 10.4 100.00 99.90 methanol 1 Clear, Colorless 8.4 10.2 98.10 99.52 2 Clear, Colorless 8.4 10.3 98.90 99.73 7 Clear, Colorless 8.4 10.5 100.41 99.43 Add water 0 Clear, Colorless 8.1 10.5 100.00 99.32 1 Clear, Colorless 8.1 10.3 98.56 99.72 2 Clear, Colorless 8.0 10.6 101.14 99.47 7 Clear, Colorless 8.1 10.6 100.68 99.61 Inverted No 0.sup.c Clear, Colorless 8.0 10.5 100.00 99.80 treatment 1 Clear, Colorless 8.1 10.5 100.11 99.74 2 Clear, Colorless 8.0 10.4 99.18 99.78 7 Clear, Colorless 8.0 10.5 99.41 99.77 Add 0.sup.c Clear, Colorless 8.3 10.4 100.00 99.90 methanol 1 Clear, Colorless 8.4 10.6 102.34 99.46 2 Clear, Colorless 8.4 10.7 103.30 99.64 7 Clear, Colorless 8.4 10.7 102.56 99.44 Add water 0.sup.c Clear, Colorless 8.1 10.5 100.00 99.32 1 Clear, Colorless 8.1 10.4 99.50 99.46 2 Clear, Colorless 8.0 10.4 99.53 99.47 7 Clear, Colorless 8.0 10.6 100.21 99.54 25 Upright No 0 Clear, Colorless 8.0 10.5 100.00 99.80 treatment 1 Clear, Colorless 8.1 10.5 99.78 99.47 2 Clear, Colorless 8.0 10.4 99.02 99.66 7 Clear, Colorless 8.0 10.4 98.72 99.73 Add 0 Clear, Colorless 8.3 10.4 100.00 99.90 methanol 1 Clear, Colorless 8.4 10.6 101.66 99.57 2 Clear, Colorless 8.3 10.3 98.83 99.61 7 Clear, Colorless 8.4 10.5 100.52 99.74 Add water 0 Clear, Colorless 8.1 10.5 100.00 99.32 1 Clear, Colorless 8.1 10.6 101.21 99.61 2 Clear, Colorless 8.1 10.6 101.00 99.76 7 Clear, Colorless 8.1 10.6 100.13 99.70 Inverted No 0.sup.c Clear, Colorless 8.0 10.5 100.00 99.80 treatment 1 Clear, Colorless 8.1 10.6 101.40 99.57 2 Clear, Colorless 8.0 10.6 101.08 99.82 7 Clear, Colorless 8.0 10.5 99.22 99.60 Add 0.sup.c Clear, Colorless 8.3 10.4 100.00 99.90 methanol 1 Clear, Colorless 8.4 10.7 102.92 99.60 2 Clear, Colorless 8.4 10.6 101.97 99.61 7 Clear, Colorless 8.4 10.5 100.47 99.81 Add water 0.sup.c Clear, Colorless 8.1 10.5 100.00 99.32 1 Clear, Colorless 8.0 10.5 100.55 99.47 2 Clear, Colorless 8.0 10.5 100.56 99.63 7 Clear, Colorless 8.0 10.5 99.92 99.68 .sup.1Average value of the three (3) replicates of samples that received treatment. .sup.2BCV recovery = c(BCV).sub.Day n/c(BCV).sub.Day 0 .times. 100%. It was defined as 100% at the initial time point. BCV recovery was defined as 100.00% on the initial time point for samples with and without treatment. .sup.3Not applicable for samples without treatment. .sup.4The sample for the initial time point was not subjected to any conditions.

TABLE-US-00017 TABLE 17 Test results of the 0.2 mg/mL brincidofovir formulation in adsorption assessment Sample conditions Station Time Observed BCV BCV temp. Vial point Measured c(BCV).sup.a Recovery.sup.a,b purity (.degree. C.) orientation Treatment (day) Appearance pH (mg/mL) (%) (%) 2-8 Upright No 0 Clear, Colorless 8.0 0.19 100.00 99.37 treatment 1 Clear, Colorless 8.1 0.19 100.23 99.37 2 Clear, Colorless 8.1 0.19 100.01 99.45 7 Clear, Colorless 8.1 0.20 100.26 99.52 Add 0 Clear, Colorless 8.3 0.20 100.00 99.28 methanol 1 Clear, Colorless 8.4 0.20 100.91 99.45 2 Clear, Colorless 8.4 0.20 100.79 99.55 7 Clear, Colorless 8.4 0.20 101.74 99.57 Add water 0 Clear, Colorless 8.0 0.19 100.00 99.48 1 Clear, Colorless 8.1 0.20 100.38 99.27 2 Clear, Colorless 8.1 0.20 100.78 99.35 7 Clear, Colorless 8.1 0.20 100.30 99.37 Inverted No 0.sup.c Clear, Colorless 8.0 0.19 100.00 99.37 treatment 1 Clear, Colorless 8.1 0.19 99.21 99.38 2 Clear, Colorless 8.1 0.19 99.68 99.50 7 Clear, Colorless 8.0 0.19 99.61 99.66 0.sup.c Clear, Colorless 8.3 0.20 100.00 99.28 Add 1 Clear, Colorless 8.5 0.20 100.09 99.51 methanol 2 Clear, Colorless 8.4 0.20 100.17 99.54 7 Clear, Colorless 8.4 0.20 100.04 99.66 Add water 0.sup.c Clear, Colorless 8.0 0.19 100.00 99.48 1 Clear, Colorless 8.1 0.19 99.81 99.30 2 Clear, Colorless 8.0 0.19 99.67 99.42 7 Clear, Colorless 8.1 0.19 99.27 99.62 25 Upright No 0 Clear, Colorless 8.0 0.19 100.00 99.37 treatment 1 Clear, Colorless 8.1 0.19 100.18 99.46 2 Clear, Colorless 8.0 0.19 100.12 99.27 7 Clear, Colorless 8.1 0.19 99.92 99.42 Add 0 Clear, Colorless 8.3 0.20 100.00 99.28 methanol 1 Clear, Colorless 8.4 0.20 101.00 99.54 2 Clear, Colorless 8.4 0.20 101.25 99.45 7 Clear, Colorless 8.4 0.20 101.87 99.53 Add water 0 Clear, Colorless 8.0 0.19 100.00 99.48 1 Clear, Colorless 8.1 0.20 100.60 99.54 2 Clear, Colorless 8.1 0.20 100.22 99.31 7 Clear, Colorless 8.1 0.20 100.45 99.44 Inverted No 0.sup.c Clear, Colorless 8.0 0.19 100.00 99.37 treatment 1 Clear, Colorless 8.1 0.19 99.38 99.45 2 Clear, Colorless 8.1 0.19 98.91 99.55 7 Clear, Colorless 8.1 0.19 98.96 99.62 Add 0.sup.c Clear, Colorless 8.3 0.20 100.00 99.28 methanol 1 Clear, Colorless 8.5 0.20 100.25 99.62 2 Clear, Colorless 8.4 0.20 99.70 99.39 7 Clear, Colorless 8.4 0.20 99.76 99.52 Add water 0.sup.c Clear, Colorless 8.0 0.19 100.00 99.48 1 Clear, Colorless 8.1 0.19 99.28 99.37 2 Clear, Colorless 8.1 0.19 99.31 99.43 7 Clear, Colorless 8.1 0.19 98.99 99.53 .sup.aAverage value of the three (3) replicates of samples that received treatment. .sup.bBCV recovery = c(BCV).sub.Day n/c(BCV).sub.Day 0 .times. 100%. It was defined as 100% at the initial time point. BCV recovery was defined as 100.00% on the initial time point for samples with and without treatment. .sup.cThe sample for the initial time point was not subjected to any conditions.

[0325] A comparison of differently treated samples did not reveal any evidence for an adsorption phenomenon; no significant differences in the studied parameters, i.e., no significant change in appearance or purity and no significant loss of brincidofovir, were observed. The observed mass deficits were within the range of the sample preparation errors as expressed by the standard deviation of replicate preparations.

Formulation Compatibility Assessments

[0326] An array of materials was tested for compatibility with the formulation. This includes testing materials from the manufacturing stage, pre-clinical toxicology as well as clinical tests. The applied compatibility tests evaluate 1) a potential loss of brincidofovir by adsorption or precipitation (appearance, brincidofovir recovery by HPLC) and 2) a shift in pH that could compromise stability. Materials that were tested include the infusion vehicle, filters for sterilization, product vials and stoppers, infusion systems for testing animals (rats) and infusion bags and IV systems for clinical applications.

[0327] A brincidofovir formulation comprising 10 mg/mL brincidofovir, 200 mM sodium phosphate, and 69 mM dextrose (pH=8.0) was diluted with the testing infusion vehicle (5% dextrose solution) to yield c(BCV)=0.5 and 1.0 mg/mL solutions. The diluted solutions were stored at ambient conditions, and samples were collected at initially at the beginning of the experiment, and after 2 h, 8 h, and 24 h and evaluated for appearance, pH, and recovery of c(BCV) (via HPLC). No significant changes in appearance, pH, or c(BCV) were observed for the tested solutions over 24 hours at ambient conditions. Detailed results of the study are summarized below (Table 18).

TABLE-US-00018 TABLE 18 Results of infusion vehicle compatibility assessment c(BCV) in infusion Time Observed BCV vehicle point Measured c(BCV).sup.a Recovery.sup.a,b (mg/mL) (h) Appearance pH (mg/mL) (%) 0.5 0 Clear, colorless 8.2 0.51 100 2 Clear, colorless 8.3 0.51 100 8 Clear, colorless 8.2 0.51 100 24 Clear, colorless 8.2 0.49 99 1.0 0 Clear, colorless 8.2 1.0 100 2 Clear, colorless 8.2 1.0 99 8 Clear, colorless 8.2 1.0 99 24 Clear, colorless 8.2 1.0 100 .sup.aAverage value of the three (3) replicates. .sup.bBCV recovery = c(BCV)Hour n/c(BCV)Hour 0 .times. 100%. It was defined as 100% for samples at initial time point.

Sterilization Filter Compatibility

[0328] Formulations of the disclosure were evaluated for sterilization filter compatibility. Aliquots of a testing brincidofovir formulation were filtered through the corresponding syringe filters as follows:

A: Filter: 0.2 .mu.m, 25 mm, syringe filter, Supor.RTM. membrane (PES), sterile; testing solution: 10 mg/mL brincidofovir, 200 mM sodium phosphate, pH=8.0; testing volume: 10 mL. B: Filter: 0.2 .mu.m, 25 mm, syringe filter, Posidyne.RTM. membrane, sterile; testing solution: 10 mg/mL brincidofovir, 200 mM sodium phosphate, pH=8.0; testing volume: 10 mL. C: Filter: 0.22 .mu.m, 33 mm, syringe filter, Durapore.RTM. PVDF membrane, .gamma.-irradiated; testing solution: 2 mg/mL brincidofovir, 4 mM sodium phosphate, pH-8, approx. 5% dextrose (50.times. dilution of 10 mg/mL brincidofovir, 200 mM sodium phosphate, pH=8.0 with 5% dextrose solution; testing volume: 50 mL.

[0329] The first and last 10% of the filtrate were collected. Samples of the collected filtrates, along with samples of the pre filtration formulation, were assessed for their appearance, pH, and c(BCV).

TABLE-US-00019 TABLE 19 Results of sterilization filter compatibility assessment Observed BCV Filter Measured c(BCV).sup.a Recovery.sup.a,b membrane Conditions Appearance pH (mg/mL) (%) PES Pre filtration Clear, 8.1 9.9 100 colorless First 10% filtrate Clear, 8.0 9.8 99 volume colorless Last 10% filtrate Clear, 8.0 9.8 99 volume colorless Posidyne Pre filtration Clear, 8.1 9.9 100 colorless First 10% filtrate Clear, 8.0 9.7 98 volume colorless Last 10% filtrate Clear, 8.0 9.8 99 volume colorless PVDF Pre filtration Clear, 8.0 0.20 100 colorless First 10% filtrate Clear, 8.1 0.20 99 volume colorless Last 10% filtrate Clear, 8.1 0.20 100 volume colorless .sup.aAverage value of the three (3) replicates. .sup.bBCV recovery = c(BCV).sub.Post filtration/c(BCV).sub.Pre filtration .times. 100%; defined as 100% for pre filtration samples.

[0330] No significant changes in appearance, pH, or c(BCV) were observed when comparing the filtrate solutions with the corresponding pre-filtration samples and no impurity was introduced during filtration.

Drug Product Vial and Stopper Compatibility

[0331] Formulations of the disclosure were evaluated for sterilization filter compatibility. The testing formulation (5 mL, 10 mg/mL brincidofovir, 200 mM sodium phosphate, pH=8.0) was dispensed into a set of Afton sterilized, Ready-To-Fill.RTM. vials. The vials were stoppered with West NovaPure.RTM. stoppers and crimped with Afton Ready-To-Fill.RTM. sterilized seals, and stored for 6 hours at ambient conditions in upright and inverted orientations. Samples of the filling solution were collected before filling and after 6 hours of storage under the respective condition. The collected samples were assessed for their appearance, pH, and c(BCV). No significant changes in appearance, pH, or c(BCV) were observed for the tested formulation after 6 hours of contact with the tested vials and stoppers. No additional impurity was introduced following contact.

TABLE-US-00020 TABLE 20 Results of drug product vial and stopper compatibility assessment Observed BCV Storage Measured c(BCV).sup.a Recovery .sup.a,b condition Appearance pH (mg/mL) (%) Pre filling Clear, colorless 8.0 9.5 100 Upright, 6 h Clear, colorless 8.0 9.5 100 Inverted, 6 h Clear, colorless 8.0 9.4 99 .sup.aAverage value of the three (3) replicates. .sup.bBCV recovery = c(BCV)Hour n/c(BCV)Pre filling .times. 100%. It was defined as 100% for pre filling samples.

Preclinical Material Compatibility Assessment (Rats)

Infusion System Assembly (Rats)

[0332] The compatibility of the following brincidofovir formulations with an infusion system assembly for dosing of rats (consisting of a syringe, tether, catheter and syringe pump) was tested

a.) 0.2 mg/mL brincidofovir, 4 mM sodium phosphate, pH .about.8.0, approx. 4.90% dextrose; b.) 1.5 mg/mL brincidofovir, 30 mM sodium phosphate, pH=8.0, approx. 4.25% dextrose.

[0333] Both solutions were prepared via dilution of the developed brincidofovir formulation (10 mg/mL brincidofovir, 200 mM sodium phosphate, pH=8.0) with a 5% dextrose solution. The testing procedure can be summarized as follows: [0334] 1. The appearance, pH, and c(BCV) of the bulk testing solutions (0.2 mg/mL BCV, 4 mM sodium phosphate, pH-8.0, approx. 4.90% dextrose, and 1.5 mg/mL BCV, 30 mM sodium phosphate, pH .about.8.0, approx. 4.25% dextrose were assessed pre-contact. [0335] 2. Approximately 5 mL of the testing solution was drawn into a syringe which was then connected to the assembled infusion system (Vascular Access Harness (VAH) tether kit, the VAH port for rat jacket, and catheter). [0336] 3. The infusion system was manually filled with the testing solution. Approximately 200 .mu.L of the testing solution were pushed out of the infusion system and collected as an end-of-line sample (t.sub.0) for the assessments of appearance and c(BCV). [0337] 4. The syringe of the testing infusion system was mounted onto the syringe pump, and a flow rate was set to a flow of 1.2 mL/h (20 .mu.L/min) for a total dispensing volume of 2.4 mL (120 min). [0338] 5. The entire flow-through was collected over the course of the experiment. At the respective time points (t.sub.10min, t.sub.105 min), end-of-line samples (approx. 200 .mu.L) were collected for analysis. [0339] 6. The pH of the bulk flow-through was measured at the end of the experiment (tin min), since the volume of samples acquired from previous time points (200 .mu.L) was too small for a standard pH measurement. The results of the study are summarized in Table 21.

TABLE-US-00021 [0339] TABLE 21 Results of preclinical infusion system compatibility assessment Testing solution Observed BCV c(BCV) Measured c(BCV).sup.a Recovery .sup.a,b (mg/mL) Conditions Appearance pH (mg/mL) (%) 0.2 Pre contact (bulk Clear, colorless 8.0 0.21 100 (Test #1) solution) t.sub.0 Clear, colorless n.a. 0.19 90 t.sub.10 min Clear, colorless n.a. 0.16 93 t.sub.105 min Clear, colorless n.a. 0.20 94 t.sub.120 min (bulk solution) Clear, colorless 8.0 n.a. n.a. 1.5 Pre contact (bulk Clear, colorless 8.2 1.5 100 (Test #2) solution) t.sub.0 Clear, colorless n.a. 1.5 100 t.sub.10 min Clear, colorless n.a. 1.5 100 t.sub.105 min Clear, colorless n.a. 1.5 100 t.sub.120 min (bulk solution) Clear, colorless 8.2 n.a. n.a. .sup.aAverage value of the three (3) replicates. .sup.bBCV recovery = c(BCV)Minute n/c(BCV)Pre contact .times. 100%, defined as 100% for pre contact samples. n.a.: Not available or not applicable.

[0340] For test #1 (0.2 mg/mL brincidofovir), an initial 10% loss in c(BCV) was observed, which can be assigned to absorption to the testing infusion system. At low concentrations an initial adsorption effect (surface saturation) is frequently observed, in the present case a full recovery of the observed loss is not observed over the time of testing; at t=105 min a brincidofovir loss of 6% was observed. No significant changes in appearance or pH were observed. For test #2, no significant loss in c(BCV) or changes in appearance or pH were observed over the course of the experiment.

Clinical Material Assessment

Infusion Bag Compatibility

[0341] A standard mini-infusion bag (100 mL) was subjected to material compatibility testing with the following formulation: [0342] 15 mg/mL brincidofovir, 200 mM sodium phosphate, pH=8.0. The tested formulation strengths after dilution into the infusion bag were approximately: [0343] 0.1 mg/mL brincidofovir, 5.3 mM sodium phosphate, pH.apprxeq.8.0 (in 5% dextrose), [0344] 1.0 mg/mL brincidofovir, 13.3 mM sodium phosphate, pH.apprxeq.8.0 (in 5% dextrose).

[0345] The testing clinical infusion bag is specified as follows: [0346] 5% dextrose for injection, USP, 100 mL VIAFLEX infusion bag

[0347] The developed brincidofovir formulation (15 mg/mL brincidofovir, 200 mM sodium phosphate, pH=8.0) was diluted to c(BCV)=0.1 and c(BCV)=1.0 mg/mL by injection into the infusion bags. The infusion bags were then stored at ambient conditions, and samples were collected from the infusion bags at the time points (t.sub.0, t.sub.1h, t.sub.8h, and t.sub.24h) for assessments of appearance, pH, and c(BCV). As a control, the 5% dextrose solution was removed from a separate infusion bag, and external dilutions (standard laboratory glassware) of the testing formulation with the 5% dextrose solution were performed applying identical dilution factors. Samples of the external diluted solution were likewise analyzed. The results are summarized in Table 22.

TABLE-US-00022 TABLE 22 Results of clinical infusion bag compatibility assessment c(BCV) in testing infusion Observed BCV bag Appear- Measured c(BCV).sup.a Recovery .sup.a,b (mg/mL) Conditions ance pH (mg/mL) (%) 0.1 External Clear, 7.9 0.098 106 dilution colorless t.sub.0 Clear, 7.9 0.092 100 colorless t.sub.1 h Clear, 7.9 0.093 101 colorless t.sub.8 h Clear, 7.9 0.093 101 colorless t.sub.24 h Clear, 7.9 0.093 101 colorless 1.0 External Clear, 8.2 0.94 108 dilution colorless t.sub.0 Clear, 8.2 0.87 100 colorless t.sub.1 h Clear, 8.2 0.87 100 colorless t.sub.8 h Clear, 8.2 0.87 100 colorless t.sub.24 h Clear, 8.1 0.86 99 colorless .sup.aAverage value of the three (3) replicates. .sup.bBCV recovery = c(BCV).sub.Hour n/c(BCV).sub.hour 0 .times. 100%. It was defined as 100% for t.sub.0 samples.

[0348] No significant change in appearance, pH, or c(BCV) was observed for the diluted solutions in the testing infusion bag at either c(BCV). The higher brincidofovir recoveries obtained from the external dilutions most likely resulted from the fact that infusion bags are generally overfilled (105-115 mL with an average of 110 mL, for 100 mL bags--information communicated by Baxter). By applying an identical dilution factor and using the to time point (dilution into the infusion bag) for normalization (defined as 100%) the concentrations determined for the external dilution are overestimated. An internal study on a single infusion bag assessed the volume of the infusion solution in a 100 mL infusion bag with .apprxeq.109 mL.

Clinical Infusion System Compatibility

[0349] Two infusion systems (Baxter Non-DEHP CONTINUO-FLO solution set with 2 INTERLINK injection sites, male Luer-lok adaptor; and Brown Rate Flow.RTM. regulator IV set with 15 micron filter, 1 non needle free injection site) were tested for compatibility with the following formulation: [0350] 15 mg/mL brincidofovir, 200 mM sodium phosphate, pH=8.0. The tested formulation strengths after dilution into the infusion bag were approximately: [0351] 0.1 mg/mL brincidofovir, 5.3 mM sodium phosphate, pH.apprxeq.8.0 (in 5% dextrose), [0352] 1.0 mg/mL brincidofovir, 13.3 mM sodium phosphate, pH.apprxeq.8.0 (in 5% dextrose). The testing procedure can be summarized as follows: [0353] 1. The developed brincidofovir formulation (15 mg/mL brincidofovir, 200 mM sodium phosphate, pH 8.0), was injected into infusion bags (reservoir function) mentioned above to yield the testing solutions at c(BCV)=0.1 or 1.0 mg/mL. A sample was collected from the infusion bag for assessment of c(BCV) (t.sub.24 h, bag). [0354] 2. The infusion bag was incubated ("aged") for 24 h at ambient conditions before another sample (t.sub.0, bag) was collected from the infusion bag for assessment (appearance, pH, c(BCV)). [0355] 3. The tested infusion system was attached to the infusion bag and manually filled with solution from the infusion bag. Approximately 1 mL (triplicates) of the testing solution was collected as an end-of-line sample for analysis (t.sub.0, eol). [0356] 4. The testing infusion system was shut off, and stored at ambient conditions until the next time point. Approximately 17 mL of the testing solution (volume of the infusion system) was drained before another end-of-line sample (1 mL, triplicates) was collected for analysis (t.sub.10 min, eol, t.sub.3 h, eol, and t.sub.6 h, eol).

TABLE-US-00023 [0356] TABLE 23 Results of clinical infusion system compatibility assessment c(BCV) Testing in infusion Observed BCV infusion bag Measured c(BCV).sup.a Recovery.sup.a,b system (mg/mL) Conditions Appearance pH (mg/mL) (%) Baxter 0.1 t.sub.-24 h, bag n.a. n.a. 0.090 100 t.sub.0, bag Clear, colorless 7.7 0.091 100 t.sub.0, eol Clear, colorless 7.7 0.088 96 t.sub.10 min, eol Clear, colorless 7.7 0.092 100 t.sub.3 h, eol Clear, colorless 7.7 0.091 100 t.sub.6 h, eol Clear, colorless 7.7 0.092 100 1.0 t.sub.-24 h, bag n.a. n.a. 0.85 100 t.sub.0, bag Clear, colorless 8.1 0.86 100 t.sub.0, eol Clear, colorless 8.1 0.85 100 t.sub.10 min, eol Clear, colorless 8.1 0.86 100 t.sub.3 h, eol Clear, colorless 8.1 0.86 100 t.sub.6 h, eol Clear, colorless 8.1 0.86 100 B. Braun 0.1 t.sub.-24 h, bag n.a. n.a. 0.090 100 t.sub.0, bag Clear, colorless 7.8 0.091 100 t.sub.0, eol Clear, colorless 7.7 0.086 94 t.sub.10 min, eol Clear, colorless 7.7 0.091 100 t.sub.3 h, eol Clear, colorless 7.7 0.090 99 t.sub.6 h, eol Clear, colorless 7.7 0.091 100 1.0 t.sub.-24 h, bag n.a. n.a. 0.86 100 t.sub.0, bag Clear, colorless 8.1 0.86 100 t.sub.0, eol Clear, colorless 8.1 0.86 100 t.sub.10 min, eol Clear, colorless 8.1 0.86 100 t.sub.3 h, eol Clear, colorless 8.1 0.86 100 t.sub.6 h, eol Clear, colorless 8.1 0.86 100 .sup.aAverage value of the three (3) replicates. .sup.bBCV recovery = c(BCV).sub.Hour n/c(BCV).sub.Hour 0 .times. 100%. It was defined as 100% for t.sub.0, bag samples.

[0357] An initial loss in c(BCV) (up to 6%) was observed for the testing solution with low c(BCV)=0.1 mg/mL in both testing clinical infusion systems. This observation can most likely be explained with an initial saturating adsorption of the tubing material. The brincidofovir concentration recovered to approx. 100% over the course of the experiment (6 h). No significant overall changes in appearance, pH, or c(BCV) were observed for both of the tested infusion systems.

Example 4--Stability and Characteristics of Lyophilized and Reconstituted Formulations

[0358] Pre-lyophilization formulations were prepared as set forth in Table 24, below and subjected to the lyophilization process.

TABLE-US-00024 TABLE 24 Pre-lyophilization Formulations. c(bulking Form. c(buffer) c(BCV) Target Bulking agent) (w/v) # Buffer [mM] [mg/mL] pH agent [%] 1 Na-Phosphate 200 10.0 8.0 Mannitol 5 2 Na-Phosphate 200 10.0 8.0 Sucrose 9 3 Arginine 200 10.0 8.0 Mannitol 5 3a Arginine 100 10.0 8.0 Mannitol 5 3b Arginine 50 10.0 8.0 Mannitol 5 3c Arginine 25 10.0 8.0 Mannitol 5 4 Arginine 200 10.0 8.0 Sucrose 9 5 Tromethamine (Tris) 200 10.0 8.0 Mannitol 5 6 Tromethamine (Tris) 200 10.0 8.0 Sucrose 9 7 water (pH-adjusted) -- 10.0 8.0 Mannitol 5 8 water (pH-adjusted) -- 10.0 8.0 Sucrose 9

[0359] The glass transition temperature T.sub.g' (amorphous), the eutectic melting point T.sub.eu (crystalline), the onset temperature of melting T.sub.melt, onset, and the freezing temperature T.sub.freeze of the formulations 1-8 of Table 27 were determined by differential scanning calorimetry (DSC) analysis. For this, 5 .mu.L of each formulation was dispensed into an aluminum sample pan and hermetically sealed. DSC scans were performed by down and up scanning in the temperature interval of +25.degree. C. to -65.degree. C. to +25.degree. C. with a ramp rate of 5.degree. C./min. The tested formulations displayed a sharp freezing point and a well-defined melting transition. The results of the DSC study are summarized in Table 25.

TABLE-US-00025 TABLE 25 Results of the DSC analysis Form. Bulking T.sub.g'/T.sub.eu T.sub.freeze T.sub.melt T.sub.melt, onset # Buffer agent [.degree. C.] [.degree. C.] [.degree. C.] [.degree. C.] 1 Na-Phosphate Mannitol -45.0 -20.0 -0.5 -3.0 2 Na-Phosphate Sucrose -32.0 -18.0 -0.2 -3.4 3 Arginine Mannitol -45.0 -20.0 -0.5 -4.2 4 Arginine Sucrose -32.0 -18.0 -0.2 -4.2 5 Tromethamine Mannitol -16.0 -21.0 -0.4 -5.2 6 Tromethamine Sucrose -43.0 -20.0 -0.4 -5.2 7 DI water (pH- Mannitol -30.5 -21.2 -0.01 -2.2 adjusted) 8 DI water (pH- Sucrose -34.8 -16.6 -0.8 -2.9 adjusted)

Lyophilization Cycle

[0360] A conservative lyophilization cycle was applied for lyophilization of brincidofovir formulations including parameters informed by the DSC analysis (see Table 28). The cycle consisted of the following steps: freezing, annealing, primary drying and a secondary drying. The set point temperature, ramp rate, step time and load time for each step in an exemplary lyophilization cycle are summarized in Table 26.

TABLE-US-00026 TABLE 26 Lyophilization cycle parameters (Cycle # 1) T.sub.shelf Ramp Step Total (Set point) rate time time Phase Type [.degree. C.] [.degree. C./h] [min] [h] Loading Hold 5 N/A 0 0 Freeze/Anneal Hold 5 0 60 1 Freeze/Anneal Rate -50 20 165 3.8 Freeze/Anneal Hold -50 N/A 180 6.8 Freeze/Anneal Rate -8 20 174 9.7 Freeze/Anneal Hold -8 N/A 240 13.7 Freeze/Anneal Rate -35 20 81 15.0 Freeze/Anneal Hold -35 N/A 60 16.0 Primary Drying Hold -35 N/A 1200 36.0 Secondary drying Rate 20 2.5 1300 57.7 Secondary drying Hold 20 N/A 360 63.7

[0361] Sample vials (5 mL vial size, 1 mL filling volume) were processed under best clean conditions in a biosafety cabinet. For each formulation, nine samples were prepared. For each formulation condition, one sample vial was equipped with a product probe to monitor the product temperature (T.sub.product) along with the shelf temperature of the lyophilizer (T.sub.shelf) throughout the lyophilization cycle. The end of the primary drying was determined as the time when T.sub.product.apprxeq.T.sub.shelf is observed. At the end of the lyophilization cycle, the sample vials were back-filled nitrogen (N.sub.2), stoppered and removed from the lyophilizer to be inspected and analyzed.

[0362] The step time for the primary drying phase was set to 20 h. FIG. 1 displays the product temperature profiles of Formulations 1-8 during this step. In the tested formulations, after activation of the vacuum pump and drop of the product temperature to T.sub.(product)<T.sub.(shelf) at around 16 hours of total cycle time, temperature jumps were observed in the different formulations within 15 hours. The temperature jump is taken to be indicative of successful sample drying.

[0363] The lyophilized products were evaluated based on structure and uniformity of the lyophilization cakes. The observation of a `melt-back` or `cake collapses` led to a lowered ranking. Four vials from each formulation were randomly selected for the appearance evaluation. The assessment and ranking are summarized in Table 27.

TABLE-US-00027 TABLE 27 Assessment of lyophilized products and reconstitution performance Reconstitution Foam. Formulation Dissip. Ranking App. Lyophilized Product Reconst. time Meas. c(BCV) Appear. Reconst. # Buffer Bulking agent pH Cake appearance time [s] Foam* [min] pH [mg/mL] ranking ranking 1 Na-Phosphate Mannitol (5%) 7.9 White uniform solid. 50 + 7 7.8 10.4 2 4 Minor melt-back 2 Na-Phosphate Sucrose (9%) 7.9 White uniform solid. 80 ++ 45 7.8 10.5 5 7 Minor melt-back. Minor detachment from vial. 3 Arginine Mannitol (5%) 8.0 White uniform solid. 60 - -- 7.9 10.3 3 1 Minor melt-back. Minor detachment from bottom (cake collapse). 4 Arginine Sucrose (9%) 8.0 Major melt-back. 45 ++ 35 7.9 9.8 7 5 Significant cake collapse. 5 Tris Mannitol (5%) 8.0 Minor-melt back and 90 - -- 8.1 10.6 4 2 detachment from bottom. Minor cake collapse. 6 Tris Sucrose (9%) 8.0 Extensive melt-back 15 ++ 45 8.0 10.4 8 6 and cake collapse. 7 DI water(pH- Mannitol (5%) 8.3 White uniform solid. 45 + 3 8.4 10.6 1 3 adjusted) Minimal melt-back. 8 DI water(pH- Sucrose (9%) 8.3 Strong melt back and 60 ++ 60 8.3 10.3 6 4 adjusted) cake collapse. Cake detached from vial.

[0364] Of the tested formulations, the mannitol based formulations (Formulations 1, 3, 5 and 7) yielded a more compact and uniform lyophilization cake, and the sucrose based formulations (Formulations 2, 4, 6 and 8) revealed partial cake collapses as well as an increased melt-back.

Reconstitution Assessment of Lyophilized Formulations

[0365] Lyophilization product vials were weighed before and after lyophilization in order to determine the reconstitution solution volume. The lyophilized products were reconstituted with DI water. After addition of the solvent the vials were gently swirled and the reconstitution appearance as well as the reconstitution time were recorded. The reconstituted product was further analyzed for its pH and brincidofovir recovery (determined via HPLC). For a reconstitution ranking the described parameters were complemented by recording the intensity of foaming as well as the foam dissipation time. The results of the reconstitution assessment for Formulations 1-8 are summarized in Table 30.

[0366] Formulation 3 was found to lack foaming upon reconstitution, and Formulation 7 was found to be able to be reconstituted with different buffers or infusion vehicles.

Example 5--Lyophilization Process Development

[0367] For the development of a lyophilized drug product of brincidofovir, five different activities have been executed: i) lyophilization feasibility assessment, ii) lyophilization process development, iii) short-term stability studies, iv) preparation of drug product batches for stability testing, and v) material compatibility assessment.

[0368] A project overview capturing the purpose, results and the outcome of each activity is summarized in Table 28. The details of the lyophilization cycle parameters for certain lyophilization runs during process development can be found in Table 29.

TABLE-US-00028 TABLE 28 Project overview and decision rationale. Activity Cycle # Purpose Formula Results Feasibility 1 Lyophilization of 1-8 Tromethamine and sucrose containing assessment starting matrix formulations formulations did not have sufficient (feasibility); 1 mL fill stability. volume, 5 mL vials 2 Scale of product unit 1, 3, 7 .sub.i) Scale-up prevented completion of drying size (5 mL fill, 20 mL during the prim,. drying phase. vials) .sub.ii) Less foaming (reconstitution) observed for # 3. 3 .sub.i) Cycle parameter 1, 3, 7 Products obtained w/2% volatiles. A adjustment. reconstitution analysis was not performed .sub.ii) Obtain products w/ for this lyophilization cycle. acceptable residual moisture. Process 4 --- Cycle fail --- Development 5 .sub.i) Investigate the extent 1, 3, 3a, .sub.i) Comparable foaming for #3, #3a. of foaming 3b, 3c .sub.ii) No loss of brincidofovir was observed .sub.ii) Evaluate the within 48 h after reconstitution (all reconstitution process formulations) incl. 48 h reconst. stability Short-term 6 Evaluate the stability of 1, 3, 3a (i) Increased residual moisture for #1 and stability the lyophilized products therefore stronger degradation. (2 weeks) (solid) (ii) Superior stability of #3a vs. #3. (iii) Loss in brincidofovir recovery observed for #1, #3 pre/post lyophilization (pot. handling errors, high assay variability) DP 7 DP preparation for 3a Preparation of 200 vials (5 mL filling, 20 preparation stability testing mL vial). A loss in recovery (not degradation) observed pre/post lyophilization, (pot. handling errors, high assay variability) Material NA Evaluate the 3a The formulation (3a) was compatible with compatibility compatibility of sterile all tested materials. An initial assessment filters, vials, stoppers, adsorption/saturation effect was observed infusion bags, IV for the IV systems at low c(BCV). systems. Samples at c(BCV) = 0.1 mg/mL showed a more significant pH shift.

TABLE-US-00029 TABLE 29 Lyophilization cycle parameters (Cycles #2, #3, #5, #6) Cycle #2 Cycle #3 Cycle #5 Cycle #6 Ramp Total Ramp Total Ramp Total Ramp Total rate T.sub.shelf Time Time rate T.sub.shelf Time Time rate T.sub.shelf Time Time rate T.sub.shelf Time Time Step Type [.degree. C./h] [.degree. C.] [min] [h] [.degree. C./h] [.degree. C.] [min] [h] [.degree. C./h] [.degree. C.] [min] [h] [.degree. C./h] [.degree. C.] [min] [h] Loading Hold N/A 5 0 0 N/A 5 0 0 N/A 5 0 0 N/A 5 0 0 Freeze/ Hold 0 5 60 1 0 5 60 1 0 5 60 1 0 5 60 1 Anneal Freeze/ Rate 20 -50 165 3.8 20 -50 165 3.8 20 -50 165 3.8 20 -50 165 3.8 Anneal Freeze/ Hold N/A -50 180 6.8 N/A -50 180 6.8 N/A -50 180 6.8 N/A -50 180 6.8 Anneal Freeze/ Rate 20 -4 174 9.7 20 -4 174 9.7 20 -4 174 9.7 20 -4 174 9.7 Anneal Freeze/ Hold N/A -4 240 13.7 N/A -4 240 13.7 N/A -4 240 13.7 N/A -4 240 13.7 Anneal Freeze/ Rate 20 -38 81 15 20 -35 81 15 20 -35 81 15 20 -35 81 15 Anneal Freeze/ Hold N/A -38 60 16 N/A -35 60 16 N/A -35 180 18 N/A -35 180 18 Anneal Primary Hold N/A -38 1200 36 N/A -35 2160 52 N/A -35 2160 54 N/A -35 2160 54 Drying Secondary Rate 2.5 20 1300 57.7 30 30 130 54.2 30 30 130 56.2 30 30 390 60.5 drying Secondary Hold N/A 20 360 63.7 N/A 30 360 60.2 N/A 30 360 62.2 N/A 30 360 66.5 drying

i) Feasibility Assessment: Compounding and Reconstitution

[0369] To further evaluate the formulations upon reconstitution following lyophilization according to the different lyophilization cycles, liquid fill solutions were prepared and evaluated as outlined below. For lyophilization cycles #1, #2, and #5, liquid fill solutions were prepared comprising: [0370] a) Brincidofovir stock solution at c(BCV)=10 mg/mL (pH adjustment to pH=8.0), [0371] b) 10.times. buffer stock solutions or DI water (pH adjustment to pH=8.0), and [0372] c) 12.5% (w/v) mannitol stock solution (pH adjustment to pH=8.0) [0373] d) DI water.

[0374] For cycles #6 and #7, liquid fill solutions were prepared comprising: [0375] a) brincidofovir (solid), [0376] b) mannitol (solid), [0377] c) 10.times. arginine stock solution (pH adjustment to pH=8.0), [0378] d) NaOH-solution (1M), and [0379] e) DI water.

[0380] For cycles #1, #2, and #5 the applied reconstitution volumes were determined by gravimetric analysis of the product vials before and after lyophilization for each of the formulations. In case of cycles #1 and #2 the specific measured volumes were applied. For reconstitution of lyophilized products of cycle #5 an average reconstitution volume was applied to all formulations. For the lyophilized product of lyophilization cycle #7, the liquid fill volume was scaled in accordance to the previously determined difference between liquid fill and reconstitution volume.

[0381] After each lyophilization cycle the lyophilized products were evaluated for their cake structure, uniformity and defects in appearance. Following reconstitution with DI water, the appearance of the solution during and after reconstitution (e.g. foaming intensity, foam dissipation time), pH, and recovery of brincidofovir were evaluated. The recovery of brincidofovir was determined by HPLC analysis. The fraction of volatiles was determined by thermogravimetric analysis (TGA). The results of the reconstitution assessments are summarized in Table 30.

TABLE-US-00030 TABLE 30 Assessment of the drug product and reconstitution process. Average Fraction Average Foam. Pre lyo Post lyo Purity Cycle Form. Volatiles Reconst. dissip. time* c(BCV) c(BCV) c(BCV) # # [%] Average Appear. Time [s] [min] pH [mg/mL] [mg/mL] [%] 1 1 N/A Minor melt-back 50 7 7.8 N/A 10.43 N/A 2 N/A Minor melt-back 80 45 7.8 N/A 10.53 N/A 3 N/A Minor melt-back 60 <1 7.9 N/A 10.33 N/A 4 N/A Cake collapse 45 35 7.9 N/A 9.83 N/A 5 N/A Cake collapse 90 <1 8.1 N/A 10.63 N/A 6 N/A Cake collapse 15 45 8.0 N/A 10.43 N/A 7 N/A Porous, uniform 45 3 8.4 N/A 10.63 N/A 8 N/A Cake collapse 60 60 8.3 N/A 10.33 N/A 2 1 0.3 Little cracks 100 32 8.0 9.8 9.6 N/A 3 1.5 Porous, uniform 45 8 8.0 9.9 9.6 N/A 7 3.8 Minor melt-back 105 30 8.4 9.5 9.5 N/A 3 1 0.3 Little cracks N/A N/A N/A N/A N/A N/A 3 2.0 Porous, uniform N/A N/A N/A N/A N/A N/A 7 0.8 Minor melt-back N/A N/A N/A N/A N/A N/A 5 1 N/A Minor cracks 45 >30 (minor 7.9 N/A 10.4 N/A foaming) 3 N/A Porous, uniform 45 <20 8.0 N/A 10.2 N/A 3a N/A Porous, uniform 130 <20 7.9 N/A 10.1 N/A 3b N/A Porous, uniform 95 >30 (minor 7.9 N/A 10.0 N/A foaming) 3c N/A Porous, uniform 80 >30 (minor 7.9 N/A 10.0 N/A foaming) 7 N/A Minor melt-back 15 >30 8.2 N/A 10.2 N/A 6 1 4.2 Several cracks 40 >30 (minor 7.9 10.3 9.8 N/A foaming) 3 1.0 Porous, uniform 30 <20 7.9 10.7 9.5 99.7 3a 0.3 Porous, uniform 91 >30 (minor 7.9 10.5 9.6 99.6 foaming) 7 3a 0.5 Porous, uniform 83 <20 8.0 10.7 9.9 N/A *Average value of three vials. For cycle #1-2 the approximate dissipation time was recorded (constant observation). For cycle #5-7 the foaming intensity was evaluated at 10, 20 and 30 min after reconstitution. <20 indicates that the foaming dissipation time was between the 10 and 20 min interval. <30 indicates that the foaming dissipation time was between the 20 and 30 min interval. If foaming still persisted over 30 min, the dissipation time was indicated by >30 min.

ii) Process Development: Stability Studies of Lyophilized Formulations--48 h Study

[0382] To evaluate the physical and chemical stability of the reconstituted solutions the appearance and pH of each of Formulations 1, 3, 3a, 3b, 3c, and 7, as well as recovery of c(BCV) at the time of reconstitution (t.sub.0), after 1 day (t.sub.24h) and after 2 days 4480 was assessed at ambient temperature. The recovery of c(BCV) was determined via HPLC.

[0383] All tested formulations (Formulations #1, 3, 3a, 3b, 3c, and 7), were clear and colorless in appearance and yielded complete recovery of brincidofovir within 48 h after reconstitution (i.e., no significant loss of brincidofovir, variations in recovery are within the error range). Formulations 3 and 3a were found to display faster dissipation of foam/bubbles than Formulations 3b, and 3c (Table 33).

iii) Short-Term Stability Study: Stability Studies of Lyophilized Formulations--2 Week Study

[0384] The stability of Formulations 1, 3, and 3a was assessed at 25.degree. C. and 60.degree. C. by evaluating appearance and pH of each formulation, as well as recovery of c(BCV), at the time of reconstitution (t.sub.0), after 2 days (t.sub.2 days), 7 days (t.sub.7 days), 10 days (t.sub.10 days), and 14 days (t.sub.14 days). In addition, the foaming intensity was evaluated at 10, 20 and 30 minutes after reconstitution.

[0385] The recovery of brincidofovir was determined via HPLC. Degradants observed in the HPLC chromatograms were identified as the deamination product, likely Cidofovir, as well as additional degradants No shift or changes in pH were associated with degradation. Of the tested formulations, Formulation 3a showed the least degree of degradation under the applied conditions with a purity of 99.2% after 14 days at 60.degree. C. The results of the experiments are summarized in Table 31, Table 32, and Table 36.

TABLE-US-00031 TABLE 31 Stability study results of the formulation 1 (200 mM Phosphate) Conditions Station Average Average Average Observed BCV BCV temp. Cake Average Recon. Foam. Time c(BCV).sup.a Reovery .sup.b Purity (.degree. C.) Day appearance Recon. Appear. Time [s] [min]* pH (mg/mL) [%] [%] Pre lyo N/A N/A N/A N/A 7.9 10.3 0.4 N/A Pre lyo 25 0** Several cracks Clear, colorless 40 >30 (minor 7.9 9.8 100 N/A foaming) 2 Several cracks Clear, colorless 41 >30 (minor 8.0 9.9 101 N/A foaming) 7 Several cracks Clear, colorless 33 >30 (minor 8.0 10.0 102 N/A foaming) 10 N/A N/A N/A N/A N/A N/A N/A N/A 14 N/A N/A N/A N/A N/A N/A N/A N/A 60 0** Several cracks Clear, colorless 40 >30 (minor 7.9 9.8 100 N/A foaming) 2 Major collapse Clear, colorless 30 >30 (minor 8.0 9.8 100 N/A foaming) 7 Major collapse Clear, colorless 32 >30 (minor 8.0 9.6 98 N/A foaming) 10 N/A N/A N/A N/A N/A N/A N/A N/A 14 N/A N/A N/A N/A N/A N/A N/A N/A *Average value of three vials. The foaming intensity was evaluated at 10, 20 and 30 min after reconstitution. <20 indicates that the foaming dissipation time was between the 10 and 20 min interval. <30 indicates that the foaming dissipation time was between the 20 and 30 min interval. If foaming still persisted over 30 min, the dissipation time was indicated by >30 min. .sup.aAverage value of the three replicates of samples that received treatment. .sup.b BCV recovery = c(BCV).sub.Day n/c(BCV).sub.Day 0 .times. 100%. **The sample for t.sub.0 at T = 25.degree. C. and t.sub.0 at T = 60.degree. C. is identical. Note: HPLC sample variability accounts for the assay variability between samples. Vial to vial variability accounts for the variability between drug product vials.

TABLE-US-00032 TABLE 32 Stability study results of the formulation 3 (200 mM Arginine) Conditions Station Average Average Average Observed BCV BCV temp. Cake Average Recon. Foam. Time c(BCV).sup.a Recovery.sup.b Purity (.degree. C.) Day appear. Recon. Appear. Time [s] [min]* pH [mg/mL] [%] [%] Pre lyo N/A N/A N/A N/A 8.0 10.7 N/A N/A 25 0** Uniform Clear, colorless 30 10 7.9 9.5 100 99.7 2 Uniform Clear, colorless 24 10 7.9 9.6 101 99.6 7 Uniform Clear, colorless 24 <10 7.9 9.7 102 99.7 10 Uniform Clear, colorless 18 <10 8.0 9.9 104 99.8 14 Uniform Clear, colorless 16 <10 8.0 9.8 103 99.7 60 0** Uniform Clear, colorless 30 10 7.9 9.5 100 99.7 2 Uniform Clear, colorless 34 10 7.9 9.5 100 99.6 7 Uniform Clear, colorless 20 10 7.9 9.6 101 99.0 10 Uniform Clear, colorless 18 <30 (minor 7.9 9.7 102 98.9 foaming) 14 Uniform Yellow tint 20** <20 7.9 9.6 101 95.9 *Average value of three vials. The foaming intensity was evaluated at 10, 20 and 30 min after reconstitution. <20 indicates that the foaming dissipation time was between the 10 and 20 min interval. <30 indicates that the foaming dissipation time was between the 20 and 30 min interval. If foaming still persisted over 30 min, the dissipation time was indicated by >30 min. .sup.aAverage value of the three replicates of samples that received treatment. .sup.bBCV recovery = c(BCV).sub.Day n/c(BCV).sub.Day 0 .times. 100%. **The sample for t.sub.0 at T = 25.degree. C. and t.sub.0 at T = 60.degree. C. is identical. Note: HPLC sample variability accounts for the assay variability between samples. Vial to vial variability accounts for the variability between drug product vials.

TABLE-US-00033 TABLE 33 Stability study results of the formulation #3a (100 mM Arginine) Conditions Station Average Average Average Observed BCV BCV temp. Cake Average Recon. Foam. Time c(BCV).sup.a Recovery.sup.b Purity (.degree. C.) Day appear Recon. Appear. Time [s] [min]* pH (mg/mL) (%) (%) Pre lyo N/A N/A N/A N/A 7.9 10.5 N/A N/A 25 0** Uniform Clear, colorless 91 >30 (minor 7.9 9.6 100 99.6 foaming) 2 Uniform Clear, colorless N/A N/A N/A N/A N/A N/A 7 Uniform Clear, colorless N/A N/A N/A N/A N/A N/A 10 Uniform Clear, colorless 80 >30 (minor 8.0 10.0 104 99.7 foaming) 14 Uniform Clear, colorless 65 >30 (minor 8.0 9.9 103 99.7 foaming) 60 0** Uniform Clear, colorless 91 >30 (minor 7.9 9.6 100 99.6 foaming) 2 Uniform Clear, colorless N/A N/A N/A N/A N/A N/A 7 Uniform Clear, colorless N/A N/A N/A N/A N/A N/A 10 Uniform Clear, colorless 49 >30 (minor 7.9 9.8 102 99.8 foaming) 14 Uniform Clear, colorless 26 >30 (minor 8.0 9.8 102 99.2 foaming) .sup.aAverage value of the three replicates of samples that received treatment. .sup.bBCV recovery = c(BCV).sub.Day n /c(BCV).sub.Day 0 .times. 100%. **The sample for t.sub.0 at T = 25.degree. C. and t.sub.0 at T = 60.degree. C. is identical. Note: HPLC sample variability accounts for the assay variability between samples. Vial to vial variability accounts for the variability between drug product vials

iv) Preparation of Drug Product Batches for Stability Testing

[0386] Preparation of Pre-Lyophilization Formulations: Formulation 3a was compounded according to the following procedure: The pH of a 1M buffer solution of arginine in DI water was adjusted to 8 using Hydrochloric acid (35-37%) under pH-control before Q.S. Then Mannitol was added to the buffer solution followed by BCV. The pH of the resulting solution was adjusted to 8 using sodium hydroxide. DI water was used to Q.S. the solution in separate volumetric flasks. The two solutions were then mixed and sterile filtered to yield the desired Formulation 3a. The procedure yielded Formulation 3a with an observed c(BCV) of 9.9 mg/mL.

[0387] Preparation of Lyophilized Formulations:

[0388] Under best clean conditions approximately 200 vials of liquid Formulation 3a, made according to the procedure outlined above, were vials were stoppered in lyophilization position and subjected to lyophilization according to lyophilization cycle #7. The resulting lyophilized product was tested for appearance, reconstitution appearance, pH, and recovery of brincidofovir. The recovery of brincidofovir was determined via HPLC, and thermogravimetric analysis. The results of the testing of appearance, reconstitution appearance and recovery of brincidofovir in the lyophilized formulation prepared according to the procedure outlined herein are summarized in Table 34.

TABLE-US-00034 TABLE 34 Appearance, reconstitution and recovery of formulations batch pre and post Lyophilization. Foam. dissip. Average Fraction Reconst. time c(BCV).sup.b Condition Volatiles Appear. time [s] [min]* pH [mg/mL] Pre lyo. N/A N/A N/A N/A 8.0 10.7 Release 0.5 Porous, 83 <20 8.0 9.9 *Average value of three vials. The foaming intensity was evaluated at 10, 20 and 30 min after reconstitution. <20 indicates that the foaming dissipation time was between the 10 and 20 min interval. <30 indicates that the foaming dissipation time was between the 20 and 30 min interval. If foaming still persisted over 30 min, the dissipation time was indicated by >30 min. .sup.aScan range = 25.degree. C. to 150.degree. C., ramp rate = 10.degree. C./min. .sup.bAverage value of the three (3) replicates of samples that received treatment. indicates data missing or illegible when filed

v) Compatibility of Lyophilized Formulations

[0389] The liquid fill solution for the lyophilized Formulation 3a was tested for compatibility with infusion vehicles as well as a series of materials from the manufacturing and clinical stage. The results of the infusion vehicle compatibility testing are summarized in Table 38. No precipitation, pH shift or significant loss in recovery of brincidofovir were observed at the end of the study.

Infusion Vehicle Compatibility

[0390] The Formulation 3a was diluted with the infusion vehicle (5% dextrose) solution to yield c(BCV)=0.5 mg/mL and 1.0 mg/mL solutions. The appearance, pH, and brincidofovir recovery (determined via HPLC) were evaluated at the time of preparing the test solution (t.sub.0), and after 2 h (t.sub.2h), 8 h (t.sub.8h), and 24 h (t.sub.24h)

TABLE-US-00035 TABLE 35 Results of the infusion vehicle compatibility assessment. c(BCV) in infusion Time Observed BCV vehicle point Measured c(BCV).sup.a Recovery.sup.a,b (mg/mL) (h) Appearance pH (mg/mL) (%) 0.5 t.sub.0 Clear, 7.9 0.492 100 Colorless t.sub.2 h Clear, 7.9 0.493 100 Colorless t.sub.8 h Clear, 7.7 0.492 100 Colorless t.sub.24 h Clear, 7.7 0.492 100 Colorless 1.0 t.sub.0 Clear, 7.9 0.967 100 Colorless t.sub.2 h Clear, 7.8 0.966 100 Colorless t.sub.8 h Clear, 7.8 0.969 100 Colorless t.sub.24 h Clear, 7.7 0.967 100 Colorless .sup.aAverage value of the three replicates. .sup.bBCV recovery = c(BCV).sub.Hour n/c(BCV).sub.Hour 0 .times. 100%. The value for BCV recovery at t.sub.0 was defined as 100%.

Sterilization Filter Compatibility

[0391] The liquid Formulation 3a was tested for compatibility with two syringe filters for aseptic processing. 10 mL of the liquid Formulation 3a, were filtered through one of the filters a (0.2 .mu.m, 25 mm, syringe filter, Posidyne.RTM. membrane) orb (0.2 .mu.m, 25 mm, syringe filter, Supor.RTM. membrane (PES)), and the first and last 10% filtrate were evaluated for appearance, pH and recovery of brincidofovir.

[0392] The results of the evaluation are summarized in Table 36. No precipitation, pH shift or significant loss in brincidofovir recovery were observed. Samples appeared clear and colorless. Degradant peaks were not observed.

TABLE-US-00036 TABLE 36 Results of the sterilization filter compatibility assessment of formulation 3a. Observed Filter Measured c(BCV).sup.a BCV membrane Conditions Appearance pH (mg/mL) Recovery.sup.a,b Posidyne .RTM. Pre filtration Clear, 7.9 10.25 100 Colorless First 10% filtrate Clear, 7.9 10.16 99 volume Colorless Last 10% filtrate Clear, 7.9 10.12 99 volume Colorless PES .RTM. Pre filtration Clear, 7.9 10.25 100 Colorless First 10% filtrate Clear, 7.9 10.21 100 volume Colorless Last 10% filtrate Clear, 7.9 10.15 99 volume Colorless .sup.aAverage value of the three replicates. .sup.bBCV recovery = c(BCV).sub.Hour n/c(BCV).sub.Pre filling .times. 100%. The value for BCV recovery at t.sub.0 was defined as 100%.

Drug Product Vial and Stopper Compatibility

[0393] The liquid Formulation 3a was tested for compatibility with vials and stoppers. The vials were filled with 5 mL of the liquid Formulation 3a, stoppered, crimped, and stored for 6 hours at ambient conditions in upright and inverted orientations. The vials used for testing were clear, 20 mL, Class A borosilicate glass serum vials, and the stoppers were 20 mm Novapure.RTM. stoppers, with FluroTec.RTM. coating.

[0394] The results of the study are summarized in Table 37. No precipitation, pH shift or significant loss in brincidofovir recovery was observed throughout the study. No degradant peaks were observed in HPLC.

TABLE-US-00037 TABLE 37 Results of the drug product vial and stopper compatibility assessment. Observed BCV Measured c(BCV).sup.a Recovery.sup.a,b Conditions Appearance pH (mg/mL) (%) Pre filling Clear, 7.9 10.25 100 Colorless Upright, 6 h Clear, 7.9 10.15 99 Colorless Inverted, 6 h Clear, 7.9 10.20 100 colorless .sup.aAverage value of the three replicates. .sup.bBCV recovery = c(BCV).sub.Hour n/c(BCV).sub.Pre filling .times. 100%. The value for BCV recovery at t.sub.0 was defined as 100%.

Infusion Bag Compatibility

[0395] The liquid Formulation 3a was tested for compatibility with a 100 mL infusion bag. Solutions of Formulation 3a, in a 5% dextrose injection vehicle, were prepared at two different formulation strengths (1.0 mg/mL and 0.1 mg/mL brincidofovir), and stored in the infusion bag at ambient conditions. Samples form the infusion bags and from an externally diluted standard were evaluated for appearance, pH and brincidofovir recovery (determined via HPLC) at the beginning of the study (t.sub.0), after 8 h (t.sub.8h), and after 24 h (t.sub.24h). The results of the infusion bag compatibility assessment are summarized in Table 38.

TABLE-US-00038 TABLE 38 Results of the infusion bag compatibility assessment c(BCV) Bag BCV in testing repli- Mea- Observed Recov- infusion cate Condi- Appear- sured c(BCV).sup.a ery.sup.a,b bag # tions ance pH (mg/mL) (%) 0.1 1 External Clear, 7.2 0.0909 101 dilution Colorless t.sub.0 Clear, 7.3 0.0896 100 Colorless t.sub.1 h Clear, 7.1 0.0893 100 Colorless t.sub.8 h Clear, 7.3 0.0909 101 Colorless t.sub.24 h Clear, 7 0.0906 101 Colorless 2 t.sub.0 N/A N/A 0.0886 100 t.sub.24 h Clear, 7 0.0883 100 Colorless 3 t.sub.0 N/A. N/A 0.111 100 t.sub.24 h Clear, 7 0.0891 81 Colorless 1 1 External Clear, 7.9 0.927 101 dilution Colorless t.sub.0 Clear, 7.9 0.915 100 Colorless t.sub.1 h Clear, 7.7 0.908 99 Colorless t.sub.8 h Clear, 7.8 0.914 100 Colorless t.sub.24 h Clear, 7.7 0.915 100 Colorless 2 t.sub.0 N/A N/A 1.108 100 t.sub.24 h Clear, 7.7 1.106 100 Colorless 3 t.sub.0 N/A. N/A 0.903 100 t.sub.24 h Clear, 7.7 0.903 100 Colorless .sup.aAverage value of the three replicates. .sup.bBCV recovery = c(BCV).sub.Hour n/c(BCV).sub.Hour 0 .times. 100%. The value for BCV recovery at t.sub.0 was defined as 100%.

[0396] The samples appeared clear and colorless. For both sample concentrations, no significant loss in brincidofovir recovery was observed over 24 h. For one bag replicate at c(BCV)=0.1 mg/mL an apparent loss of brincidofovir (19%) is observed; It was further observed that a dilution of the brincidofovir formulation into the infusion vehicle, yielding a concentration of c(BCV)=0.1 mg/mL caused a significant pH shift to pH=7. No degradation was observed in the experiment.

Infusion System Compatibility

[0397] The brincidofovir formulation (10 mg/mL brincidofovir, 100 mM arginine, 5% mannitol, pH=8.0) was tested for compatibility with two infusion systems (Baxter, Non-DEHP CONTINU-FLO; B Braun, Rate Flow.RTM. regulator IV set). The liquid formulation was diluted into infusion bags as previously described (tested final concentrations: 1.0 mg/mL and 0.1 mg/mL) and an aliquot was collected from the infusion bag for analysis of brincidofovir recovery after 24 h (t0, bag). The infusion systems were then attached to the infusion bag and manually filled with solution from the infusion bag. Aliquots were collected as an end-of-line sample for analysis (t0, eol). The IV lines were closed between time points with the infusate resting within the infusion system and stored at ambient temperature. At testing time points, approximately 17 mL (approx. volume of the IV system) of infusate solution were drained, which led to a replacement of the solution in the IV system from the bag reservoir. Following this replacement, another end-of-line sample was collected for the assessments (t10 min, eol, t3 h, eol, and t6 h, eol). The samples were evaluated for appearance, pH, and brincidofovir recovery. The results of infusion system compatibility testing are summarized in Table 42.

[0398] An initial adsorption/saturation effect was observed for both IV systems at low c(BCV), indicated by a loss in brincidofovir recovery of 7-11%. This loss partially recovered over the following 6 h. The appearance of degradants was not observed over the course of this study. As previously noted, samples at c(BCV)=0.1 mg/mL showed a pH-shift by approximately one pH-unit to pH=7 upon dilution with 5% dextrose solution. The samples appeared clear and colorless over the course of the study.

TABLE-US-00039 TABLE 39 Results of the infusion system compatibility assessment. c(BCV) in BCV Testing infusion Observed Recov- infusion bag Appear- c(BCV).sup.a ery.sup.a,b system (mg/mL) Conditions ance pH (mg/mL) (%) Baxter 0.1 t.sub.0, bag Clear, 7 0.0883 100 Colorless t.sub.0, eol Clear, 6.9 0.0826 93 Colorless t.sub.10 min, eol Clear, 6.9 0.0868 98 Colorless t.sub.3 h, eol Clear, 7 0.0876 99 Colorless t.sub.6 h, eol Clear, 7 0.0863 98 Colorless 1 t.sub.0, bag Clear, 7.7 1.106 100 Colorless t.sub.0, eol Clear, 7.7 1.095* 99 Colorless t.sub.10 min, eol Clear, 7.7 1.121 101 Colorless t.sub.3 h, eol Clear, 7.7 1.136 103 Colorless t.sub.6 h, eol Clear, 7.7 1.107 100 Colorless B Braun 0.1 t.sub.0, bag Clear, 7 0.0891 100 Colorless t.sub.0, eol Clear, 6.9 0.0796 89 Colorless t.sub.10 min, eol Clear, 6.9 0.0854 96 Colorless t.sub.3 h, eol Clear, 7 0.0849 95 Colorless t.sub.6 h, eol Clear, 6.9 0.0844 95 Colorless 1 t.sub.0, bag Clear, 7.7 0.903 100 Colorless t.sub.0, eol Clear, 7.7 0.9 100 Colorless t.sub.10 min, eol Clear, 7.6 0.908 101 Colorless t.sub.3 h, eol Clear, 7.7 0.909 101 Colorless t.sub.6 h, eol Clear, 7.7 0.909 101 Colorless .sup.aAverage value of the three replicates. .sup.bBCV recovery = c(BCV).sub.Hour n/c(BCV).sub.Hour 0 .times. 100%. The value for BCV recovery at t.sub.0 was defined as 100%.

Example 6--Stability Testing

[0399] The bulk liquid formulations and lyophilized formulations of the disclosure were further tested for stability. A liquid formulation comprising arginine, mannitol, and brincidofovir at a concentration of 10 mg/mL was kept in a 10 mL glass vial, stoppered (rubber stopper or 20 mm serum stopper) and sealed (aluminum seals), and kept at about 5.degree. C. or about 25.degree. C. for up to 12 months. The appearance, pH and brincidofovir content of the stored formulation were monitored along with impurities and particulate matter, at various time intervals (i.e., after 2 weeks, after 1 month, after 2 months, after 3 months, after 6 months, after 9 months and after 12 months). Impurities were evaluated via HPLC and are identified herein by their relative retention time. The same experiment was repeated for a solution of the lyophilized powder, comprising 10 mg/mL of brincidofovir. For the lyophilized formulations, a 20 mL glass vial with a 20 mm rubber stopper was used. Furthermore, the appearance of the lyophilized powder (i.e., the lyophilization cake) was recorded.

[0400] The results of the stability study of the bulk liquid formulations at 5.degree. C. and 25.degree. C. are summarized in Tables 40A and 40B, respectively. The results of the stability study of the lyophilized formulations at 5.degree. C. and 25.degree. C. are summarized in Tables 41A, 41B, and 42, respectively.

TABLE-US-00040 TABLE 40A Stability of liquid formulation at 5.degree. C. (.+-.3.degree. C.) Specifications Acceptance Stability Intervals Test Criteria Initial 2 week 1 month 2 month 3 month 6 month 9 month 12 month Appearance Clear, colorless; Conforms Conforms Conforms Conforms Conforms Conforms Does not Conforms free of visible conform particulates Brincidofovir 90.0 to 110.0% 100.3 100.7 100.4 100.0 101.2 101.3 99.9 99.8 Content - Average Drug Related Report Results <0.05 <0.05 <0.05 0.05 0.06 0.07 0.12 0.15 Impurities - RRT 0.80 Drug Related Report Results <0.05 <0.05 <0.05 <0.05 <0.05 <0.05 <0.05 <0.05 Impurities - RRT 1.13 Drug Related Report Results <0.05 <0.05 <0.05 <0.05 <0.05 <0.05 <0.05 <0.05 Impurities - RRT 1.19 Drug Related Report Results <0.05 <0.05 <0.05 <0.05 <0.05 <0.05 <0.05 <0.05 Impurities - RRT 1.34 Drug Related Report Results <0.05 <0.05 <0.05 <0.05 <0.05 <0.05 <0.05 <0.05 Impurities - RRT 1.42 Drug Related Report Results <0.05 <0.05 <0.05 0.05 0.06 0.07 0.12 0.15 Impurities - Total Impurities pH - Average 7.5 to 8.5 8.0 8.0 7.9 7.8 8.0 8.0 7.8 7.9 Particulate .ltoreq.6000 148 N/T 274 N/T 181 135 N/T N/T Matter - 10 .mu.m Particulate .ltoreq.600 1 N/T 1 N/T 2 2 N/T N/T Matter - 25 .mu.m

TABLE-US-00041 TABLE 40B Stability of liquid formulation at 25.degree. C. (.+-.2.degree. C.) and 60% relative humidity (.+-.5% relative humidity) Specifications Acceptance Stability Intervals Test Criteria Initial 2 week 1 month 2 month 3 month 6 month 12 month Appearance Clear, colorless; Conforms Conforms Conforms Conforms Conforms Conforms Conforms free of visible particulates Brincidofovir 90.0 to 110.0% 100.3 100.9 100.1 99.4 98.9 99.9 97.5 Content - Average Drug Related Report Results <0.05 0.10 0.21 0.37 0.54 1.0 2.0 Impurities - RRT 0.80 Drug Related Report Results <0.05 <0.05 0.06 0.07 0.12 0.29 0.60 Impurities - RRT 1.13 Drug Related Report Results <0.05 <0.05 0.05 <0.05 <0.05 <0.05 <0.05 Impurities - RRT 1.19 Drug Related Report Results <0.05 <0.05 <0.05 <0.05 0.05 <0.05 <0.05 Impurities - RRT 1.34 Drug Related Report Results <0.05 <0.05 <0.05 <0.05 <0.05 <0.05 0.05 Impurities - RRT 1.42 Drug Related Report Results <0.05 0.10 0.32 0.45 0.71 1.3 2.7 Impurities - Total Impurities pH - Average 7.5 to 8.5 8.0 8.0 7.9 7.9 8.0 8.0 7.9 Particulate .ltoreq.6000 148 N/T N/T N/T N/T 325 N/T Matter- 10 .mu.m Particulate .ltoreq.600 1 N/T N/T N/T N/T 7 N/T Matter - 25 .mu.m

TABLE-US-00042 TABLE 41A Stability of lyophilized formulation at 5.degree. C. (.+-.3.degree. C.) Specifications Acceptance Stability Intervals Test Criteria Initial 2 week 1 month 2 month 3 month 6 month 9 month 12 month Appearance of Report White White White White White White White White Lyophilized Results porous porous uniform uniform porous cake in porous porous Cake-Appearance uniform uniform cake with cake with uniform the uniform uniform structure structure some deposited structure bottom of structure structure with some with some deposited material with some the bottle with some with some solid solid material above on solid solid solid deposits on deposits above the wall. deposits deposits deposits vial above above the One cake above the above the above the the cake cake on at -20.degree. C., cake in cake in cake on the vials. both the vial. the vial. the vial. The vials cakes at 5.degree. had some C., and cake one cake particulates at 40.degree. C., are on the removable stoppers from the of the bottom of vials. the vial. Completeness Meets the N/T N/T N/T Conforms Not tested Conforms Conforms Conforms and Clarity of requirements Solution of USP <1> Brincidofovir 90.0 to 110.0% 100.3 100.4 98.6 98.7 98.2 100.1 98.4 98.2 Content - Average Drug Related Report None None None None None None None None Impurities - Results Detected Detected Detected Detected Detected Detected Detected Detected Synergi imp Drug Related Report None None None None None None None None Impurities - Results Detected Detected Detected Detected Detected Detected Detected Detected XBridge imp Drug Related Report None None None None None None None None Impurities - Total Results Detected Detected Detected Detected Detected Detected Detected Detected Impurities = XBridge + Synergi pH 7.5 to 8.5 7.9 7.9 7.9 7.9 7.9 7.8 7.9 7.9 Reconstitution Report 90 N/T N/T 136 129 105 152 160 Time - Prep 1 Results Reconstitution Report 95 N/T N/T 160 90 125 148 130 Time - Prep 2 Results Particulate .ltoreq.6000 188 N/T 52 N/T N/T 84 N/T N/T Matter - 10 .mu.m Particulate .ltoreq.600 4 N/T 0 N/T N/T 3 N/T N/T Matter - 25 .mu.m Moisture Report N/T N/T N/T 0.3 0.8 0.5 N/T 0.5 Average Results

TABLE-US-00043 TABLE 41B Stability of lyophilized formulation at 25.degree. C. (.+-.2.degree. C.) and 60% relative humidity (.+-.5% relative humidity) Specifications Acceptance Stability Intervals Test Criteria Initial 2 week 1 month 3 month 6 month 9 month 12 month Appearance of Report White White White White White cake White White Lyophilized Results porous porous uniform porous in the porous porous Cake-Appearance uniform uniform cake with uniform bottom of uniform uniform structure structure some structure the bottle structure structure with some with some deposited with some with some with some solid solid material solid solid solid deposits on deposits above deposits deposits deposits vial above above the above the above the above the the cake cake on the cake on cake on cake on vials. The the vial. the vial. the vial. vials had some cake particulates on the stoppers of the vials. Completeness Meets the N/T N/T N/T Not tested Conforms Conforms Conforms and Clarity of requirements Solution of USP <1> Brincidofovir 90.0 to 110.0% 100.3 98.0 98.5 98.4 101.2 98.2 98.5 Content - Average Drug Related Report None None None None None None None Impurities - Results Detected Detected Detected Detected Detected Detected Detected Synergi imp Drug Related Report None None None None None None None Impurities - Results Detected Detected Detected Detected Detected Detected Detected XBridge imp Drug Related Report None None None None None None None Impurities - Total Results Detected Detected Detected Detected Detected Detected Detected Impurities = XBridge + Synergi pH 7.5 to 8.5 7.9 7.9 7.9 7.9 7.8 7.9 7.9 Reconstitution Report 90 N/T N/T 92 111 147 177 Time - Prep 1 Results Reconstitution Report 95 N/T N/T 91 103 135 189 Time - Prep 2 Results Particulate .ltoreq.6000 188 N/T N/T N/T 83 N/T N/T Matter-10 .mu.m Particulate .ltoreq.600 4 N/T N/T N/T 2 N/T N/T Matter-25 .mu.m Moisture Report N/T N/T N/T 0.6 0.5 0.6 0.6 Average Results

TABLE-US-00044 TABLE 42 Stability of lyophilized formulation at 40.degree. C. (.+-.2.degree. C.) and 60% relative humidity (.+-.5% relative humidity) Specifications Acceptance Stability Intervals Test Criteria Initial 2 week 1 month 2 month 3 month 6 month Appearance of Report White White White White White White cake in Lyophilized Results porous porous uniform uniform porous the bottom of Cake-Appearance uniform uniform cake with cake with uniform the bottle structure structure some deposited structure with some with some deposited material with some solid solid material above on solid deposits deposits above the wall. deposits on vial above the One cake above the above the cake on at -20.degree. C., cake in cake the vials. both the vials. The vials cakes at 5.degree. had some C., and cake one cake particulates at 40.degree. C. are on the removable stoppers of from the the vials. bottom of the vial. Completeness Meets the Not tested Not tested Not tested Conforms Not tested Conforms and Clarity of requirements per protocol per protocol Solution of USP <1> Brincidofovir 90.0 to 110.0% 100.3 97.5 99.1 98.8 98.5 99.6 Content - Average Drug Related Report None None None None None None Impurities - Results detected detected detected detected detected detected Synergi imp Drug Related Report None None None None None None Impurities - Results detected detected detected detected detected detected XBridge imp Drug Related Report None None None None None None Impurities - Total Results detected detected detected detected detected detected Impurities = XBridge + Synergi pH 7.5 to 8.5 7.9 7.9 7.9 7.9 7.9 7.8 Reconstitution Report 90 Not tested Not tested 135 82 99 Time - Prep 1 Results per protocol per protocol Reconstitution Report 95 Not tested Not tested 126 77 106 Time - Prep 2 Results per protocol per protocol Particulate .ltoreq.6000 188 Not tested Not tested Not tested Not tested Not tested Matter - 10 .mu.m per protocol per protocol per protocol per protocol per protocol Particulate .ltoreq.600 4 Not tested Not tested Not tested Not tested Not tested Matter - 25 .mu.m per protocol per protocol per protocol per protocol per protocol Moisture Report Not tested Not tested Not tested 0.6 0.6 Average Results per protocol per protocol per protocol

Example 7--Determination of Hemocompatibility in Rat and Human Whole Blood Introduction

[0401] Each concentration of test item for the rat was mixed separately in a 1:4 ratio with rat whole blood while each concentration of test item for the human was mixed separately in a 1:4 ratio with human whole blood. These samples plus control samples [vehicle control (vehicle+whole blood, 1 for rat and 1 for humans), positive control (1% Saponin+whole blood), untreated whole blood control and negative control (saline+whole blood) were analyzed for hemolysis (supernatant hemoglobin concentration).

[0402] The rat and human whole blood samples were incubated in test or control items at 37.+-.1.degree. C. for 20 minutes and then immediately (within 2 minutes) centrifuged at 3500.+-.100 rpm (2740 g) at room temperature for 5-10 minutes. The supernatant was extracted and subjected to a hemoglobin analysis using the cyanmethemoglobin method employed on the Siemens Advia 120 hematology analyzer. If the test item has a potential for causing hemolysis, then hemoglobin released from lysed red blood cells into the plasma supernatant can be quantified via the reaction of heme iron in solution being oxidized from a ferrous to a ferric state in the presence of potassium cyanide. The reaction results in the formation of methemoglobin, which then combines with cyanide to form a stable cyanmethemoglobin. When measured spectrophotometrically at 546 nm the absorbance of cyanmethemoglobin follows Beer's Law and is directly proportional to the concentration of hemoglobin released into solution. In addition, the formulated test item at each concentration assessed was mixed in a 1:4 ratio with saline and then directly analyzed to ensure that the test item formulations did not interfere with the Siemens Advia measurement of hemoglobin.

Materials and Methods

[0403] The testing was conducted in 2 phases as shown below:

TABLE-US-00045 TABLE 43 Phase 1, Brincidofovir in Rat Blood Rat Test Item Positive Tube Blood* (BCV).sup.a Vehicle.sup.b Control.sup.c Saline.sup.d 1.sup.e + +0.2, 0.5 and - - - 1.6 mg/mL 2.sup.f - +0.2, 0.5 and - - + 1.6 mg/mL 3.sup.g + - + - - 4.sup.h + - - + - 5.sup.i + - - - - 6.sup.j + - - - + *1.0 mL added to Tubes 1 (separate tube for each concentration) and Tubes 3-6 .sup.a250 .mu.L added to Tubes 1-2 (separate tube for each concentration) .sup.b250 .mu.L added to Tube 3 .sup.c250 .mu.L added to Tube 4 .sup.d1.0 mL added to Tube 2; 250 .mu.L added to Tube 6 .sup.eTest item assay for hemolysis: Tube 1A contained 0.2 mg/mL Tube 1B contained 0.5 mg/mL Tube 1C contained 1.6 mg/mL .sup.fTest item interference control: Tube 2A contained 0.2 mg/mL Tube 2B contained 0.5 mg/mL Tube 2C contained 1.6 mg/mL .sup.gVehicle control for hemolysis .sup.hPositive control for hemolysis; 1% Saponin was used as a positive control .sup.iUntreated whole blood control for hemolysis .sup.jNegative control for hemolysis + Added to the tube - Not added to the tube Note: Tubes 3-6 for each phase were run once per phase.

TABLE-US-00046 TABLE 44 Phase 2, Brincidofovir in Human Blood Human Test Item Positive Tube Blood* (BCV).sup.a Vehicle.sup.b Control.sup.c Saline.sup.d 1.sup.e + +0.2, 0.5 and - - - 1.6 mg/mL 2.sup.f - +0.2, 0.5 and - - + 1.6 mg/mL 3.sup.g + - + - - 4.sup.h + - - + - 5.sup.i + - - - - 6.sup.j + - - - + *1.0 mL added to Tubes 1 (separate tube for each concentration) and Tubes 3-6 .sup.a250 .mu.L added to Tubes 1-2 (separate tube for each concentration) .sup.b250 .mu.L added to Tube 3 .sup.c250 .mu.L added to Tube 4 .sup.d1.0 mL added to Tube 2; 250 .mu.L added to Tube 6 .sup.eTest item assay for hemolysis: Tube 1A contained 0.2 mg/mL Tube 1B contained 0.5 mg/mL Tube 1C contained 1.6 mg/mL .sup.fTest item interference control: Tube 2A contained 0.2 mg/mL Tube 2B contained 0.5 mg/mL Tube 2C contained 1.6 mg/mL .sup.gVehicle control for hemolysis .sup.hPositive control for hemolysis; 1% Saponin was used as a positive control .sup.iUntreated whole blood control for hemolysis .sup.jNegative control for hemolysis + Added to the tube - Not added to the tube Note: Tubes 3-6 for each phase were run once per phase.

Control Item (Vehicle)

[0404] The control items for dose formulations was 2 sodium phosphate buffer solution (400 mM, pH=8.0); 1.0N sodium hydroxide solution; sterilized water for injection; and 5% dextrose solution.

Test Preparation and Analysis

[0405] Brincidofovir injectable liquid stock formulation was prepared to 15 mg/mL brincidofovir, compounded from stock solutions inclusive of three (3) steps: (1) 2.times. sodium phosphate buffer solution (400 mM, pH=8.0) was prepared; (2) 1.0 N sodium hydroxide solution was purchased; brincidofovir injectable liquid stock formulation (15 mg/mL brincidofovir, 200 mM sodium phosphate, pH=8.0) from solid brincidofovir, 1.0 N sodium hydroxide solution, 2.times. sodium phosphate buffer solution and WFI (water for injection) was prepared.

[0406] The brincidofovir injectable liquid stock formulation is stable for 28 days under refrigerated conditions (2 to 8.degree. C.). Brincidofovir injectable liquid stock formulation was diluted with 5% dextrose to yield the desired dose concentration.

[0407] Fresh dose formulations were prepared once on the day of testing. Dose formulations were refrigerated (2 to 8.degree. C.) when not in use.

[0408] Saponin was diluted with saline to the appropriate concentration on the day of testing.

Test System

[0409] Whole blood (.about.20 mL) from rats (samples from four animals were pooled for testing) was collected in presence of K.sub.2EDTA anticoagulant and inverted to mix.

[0410] Human whole blood (.about.20 mL) was collected for use in this study from one human volunteer in the presence of K2EDTA anticoagulant.

[0411] The blood samples were held for no more than 4 hours at room temperature following collection, for use in hemolytic potential testing in this study.

Test Methodology

[0412] Each concentration of test item for the rat was mixed separately in a 1:4 ratio with rat whole blood while each concentration of test item for the human was mixed separately in a 1:4 ratio with human whole blood. These samples plus control samples [vehicle control (vehicle+whole blood, 1 for rat and 1 for humans), positive control (1% Saponin+whole blood), untreated whole blood control and negative control (saline+whole blood) were analyzed for hemolysis (supernatant hemoglobin concentration).

[0413] The rat and human whole blood samples were incubated in test or control items at 37.+-.1.degree. C. for 20 minutes and then immediately (within 2 minutes) centrifuged at 3500.+-.100 rpm (2740 g) at room temperature for 5-10 minutes. The supernatant was extracted and subjected to a hemoglobin analysis using the cyanmethemoglobin method employed on the Siemens Advia 120 hematology analyzer. Without wishing to be bound by theory, if the test item has a potential for causing hemolysis, then hemoglobin released from lysed red blood cells into the plasma supernatant can be quantified via the reaction of heme iron in solution being oxidized from a ferrous to a ferric state in the presence of potassium cyanide. The reaction results in the formation of methemoglobin, which then combines with cyanide to form a stable cyanmethemoglobin. When measured spectrophotometrically at 546 nm the absorbance of cyanmethemoglobin follows Beer's Law and is directly proportional to the concentration of hemoglobin released into solution.

[0414] In addition, the formulated test item at each concentration assessed was mixed in a 1:4 ratio with saline and then directly analyzed to ensure that the test item formulations did not interfere with the Siemens Advia measurement of hemoglobin.

[0415] The testing was conducted in 2 phases.

[0416] Two independent analytical runs composing 1 batch were performed using whole blood from 4 normal rats and 1 human donor. The R&D Systems whole blood quality control sets were run before and after the batch to verify operation performance of the instrument using established criteria for this control set per the stated manufacturer's ranges for the control material. An independent (Pointe Scientific) set of Low, Mid, and High controls containing hemoglobin in solution was run once to verify the control rank order of the measurement of hemoglobin in solution phase. The negative and positive control, and test samples were run in singlet.

Hemolytic Potential Criteria

Run Criteria:

[0417] The 1% Saponin positive control must demonstrate hemoglobin value>(Negative Control+0.5 g/dL) for the assay run to be considered acceptable.

[0418] Untreated whole blood must demonstrate hemoglobin values<(Negative Control+0.5 g/dL).

[0419] Saline+untreated whole blood (Negative Control) must demonstrate hemoglobin values <0.5 g/dL.

[0420] R&D Systems whole blood control set should be within the defined manufacturer's target range.

[0421] The Pointe Scientific liquid hemoglobin controls have no established target range for the ADVIA instrument but should demonstrate a rank order in response (based on concentration) of High>Mid>Low.

Sample Analysis Criteria

[0422] The test item+saline sample must demonstrate hemoglobin value<(Negative Control+0.5 g/dL) for the assay run to be considered valid.

[0423] The vehicle control sample must demonstrate hemoglobin value<(Negative Control+0.5 g/dL) for the assay to be considered valid

[0424] The (Pointe Scientific) liquid QC control set containing hemolysates prepared from human erythrocytes ranging from Low, Mid, and High concentrations were used to verify the ability of the Advia to measure liquid hemolysates using the Advia cyanmethemoglobin procedure. The Low, Mid, and High liquid controls from Phase 1 returned values of: 7.9, 11.7, and 15.6 g/dL, respectively, as measured on the Siemens Advia. The Low, Mid, and High liquid controls from Phase 2 returned values of: 8.1, 11.9, and 15.6 g/dL, respectively, as measured on the Siemens Advia. The rank order of the control sets meets the protocol criteria.

[0425] The (R&D System) whole blood quality controls were used to verify the operational performance of the Siemens Advia 120 hematology instrument. The QC values recorded in the raw data were within the defined manufacturer's acceptable ranges for the Advia instrument.

[0426] The amphipathic glycoside Saponin, which complexes with cholesterol to form pores in cell membrane bilayers and cause red blood cell lysis, was utilized for the positive hemolytic control. The 1% Saponin-treated positive controls for rat and human whole blood hemolysis generated a mean hemoglobin concentration of 3.9 and 11.0 g/dL in the supernatant, respectively (Tables 47, 48, 49 and 50). These values met the protocol defined assay acceptability criterion of hemoglobin concentration>(Negative Control+0.5 g/dL).

[0427] The vehicle control (5% Dextrose) in rat and human whole blood in Phases 1 and 2 generated a mean hemoglobin concentration of 0.0 g/dL (Tables 47, 48, 49 and 50). The vehicle control from both phases met the protocol defined sample analysis criterion of <(Negative Control+0.5 g/dL).

[0428] The plasma and negative controls from both phases exhibited a mean hemoglobin concentration of 0.0 g/dL (Tables 47, 48, 49 and 50). These values met the protocol defined assay acceptability criteria of untreated whole blood hemoglobin concentration <(Negative Control+0.5 g/dL) and saline with untreated whole blood hemoglobin concentration <0.5 g/dL.

[0429] Vehicle controls were negative and method controls provided the expected targeted values. All control checks indicate acceptable runs in Phases 1 and 2.

Brincidofovir Hemolytic Potential

[0430] Brincidofovir in rat whole blood at 0.2, 0.5 and 1.6 mg/mL exhibited mean hemoglobin concentrations of 0.0, 0.25 and 1.35 g/dL, respectively (Tables 47, 48, 49 and 50) and as summarized:

TABLE-US-00047 TABLE 45 Brincidofovir Hemolysis in Rat Whole Blood Mean HgB Concentration Estimated % Tube Contents (g/dL) Hemolysis* 1A Test Item (0.2 mg/mL) + 0.0 -- whole blood 1B Test Item (0.5 mg/mL) + 0.25 2.4% whole blood 1C Test Item (1.6 mg/mL) + 1.35 13.0% whole hood *Calculated as: (HgB plasma)/(normal HgB whole blood .times. dilution factor) * 100 Based on normal whole blood rat hemoglobin value of 13 g/dL (as per our HCD)

[0431] Point scientific controls are system tests to verify the measurement of hemoglobin in solution phase. As set forth herein, brincidofovir demonstrated a hemolytic potential effect in rat whole blood that appears to be concentration dependent. Presumptive evidence of this effect was observed at the 0.5 mg/mL concentration and more distinct evidence of hemolysis was observed at the 1.6 mg/mL concentration.

[0432] The estimated % hemolysis for detectable levels of hemoglobin in this study can be calculated using the mean normal hemoglobin concentration for rat whole blood and the appropriate dilution factor. For instance, a rat whole blood normal hemoglobin value of 13 g/dL corrected for dilution by test article, results in a value of 10.4 g/dL (13.times.0.8). The calculated % hemolysis for hemoglobin values of 0.25 and 1.35 g/dL was 2.4% and 13.0% respectively. Formulations with a hemolysis value of <10% are considered to be non-hemolytic while values >25% are considered to be at risk for hemolysis (Amin and Dannenfelser, 2006). Without wishing to be bound by theory, the 0.2 mg/mL concentration showed no detectable hemoglobin and is thus considered non-hemolytic. Concentrations of 0.5 and 1.6 mg/mL resulted in detectable hemoglobin levels in rat whole blood, but the levels noted were not considered a hemolytic risk.

[0433] Results of two (n=2) independent analytical runs from Phase 2 indicated that there was no effect from brincidofovir on hemocompatibility with human whole blood at the concentrations of 0.2 and 0.5 and 1.6 mg/mL and as summarized:

TABLE-US-00048 TABLE 46 Brincidofovir Hemolysis in Human Whole Blood Mean HgB Concentration Estimated % Tube Contents (g/dL) Hemolysis* 1A Test Item (0.2 mg/mL) + 0.0 -- whole blood 1B Test Item (0.5 mg/mL) + 0.0 -- whole blood 1C Test Item (1.6 mg/mL) + 0.0 -- whole blood *Calculated as: (HgB plasma)/(normal HgB whole blood .times. dilution factor) * 100 Based on normal whole blood human female hemoglobin value of 14 g/dL (as per Mayo Clinic on-line data)

[0434] In addition, brincidofovir was not found to interfere with hemoglobin concentration determinations in both Phases 1 and 2 (Tables 47, 48, 49 and 50). Test item controls met the protocol defined sample analysis criterion which indicates acceptable runs in Phases 1 and 2.

[0435] Results of two (n=2) independent analytical runs from Phase 1 indicated without wishing to be bound by theory that brincidofovir demonstrated a hemolytic potential effect in rat whole blood that appears to be concentration dependent. Without wishing to be bound by theory, presumptive evidence of this effect was observed at the 0.5 mg/mL concentration and clear evidence of hemolysis was observed at the 1.6 mg/mL concentration. Formulations with a hemolysis value of <10% are considered to be non-hemolytic while values >25% are considered to be at risk for hemolysis. The 0.2 mg/mL concentration showed no detectable hemoglobin and is considered non-hemolytic. Concentrations of 0.5 and 1.6 mg/mL resulted in detectable hemoglobin levels in rat whole blood, but the levels noted were not considered a hemolytic risk.

[0436] Results of two (n=2) independent analytical runs from Phase 2 indicated that there was no effect from brincidofovir on hemocompatibility with human whole blood at the concentrations of 0.2, 0.5 or 1.6 mg/mL.

[0437] Without wishing to be bound by theory, not all species react the same to the positive hemolytic reagents which may represent RBC fragility differences between species.

TABLE-US-00049 TABLE 47 Summary of Hemolytic Potential Results; Brincidofovir in Rat Blood Phase 1 Mean HgB Concentration Estimated % Tube Contents (g/dL) Hemolysis* 1A Test Item (0.2 mg/mL) + 0.0 -- whole blood 1B Test Item (0.5 mg/mL) + 0.25 2.4% whole blood 1C Test Item (1.6 mg/mL) + 1.35 13.0% whole blood 2A Test Item (0.2 mg/mL) + 0.0 -- saline 2B Test Item (0.5 mg/mL) + 0.0 -- saline 2C Test Item (1.6 mg/mL) + 0.0 -- saline 3 Test Item Vehicle + whole 0.0 -- blood 4 Positive Control (whole 3.9 37.5% blood + 1.0% saponin) 5 Plasma Control (whole blood) 0.0 -- 6 Negative (saline + 0.0 -- whole blood) *Calculated as: (HgB plasma)/(normal HgB whole blood .times. dilution factor) * 100 Based on normal whole blood rat hemoglobin value of 13 g/dL (as per our HCD)

TABLE-US-00050 TABLE 48 Summary of Hemolytic Potential Results; Brincidofovir in Human Blood Phase 2 Mean HgB Concentration Estimated % Tube Contents (g/dL) Hemolysis* 1A Test Item (0.2 mg/mL) + 0.0 -- whole blood 1B Test Item (0.5 mg/mL) + 0.0 -- whole blood 1C Test Item (1.6 mg/mL) + 0.0 -- whole blood 2A Test Item (0.2 mg/mL) + 0.0 -- saline 2B Test Item (0.5 mg/mL) + 0.0 -- saline 2C Test Item (1.6 mg/mL) + 0.0 -- saline 3 Test Item Vehicle + 0.0 -- whole blood 4 Positive Control (whole 11.0 98.2% blood + 1.0% saponin) 5 Plasma Control (whole blood) 0.0 -- 6 Negative (saline + 0.0 -- whole blood) *Calculated as: (HgB plasma)/(normal HgB whole blood .times. dilution factor) * 100 Based on normal whole blood human female hemoglobin value of 14 g/dL (as per Mayo Clinic on-line data)

TABLE-US-00051 TABLE 49 Hemolytic Potential Results (Hemoglobin Concentration in g/dL): Brincidofovir in Rat Blood Phase 1 Tube Number Test Samples 1A 1B 1C 2A 2B 2C 3 Run 1 0.0 0.2 1 0.0 0.0 0.0 0.0 Run 2 0.0 0.3 1.7 0.0 0.0 0.0 Mean 0.0 0.25 1.35 0.0 0.0 0.0 0.0 SD 0.0 0.1 0.5 0.0 0.0 0.0 Tube Number Controls 4 5 6 Run 1 3.9 0.0 0.0

TABLE-US-00052 TABLE 50 Hemolytic Potential Results (Hemoglobin Concentration in g/dL): Brincidofovir in Human Blood Phase 2 Tube Number Test Samples 1A 1B 1C 2A 2B 2C 3 Run 1 0.0 0.0 0.0 0.0 0.0 0.0 0.0 Run 2 0.0 0.0 0.0 0.0 0.0 0.0 Mean 0.0 0.0 0.0 0.0 0.0 0.0 0.0 SD 0.0 0.0 0.0 0.0 0.0 0.0 Tube Number Controls 4 5 6 Run 1 11.0 0.0 0.0 1A = Test Item assay for hemolysis at 0.2 mg/mL 1B = Test Item assay for hemolysis at 0.5 mg/mL 1C = Test Item assay for hemolysis at 1.6 mg/mL 2A = Test Item interference control at 0.2 mg/mL 2B = Test Item interference control at 0.5 mg/mL 2C = Test Item interference control at 1.6 mg/mL 3 = Vehicle control for hemolysis 4 = Hemolytic positive control; 1% Saponin used as a positive control 5 = Untreated whole blood control for hemolysis 6 = Negative control for hemolysis SD = standard deviation

Example 8--Maximum Tolerated Dose and 7-Day Dose Range-Finding (DRF) Study in Rats

[0438] The purpose of this 2-phase study was to determine the maximum tolerated dose (MTD) of brincidofovir, when administered via a single 2-hour intravenous infusion to rats and to assess the toxicity, as well as the single and repeat and toxicokinetic profile of brincidofovir when administered via 2-hour intravenous infusion to rats on Days 1, 4 and 7.

[0439] The results of this study can allow for a decision regarding the dose levels for subsequent toxicity studies of brincidofovir when administered via intravenous infusion.

[0440] The study design incorporated elements of general regulatory guidelines for toxicity studies.

Summary

[0441] This study was comprised of two phases; Phase 1 was a maximum tolerated dose (MTD) study and Phase 2 was a repeat dose range-finding (DRF) study.

[0442] Parameters evaluated during Phase 1 were: viability and clinical observations.

[0443] For Phase 1, Sprague-Dawley CD.RTM. rats (2/sex/dosing interval) were dosed with brincidofovir via 2-hour IV infusion once at 2, 4, 10 or 15 mg/kg. The maximum feasible dose (MFD) was limited to 15 mg/kg because of increased hemolytic potential when the concentration of brincidofovir exceeds 1 mg/mL in the dosing solution. The dose volume was 10 mL/kg/hr for all dosing intervals. Each dose administration was followed by a 2-3 day observation period. At the end of the Phase 1 treatment period, all animals were euthanized and discarded without macroscopic examination.

[0444] Parameters evaluated during Phase 2 were: viability, clinical observations, body weights, food consumption, hematology (termination of dosing), clinical chemistry (termination of dosing) and macroscopic observations.

[0445] For Phase 2, Sprague-Dawley CD.RTM. rats (5/sex/group) were dosed with brincidofovir via 2-hour IV infusion with 0 [2.times. sodium phosphate buffer solution (400 mM, pH 8.0.+-.0.04)], 1 or 15 mg/kg on Days 1, 4 and 7. The dose volume was 10 mL/kg/hr for all groups. At the end of the Phase 2 treatment period, all animals were euthanized and necropsied. Phase 2 satellite animals (3/sex/Group 1 and 6/sex/Group 2-3) were similarly dosed and blood samples collected on Days 1 and 7 for toxicokinetic analysis of brincidofovir and one of its metabolites, cidofovir.

TABLE-US-00053 List of Abbreviations and Definitions of Terms Abbreviation or Specialist Term Explanation AUC.sub.inf Area under the plasma concentration-time curve from time 0 extrapolated to infinity AUC.sub.inf % Extrap Percentage of the AUC.sub.inf that occurred post T.sub.last AUC.sub.last Area under the plasma concentration-time curve from time 0 to time of last measurable plasma concentration AUC.sub.last/D Area under the plasma concentration-time curve from time 0 to time of last measurable plasma concentration normalized to dose BLQ Below the limit of quantitation CL Apparent total body clearance C.sub.max Maximum concentration observed during a dosing interval Brincidofovir Hexadecyloxypropyl-cidofovir CMX021 Brincidofovir metabolite: Cidofovir % CV Coefficient of variation (percent) DRF Dose range finding K.sub.2EDTA Anticoagulant LLOQ Lower limit of quantification M/P ratio: The ratio of exposure parameters (C.sub.max and/or AUC.sub.last) of metabolite versus parent. The M/P ratio is calculated by first converting the relevant parameters to molar units and then dividing the molar exposure value of the metabolite by the molar exposure value of the parent compound. MTD Maximum tolerated dose IV Intravenous dosing TK Toxicokinetic(s) t.sub.1/2 Apparent terminal half-life (aka t.sub.1/2elim) T.sub.last Time of last measurable concentration T.sub.max Time to maximum plasma concentration V.sub.z Apparent volume of distribution, terminal phase

Phase 1

[0446] All animals administered a single dose of brincidofovir at 2, 4, 10 or 15 mg/kg via 2-hour IV infusion survived until their scheduled euthanasia.

[0447] Administration of brincidofovir at 2, 4, or 15 mg/kg resulted in intermittent clinical signs in some male and female animals. Clinical signs were not observed in animals dosed at 10 mg/kg brincidofovir. The intermittent clinical signs consisted of rapid breath and hunched posture with and without piloerection, partially closed eyes, and decreased activity, or irregular breathing, or decreased activity with or without irregular breathing, piloerection, and bilateral partially closed eyes. The clinical signs were reversible and resolved at the end of infusion or within 45 minutes of after the end of the infusion in the Group 1 and 2 animals that displayed signs. The clinical signs were reversible and resolved within 3 hours and 22 minutes of after the end of the infusion in the Group 4 that displayed signs.

[0448] Under the conditions of the study, all doses were well tolerated. Therefore, a maximally tolerated dose level was not determined, and 15 mg/kg was considered the maximum feasible dose (MFD).

Phase 2

[0449] All animals administered three total doses of brincidofovir at 1 and 15 mg/kg/day over a 7 day period survived until their scheduled euthanasia.

[0450] Administration of brincidofovir at 1 mg/kg/day or 15 mg/kg/day did not result in any brincidofovir-related clinical signs in either male or female animals.

[0451] Plasma samples (108) were collected 2 hours following the initiation of dosing on Day 1 and Day 7 (Group 1), or at 1, 2, 8, and 24 hours following the initiation of dosing on Day 1, and prior to administration and at 2, 8, and 24 hours following the initiation of dosing on Day 7 (Groups 2 and 3), and analyzed to determine the concentrations of brincidofovir and the metabolite, cidofovir), by LC/MS/MS.

[0452] Peak concentrations for brincidofovir were generally observed at the end of the 2 hour IV infusion, and decreased rapidly. In general, the mean brincidofovir C.sub.max and AUC increased approximately proportional to the increase in dose from 1 to 15 mg/kg, however the AUC following 1 mg/kg was based on limited concentration-time data. The brincidofovir C.sub.max and AUC.sub.last values on Day 7 showed a trend toward lower values on Day 7 when compared to Day 1 [Day 7/Day 1 accumulation ratios (AR) of 0.44 to 0.54]. No sex differences in brincidofovir TK parameters were observed. Overall, the metabolite cidofovir increased less than proportional to the increase in brincidofovir dose. There were no apparent sex differences in cidofovir TK parameters and no change in TK parameters after repeat administration.

[0453] Brincidofovir-related hematologic findings were limited to slight decreases in reticulocytes in males and females administered 15 mg/kg brincidofovir (-33% and -30% controls, respectively; statistically significant in males only), associated with increases in mean cell hemoglobin concentration (MCHC) in males (+2.1% controls, statistically significant). Decreases red cell mass (hemoglobin, hematocrit, red blood cells) were negligible (to -4.2% controls) at the end of the 7-day dosing phase

[0454] There were minimal brincidofovir-related increases in total calcium in males administered 15 mg/kg brincidofovir (+3.8% controls; not statistically significant) and females administered .gtoreq.1 mg/kg brincidofovir (+4.9% controls; statistically significant) and minimal increases in urea (BUN) in females administered 15 mg/kg (+29% controls; statistically significant).

[0455] Administration of 1 and 15 mg/kg/day brincidofovir via 2-hour IV infusion on Days 1, 4 and 7 are well tolerated.

Route, Duration, and Frequency of Administration

[0456] In Phase 1, each animal was administered a single dose of brincidofovir via 2-hour intravenous infusion. At each dose interval, an escalated dose was administered to naive animals to determine the maximum tolerated dose (MTD). MTD data were used to select doses for Phase 2.

[0457] In Phase 2, the selected doses were administered via 2-hour intravenous infusion on Days 1, 4 and 7 to evaluate the toxicity of repeated doses of the brincidofovir and to aid in the selection of doses for subsequent toxicity studies. A seven-day study can be appropriate for selecting doses for subsequent repeat dose GLP toxicity studies.

Number of Animals

[0458] The number of animals in this study was considered to be the minimum necessary for statistical, regulatory and scientific reasons.

[0459] The number of animals (2/sex/dose interval) in Phase 1 (MTD) was the minimum number that would be required to determine the maximum tolerated dose in male and female rats. Four dose levels of the brincidofovir were expected to be sufficient to determine the maximum tolerated dose. Although there was a 2-3 day observation period, new animals were used at each dose level to eliminate possible additive effects of repeat dosing that could confound determination of a maximum tolerated single dose level.

[0460] The number of main study animals (5/sex/group) in Phase 2 (DRF) was the minimum number that would control for the expected variability among animals. The negative control group and the two brincidofovir-treated groups receiving a low and high multiple of the proposed human dose were considered the minimum number of groups necessary to establish a baseline and provide a range of effects and allow for extrapolation of results for additional repeat dose studies.

[0461] The number of satellite toxicokinetic study animals (6/sex/treated group) in Phase 2 (DRF) toxicokinetic evaluations was considered the minimum number necessary to provide meaningful data, given the inherent variability in distribution, metabolism and excretion processes. A control group with 3 animals per sex confirmed absence of brincidofovir exposure.

Dose Selection

[0462] The starting dose for Phase 1 of this study was 2 mg/kg.

[0463] The low and high doses for Phase 2 of this study (1 and 15 mg/kg) were selected based on the study results of Phase 1 of the present study. In some embodiments, the low and high doses were selected by the lack of clinical signs during the post-administration observation period in animals that were administered 15 mg/kg during Phase 1 of the present study.

Experimental Outline

[0464] For Phase 1 (MTD), up to four dose levels of brincidofovir were administered as escalating single doses in naive male and female rats via intravenous infusion (2-hour). Each dose administration was followed by a 2-3 day observation period. Each subsequent dose level was increased and administered to naive animals based on the response to the preceding doses until the maximum tolerated dose (MTD) was identified, or until the maximum feasible dose (MFD) based on prior evidence of hemolysis of 15 mg/kg was achieved.

TABLE-US-00054 TABLE 51 Phase 1 (MTD) Study Consisted of 4 Dosing Intervals Dose Number of Concen- Volume Interval Treat- Dose animals tration Dose (Group) ment (mg/kg).sup.a Male Female (mg/mL) (mL/kg/hr) 1 BCV 2 2 2 0.1 10 2 BCV 4 2 2 0.2 10 3 BCV 10 2 2 0.5 10 4 BCV 15 2 2 0.75 10 .sup.aDoses represent active ingredient

[0465] For Phase 2 (DRF), the test and control articles were administered, via intravenous 2-hour IV infusion to rats on Days 1, 4, and 7.

TABLE-US-00055 TABLE 52 Phase 2 Study: 1 Control and 2 Treated Groups Number of animals Volume Dose Main study Satellite study.sup.b Concentration Dose Group Treatment (mg/kg).sup.a Male Female Male Female (mg/mL) (mL/kg/hr) 1 Control 0 5 5 3 3 0 10 2 BCV 1 5 5 6 6 0.05 10 3 BCV 15 5 5 6 6 0.75 10 .sup.aDoses represent active ingredient .sup.bSatellite animals used for toxicokinetic blood sampling only.

[0466] A vehicle solution of 2.times. sodium phosphate buffer solution (400 mM, pH 8.0.+-.0.04) was prepared by mixing the appropriate amounts of monobasic sodium phosphate solution (400 mM) and dibasic sodium phosphate solution (400 mM). The pH of the solution was adjusted with monobasic sodium phosphate solution (400 mM), when necessary. The solution was filtered through a 0.22 .mu.m Millex.RTM.-GP filter under a laminar flow hood into a sterile vessel.

[0467] The vehicle (control article) was stored refrigerated 2-8.degree. C. Fresh vehicle solution was prepared once prior to each phase, and used within one month of preparation.

[0468] A brincidofovir stock of 15 mg/mL was perpared by mixing the appropriate amount of brincidofovir with 2.times. sodium phosphate buffer solution, sterile water for injection, USP, and 1N sodium hydroxide (NaOH). The solution was filtered through a 0.22 .mu.m Millex.RTM.-GP filter under a laminar flow hood into a sterile vessel.

[0469] Dose formulations for Phase 1 and 2 were prepared by diluting the appropriate amounts of the brincidofovir stock (15 mg/mL) or of the stock solution vehicle (2.times. sodium phosphate buffer solution) with the appropriate amounts of 5% dextrose for injection, USP, into a sterile vial under a laminar flow hood and inverting 10 times to mix. During Phase 1 and 2, fresh dose formulations were prepared on each dosing day and were stored refrigerated 2-8.degree. C. and protected from light when not in use.

Method of Administration

Route

[0470] Intravenous infusion over 2-hours. Treated at constant doses in 20 mL/kg/2 hours.

Infusion Catheter Implantation Procedure

[0471] Catheters for infusion were implanted approximately 1-2 weeks prior to dose administration. All animals, including the spares, were surgically implanted with a catheter.

[0472] The surgical site was prepared as per Testing Facility's SOP for aseptic, recovery surgical procedures. Animals received analgesics pre-emptively (flunixin meglumine (USP) 2 mg/kg, subcutaneously).

[0473] All animals were anesthetized (isoflurane) and shaved in the inguinal and dorsal regions. A small incision was made and the femoral vein isolated. A small incision in the vessel was performed and a sterile catheter was placed into the vessel. The catheter was inserted via the femoral vein and passed retrograde into the inferior vena cava, just caudal to the kidneys. The catheter was secured with sutures and passed subcutaneously so as to exit at the nape of the neck. A small pocket was made at the exteriorization site in which a loop of catheter remained. The inguinal region was closed by sutures.

[0474] The animals were placed in jackets and the implanted catheters were attached to pins with capped septum connectors. The catheters were locked with taurolidine citrate locking solution. Each animal received enrofloxacin 5.0 mg/kg intramuscularly on the day of surgery.

Infusion Catheter Maintenance Procedure

[0475] Implanted femoral vein catheters were assessed for patency as per Testing Facility's SOP prior to placing animals on study.

Volume Dose (Rate)

[0476] 20 mL/kg/2 hours (10 mL/kg/hour).

Individual Dose Concentration

[0477] The individual animal concentrations were calculated from the most recently recorded scheduled bodyweight.

Frequency and Duration

[0478] During Phase 1, each animal received a single dose administered via intravenous infusion (2-hours), followed by a 2-3 day observation period after each dose interval. Following the single intravenous (2-hour) infusion, the animals had their jackets and dosing sets removed and were not returned to saline maintenance.

[0479] During Phase 2, each animal received brincidofovir administrations via intravenous infusion (2-hours) on Days 1, 4 and 7. Animals were maintained on sterile saline at a rate of 0.5 mL/hour between doses. Catheters were tied off after the completion of dose administration on Day 7 (a knot was placed in the catheter and the catheter was receded under the skin) and jackets were removed.

Dose Site

[0480] Surgically implanted cannula in the femoral vein/vena cava.

Dosing Procedure

[0481] One to two days prior to dose initiation, the lock solution was withdrawn from each animal's implanted catheter (if possible) and the catheter was flushed with saline before connecting to a tether and infusion dosing set. After connecting the animals' catheters, the animals were infused with sterile saline (0.9% NaCl, USP) at a rate of 0.5 mL/hr by a calibrated Medfusion syringe pump until dose administration on Day 1.

[0482] A saline flush was infused to deliver the brincidofovir in the infusion lines and to ensure the delivery of a complete dose (an additional .about.0.5 to 1 mL volume, at the same rate as the brincidofovir, was administered to flush the catheter line).

Dosing Accuracy

[0483] The accountability (confirmation of actual dose administered) for each day of infusion was calculated and reviewed for adjustments daily. Pumps were checked for accuracy prior to dose initiation and following end of dose administration for each phase.

Collection Times and Number of Animals

[0484] Blood samples were obtained for the determination of plasma concentrations of brincidofovir during Phase 2.

TABLE-US-00056 TABLE 53 Collection Times and Number of Animals Interval Timepoints Number of Animals Day 1 2 hours after the initiation of 3 animals/sex/Group 1 dose administration 1, 2, 8 and 24 hours after the 3 animals/sex/Groups 2-3 initiation of dose administration Day 7 2 hours after the initiation of 3 animals/sex/Group 1 dose administration Predose, and 2, 8 and 24 hours 3 animals/sex/Groups 2-3 after the initiation of dose administration

[0485] On Days 1 and 7, blood samples were obtained for toxicokinetic determinations at the above timepoints.

Collection Procedures

[0486] Approximately 0.4 mL of whole blood was obtained via the tail vein from each animal at each time point. Animals were not fasted prior to blood collection. Blood was collected into tubes containing K.sub.2EDTA anticoagulant and placed on wet ice in an upright position, per facility SOP prior to processing. Plasma was separated by centrifugation (10 minutes at approximately 2000 g, at approximately 2-8.degree. C.). Approximately 0.10 mL of plasma was transferred into a single cryotube appropriately labeled with study number, animal number, time point, date of sampling and sample type. Remaining plasma was transferred into a second cryotube and retained as a backup sample. Plasma frozen at approximately -80.+-.10.degree. C. within approximately 2 hours after collection of each blood sample until analysis. Animals were euthanized (CO.sub.2 inhalation) after the final blood collection.

[0487] All plasma sample tubes were stored frozen (-80.+-.10.degree. C.) and shipped (frozen, on dry ice) to Pyxant Labs, Colorado Springs, Colorado for analysis. Samples were shipped within 1 month of collection.

[0488] In general, TK parameters were estimated using mean plasma concentrations of brincidofovir and cidofovir derived from the composite blood sampling design using Phoenix WinNonLin (V 6.3).

[0489] Animals were observed in their cages twice daily for mortality and signs of severe toxic or pharmacologic effects.

[0490] For dose observations, Phase 1: Signs of poor health or toxic or pharmacologic effects (e.g., abnormalities in general condition, appearance, activity, behavior, respiration, etc.) observed during infusion period were recorded.

[0491] Animals were removed from their cages and examined once pretest. Examinations included observations of general condition, skin and fur, eyes, nose, oral cavity, abdomen and external genitalia as well as evaluations of respiration.

Body Weight

[0492] Phase 1: Animals were removed from their cages and weighed twice pretest and prior to each dose.

[0493] Phase 2: Animals were removed from their cages and weighed twice pretest, prior to each dose and following the last dose administered during the afternoon of Day 7. Terminal, fasted body weights were obtained just prior to necropsy.

[0494] Food consumption was measured (weighed) weekly, beginning one week prior to dosing for Phase 2.

[0495] Blood obtained via the orbital sinus (retrobulbar venous plexus) as a terminal procedure under light isoflurane anesthesia was used to analyze hematology and clinical chemistry parameters for all animals at termination of dosing. Animals were fasted overnight prior to blood collection. Animals were not allowed to recover from anesthesia after blood collection.

Hematology

[0496] Blood samples (approximately 0.25 mL) were collected into tubes containing K.sub.2EDTA anticoagulant and analyzed for the following using a Siemens ADVIA 120 Hematology Analyzer: Hemoglobin concentration (HGB); Hematocrit (HCT); Erythrocyte count (RBC); Platelet count (PLT); Mean corpuscular volume (MCV); Mean corpuscular hemoglobin (MCH); Mean corpuscular hemoglobin concentration (MCHC); Red cell distribution width (RDW); Total leukocyte count (WBC); Reticulocyte count (RETIC); Differential leukocyte count (Manual differential leukocyte counts were performed for verification and absolute values were calculated if necessary); Neutrophils (ANEU); Lymphocytes (ALYM); Eosinophils (AEOS); Basophils (ABASO); Monocytes (AMONO); Large unstained cells (ALUC);

[0497] A peripheral blood smear was prepared for each animal at each blood collection interval and was available for confirmation of automated results and/or other evaluations deemed necessary by the Clinical Pathologist.

Clinical Chemistry

[0498] Blood samples (approximately 1 mL) were collected into tubes with no anticoagulant, allowed to clot, centrifuged to obtain serum and analyzed for the following using a Siemens ADVIA 1800 Chemistry Analyzer: Aspartate aminotransferase (AST) (Kinetic--Modified Bergmeyer); Alanine aminotransferase (ALT) (Kinetic--Modified Bergmeyer); Alkaline phosphatase (ALKP) (Kinetic--Tietz AMP Buffer); Blood urea nitrogen (BUN) (Enzymatic Roch-Ramek with Urease); Creatinine (CREAT) (Jaffe Picric Acid in Alkaline Medium); Glucose (GLU) (Glucose Hexokinase II Method); Cholesterol (CHOL) (Enzymatic esterase/oxidase Trinder Endpoint); Triglycerides (TRIG) (Fossati Three Step Enzymatic--Trinder Endpoint); Total protein (TP) (Biuret Technique); Albumin (ALB) (Bromocresol Green Method); Total bilirubin (TBILI) (Oxidation with Vandate); Sodium (NA.sup.+) (Ion Selective Electrode); Potassium (K.sup.+) (Ion Selective Electrode); Chloride (Cl.sup.-) (Ion Selective Electrode); Calcium (Ca.sup.++) (Michaylova & Ilkova, Arsenazo III); Inorganic phosphorus (PHOS) (Phosphomolybdate--UV Method).

Postmortem

[0499] Phase 1: All animals were euthanized by exsanguination following carbon dioxide inhalation and discarded without macroscopic examination after the completion of the observation period.

[0500] Phase 2: Main DRF study animals were euthanized by exsanguination following isoflurane inhalation and TK satellite animals via carbon dioxide inhalation.

[0501] Necropsy was performed on the Phase 2 main study animals on Day 8. Animals were fasted overnight prior to necropsy.

[0502] Complete macroscopic examinations were performed on all Phase 2 main study animals. The macroscopic examination included examination of the external surface and all orifices; the external surfaces of the brain and spinal cord; the organs and tissues of the cranial, thoracic, abdominal and pelvic cavities and neck; and the remainder of the carcass for the presence of macroscopic morphologic abnormalities. Animals were discarded after examination; no tissues were collected or preserved.

[0503] The following abbreviations were used for hematology: CS: Clotted specimen; LA: Lab accident; NVIM; Not valid due to improbable result; CLSE: Severe platelet clumping noted; CLSL: Slight platelet clumping noted; NCLP: No clumping.

[0504] Blood chemistry: TBILI values below the analytical limit (<0.2) are excluded from calculations.

Globulin (GLOB) was calculated as: total protein-albumin

Albumin/globulin ratio (A/G) was calculated as: albumin/globulin

[0505] All statistical analyses were carried out separately for males and females using the individual animal as the basic experimental unit.

[0506] The following data types were analyzed at each timepoint separately for Phase 2: body weight; body weight change from interval to interval; cumulative body weight change from baseline; food consumption, hematology, and clinical chemistry. The parameters to analyze were identified as continuous, discrete or binary. Brincidofovir treated groups were then compared to the control using the following procedures.

In-Life Study Conduct

[0507] Table 54 below summarizes details regarding the study conduct, including but not limited to test animals, study materials, study design, dosing, observations, and results.

TABLE-US-00057 TABLE 54 Experimental Design Study Summary (phase 2) Species and Strain: Vehicle: 200 mM Sodium Male and Female [Crl: CD (SD)BR] Phosphate Buffer, pH 8, Variable (Sprague-Dawley derived) dilution with 5% Dextrose Study Design Group Dose Number (mg/kg) Sampling Scheme 1 0 Day 1 and Day 7: 3 animals/sex: 2 hours after the initiation of dose vehicle administration 2 1 Day 1: 3 animals/sex/timepoint: 1, 2, 8, and 24 hours after the initiation of dose administration Day 7: 3 animals/sex/timepoint: Predose, 2, 8, and 24 hours after the initiation of dose administration 3 15 Same as Group 2

Sampling

[0508] A composite blood sampling design was used such that each rat was sampled twice per sampling day. Blood samples were collected according to the scheme shown in Table 54. Blood was collected via tail vein from unanesthetized animals into tubes containing anticoagulant and placed on wet ice in an upright position. Animals were not fasted prior to blood collection. Plasma was separated by centrifugation (for 10 minutes at approximately 2000 g, at approximately 2-8.degree. C.), and transferred into individually labeled cryotubes. All cryotubes containing the collected plasma samples were appropriately labeled as to study number, animal number, time point, date of sampling and sample type. All plasma samples were obtained and frozen at approximately -80.degree. C. (.+-.10.degree. C.) within approximately 2 hours after collection of each blood sample until analysis.

[0509] All plasma samples were stored frozen [approximately -80.degree. C. (.+-.10.degree. C.)] and shipped (frozen, on dry ice) to Pyxant Laboratories for analysis.

Bioanalytical Methods

[0510] Rat plasma bioanalysis was conducted by Pyxant Laboratories. Plasma samples were analyzed for concentrations of brincidofovir and cidofovir using a method based upon protein precipitation extraction followed by LC-MS/MS analysis; calibration ranges for brincidofovir and cidofovir were 1.00-1500 ng/mL and 5.00-750 ng/mL, respectively, (Pyxant study number 3025) for a 50 .mu.L aliquot of rat plasma. Sample analysis for ISR was not performed as part of this non-GLP study.

Data Analysis

Toxicokinetic Analysis

[0511] For the generation of Mean Profiles, all sample concentrations that were below the limit of quantitation (BLQ) were set to 0.

[0512] These values are recorded in Table 61 and Table 62, with the original BLQ result in the column labelled [Concentration] and the imputed values for calculation of the mean values in the column labelled [Imputed Concentration].

[0513] The treatment of Mean Profile BLQ values for the purposes of NCA, are recorded in Table 63 and Table 64, with the original Mean Profile BLQ result in the column labeled [Mean Concentration] and the imputed values used for NCA analysis in the column labeled [Imputed Mean Concentration]. BLQ values at early time points (prior to T.sub.max) were set to 0, BLQ at time points after T.sub.max were set to missing.

[0514] All pharmacokinetic parameter estimates were completed using a Non-Compartmental Model (Model 200-202 for IV infusion administration). The AUC.sub.last and AUC.sub.inf were calculated using the linear up log down trapezoidal rule.

[0515] Statistical Analysis was limited to descriptive statistical analysis including arithmetic mean, standard deviation, % CV of the arithmetic mean.

[0516] The coefficient of variation (% CV) for mean plasma concentrations ranged from (9.98 to 173% CV) for brincidofovir and (4.82 to 173% CV) for cidofovir.

[0517] Peak concentrations for brincidofovir were generally observed at the end of the 2 hour IV infusion, and decreased rapidly; brincidofovir concentrations were at or near the lower limit of quantification (LLOQ, 1 ng/mL) 8 hours after initiation of the 1 mg/kg dose and 24 hours after the 15 mg/kg dose. Terminal half-life (t1/2), CL, and Vss were not determinable due to insufficient data points to characterize the elimination phase. In general, mean brincidofovir C.sub.max and AUC increased approximately proportional to the increase in dose from 1 to 15 mg/kg, though AUC was determined on few concentration values after 1 mg/kg dosing. The brincidofovir C.sub.max and AUC.sub.last values on Day 7 showed a trend toward lower values on Day 7 compared to Day 1 (Day7/Day1 accumulation ratios (AR) of 0.44 to 0.53). Further, no sex differences in brincidofovir TK parameters were observed.

[0518] Overall, exposure to metabolite cidofovir increased less than proportional to the increase in brincidofovir dose, there were no apparent sex differences in TK parameters, and no change in TK parameters after repeat administration.

[0519] Bioanalysis was performed on 108 plasma samples collected only during Phase 2 to determine the concentrations of brincidofovir and the cidofovir metabolite, cidofovir. The plasma samples were collected at 2 hours following the initiation of dosing on Day 1 and Day 7 (Group 1), or at 1, 2, 8, and 24 hours following the initiation of dosing on Day 1, and prior to administration and at 2, 8, and 24 hours following the initiation of dosing on Day 7 (Groups 2 and 3) and analyzed by LC/MS/MS.

[0520] Toxicokinetic analysis of the plasma concentrations of brincidofovir and cidofovir was conducted for Phase 2 only.

[0521] On Days 1 and 7, brincidofovir C.sub.max increased in approximate proportion to dose (for female rats), or less than proportionally to dose (for male rats). For a 15-fold increase in dose from 1 mg/kg to 15 mg/kg, C.sub.max increased 17.3- to 18.2-fold for females and 9.2- to 18.6-fold for males. On Days 1 and 7, brincidofovir AUC.sub.last increased in approximate proportion (for female rats); for a 15-fold increase in dose, the AUC.sub.last increased 14.1- to 16.1-fold. Due to insufficient data points in male rats after 1 mg/kg administration, proportionality of AUC.sub.last could not be determined.

[0522] Between female and male animals, the brincidofovir C.sub.max on Days 1 and 7 for a 1 or 15 mg/kg dose were similar (difference within 54%). The brincidofovir AUC.sub.last for a 1 mg/kg dose could not be compared due to insufficient data points to calculate AUC in males. The brincidofovir AUC.sub.last on Days 1 and 7, for a 15 mg/kg dose, were similar (difference within 16%) between female and male animals.

[0523] The brincidofovir C.sub.max and AUC.sub.last values on Day 7 for female animals following twice-weekly dosing of brincidofovir at 1 mg/kg, and for both female and male animals after 15 mg/kg, showed a trend toward lower C.sub.max and AUC.sub.last on Day 7 compared to Day 1 (Day7/Day1 accumulation ratios (AR) of 0.44 to 0.53).

TABLE-US-00058 TABLE 55 Brincidofovir TK Parameters (Phase 2) Following Single and Multiple 2-Hour Intravenous Infusion Administrations of brincidofovir on Days 1, 3 and 7 to Rats Brincidofovir Dose (mg/kg) 1 15 Male Female Male Female Period Period TK Parameter Day 1 Day 7 Day 1 Day 7 Day 1 Day 7 Day 1 Day 7 T.sub.max (h) 2 2 2 2 1 2 1 2 T.sub.last (h) 2 2 8 8 24 24 24 24 C.sub.max (ng/mL) 235 208 191 95.4 4370 1910 3470 1660 C.sub.max/Dose 235 208 191 95.4 291 127 231 110 [(ng/mL)/(mg/kg)] AUC.sub.last NR NR 459 244 8780 4090 7360 3420 (h*ng/mL) AUC.sub.last/Dose NR NR 459 244 585 273 491 228 [(h*ng/mL)/(mg/kg)] AR: C.sub.max NA NR NA 0.50 NA 0.44 NA 0.48 Day 7/Day 1 AR: AUC.sub.last NA NR NA 0.53 NA 0.47 NA 0.46 Day 7/Day 1 NA, not applicable; NR, not reportable due to insufficient data points

[0524] As set forth in Tables 55 and 56, on Days 1 and 7, cidofovir C.sub.max generally increased less than proportional to dose. For a 15-fold increase in brincidofovir dose from 1 mg/kg to 15 mg/kg, cidofovir C.sub.max increased 2.9- to 4.1-fold for female animals and 5.6- to 16.8-fold for males. On Days 1 and 7, cidofovir AUC.sub.last increased less than proportionally to dose for female animals; for a 15-fold increase in brincidofovir dose, cidofovir AUC.sub.last increased 9.4- to 16.2-fold. Due to insufficient data points in male rats after 1 mg/kg administration, proportionality of AUC.sub.last could not be determined.

[0525] The cidofovir C.sub.max and AUC.sub.last on Days 1 and 7, after a 15 mg/kg brincidofovir dose, were similar (difference within 33%) between female and male animals.

[0526] The cidofovir C.sub.max and AUC.sub.last on Day 7 for female animals following twice-weekly dosing of brincidofovir at 1 mg/kg showed a trend toward higher C.sub.max and AUC.sub.last on Day 7 compared to Day 1 (Day7/Day1 AR ranged from 1.6 to 1.9). However, cidofovir C.sub.max and AUC.sub.last for both female and male animals administered 15 mg/kg (3 doses), were similar (Day7/Day1 AR ranged from 1.0 to 1.1).

[0527] The metabolite-to-parent (M/P) ratio, calculated on a molar basis, of AUC.sub.last on Day 1 for female animals were 0.190 and 0.661 for doses of 1 mg/kg and 15 mg/kg of brincidofovir, respectively. The M/P ratios on Day 7 for female animals were 0.193 and 0.444 for doses of 1 mg/kg and 15 mg/kg of brincidofovir, respectively.

[0528] Due to insufficient data points in male rats after 1 mg/kg administration, the M/P ratio for AUC.sub.last could not be determined. The M/P ratios on Day 1 and 7 after 15 mg/kg administration of brincidofovir were 0.126 and 0.307, respectively.

TABLE-US-00059 TABLE 56 Cidofovir TK Parameters Following Single and Multiple 2-Hour Intravenous Infusion Administrations of brincidofovir on Days 1, 3 and 7 to Rats Brincidofovir Dose (mg/kg) 1 15 Male Female Male Female Period Period TK Parameter Day 1 Day 7 Day 1 Day 7 Day 1 Day 7 Day 1 Day 7 T.sub.max (h) 8 8 2 2 8 8 8 8 T.sub.last (h) 8 8 8 8 24 24 24 24 C.sub.max (ng/mL) 1.95 5.73 9.35 15 32.8 32.8 38.6 43.6 C.sub.max/Dose 1.95 5.73 9.35 15 2.18 2.18 2.57 2.91 [(ng/mL)/(mg/kg)] AUC.sub.last NR NR 43.4 80.1 548 624 705 756 (h*ng/mL) AUC.sub.last/Dose NR NR 43.4 80.1 36.5 41.6 47 50.4 [(h*ng/mL)/(mg/kg)] AR: C.sub.max NA 2.94 NA 1.60 NA 1.00 NA 1.13 Day 7/Day 1 AR: AUC.sub.last NA NR NA 1.85 NA 1.14 NA 1.07 Day 7/Day 1 M/P Ratio: C.sub.max 0.017 0.056 0.099 0.316 0.035 0.015 0.022 0.053 (molar equivalents) M/P Ratio: AUC.sub.last NR NR 0.190 0.661 0.126 0.307 0.193 0.444 (molar equivalents)

[0529] For Phase 1 and Phase 2, all animals survived until scheduled termination.

Clinical Observations

Phase 1

[0530] Dose 1 (2 mg/kg): Clinical signs in both male animals consisted of rapid breath and hunched posture, with and without piloerection, partially closed eyes, and decreased activity.

[0531] Dose 2 (4 mg/kg); Clinical signs in both males and one female animal consisted of irregular breathing.

[0532] Dose 3 (10 mg/kg): No clinical signs were observed in either male or female animals.

[0533] Dose 4 (15 mg/kg): Clinical signs in both male and one female animal included decreased activity with or without irregular breathing, piloerection, and bilateral partially closed eyes.

[0534] All clinical signs were reversible and resolved shortly after the end of the infusion.

Phase 2

[0535] Dose administration of brincidofovir at 1 mg/kg/day or 15 mg/kg/day did not result in any brincidofovir-related clinical signs in either male or female animals. There were no effects on body weights (only Phase 2 evaluated) or food consumption related to treatment with brincidofovir.

Clinical Pathology

[0536] Brincidofovir-related hematologic findings were limited to slight decreases in reticulocytes in males and females at 15 mg/kg (-33% and -30% controls, respectively; statistically significant in males only), associated with increases in mean cell hemoglobin concentration (MCHC) in males (+2.1% controls, statistically significant). Decreases in red cell mass (hemoglobin, hematocrit, red blood cells) were negligible (to -4.2% controls) at the end of the 7-day dosing phase. The minimal effect of reticulocyte decreases on red cell mass was attributed to the long lifespan of red blood cells in rats (.about.45-68 days) relative to reticulocytes (2-5 days). Red cell mass decreases would be expected to be more pronounced with continued dosing. Decreases in reticulocytes were indicative of decreased erythropoiesis in hematopoietic tissues. In the absence of decreases in food consumption or body weight or poor clinical condition, they were likely to be due to brincidofovir-related suppression of erythropoiesis, which is not unexpected with this class of drug (nucleotide analog).

[0537] There were minimal brincidofovir-related increases in total calcium in males at 15 mg/kg (+3.8% controls; not statistically significant) and females at >1 mg/kg (+4.9% controls; statistically significant) and minimal increases in urea (BUN) in females at 15 mg/kg (+29% controls; statistically significant). Both findings can be observed with altered kidney function (decreased urinary excretion). In addition, without wishing to be bound by theory, increases in total calcium may reflect increased mobilization from bone or increased intestinal absorption.

[0538] Changes in Animal No. 3048 (slight increases in BUN, creatinine and phosphorus) were not considered to be brincidofovir-related and secondary to decreased glomerular filtration rate due to absence of a left kidney (noted macroscopically).

[0539] All other differences from controls, statistically significant or otherwise, were not considered to be brincidofovir-related due to their direction, small magnitude, infrequent occurrence, lack of relation to dose, lack of concordance with related endpoints, and/or because values were comparable to the study control range.

Pathology

[0540] Macroscopic findings were few, occurred sporadically, and were not considered to be brincidofovir-related as they occurred at a similar incidence in control animals, lacked a dose relationship, or were considered congenital anomalies (Animal No. 3048). Male Animal No. 3048 did not have a left seminal vesicle or a left kidney; the absence of the left kidney correlated with elevated blood urea nitrogen (BUN), creatinine, and phosphorus in this animal.

[0541] For Phase 1, under the conditions of the study, all doses were well tolerated when administered via a single 2-hour IV infusion. Therefore, the maximally tolerated dose of brincidofovir was not determined. However, 15 mg/kg delivered via 2-hour IV infusion of a 0.75 mg/mL solution of brincidofovir represents the maximum feasible dose based on in vitro evidence of hemolysis at brincidofovir concentrations above 0.75 mg/mL.

[0542] For Phase 2, peak concentrations for brincidofovir were generally observed at the end of the 2-hour IV infusion, and decreased rapidly.

[0543] In general, mean brincidofovir C.sub.max and AUC increased approximately proportional to the increase in dose from 1 to 15 mg/kg, though AUC was determined on few concentration values after 1 mg/kg dosing. The brincidofovir C.sub.max and AUC.sub.last values on Day 7 showed a trend toward lower values on Day 7 compared to Day 1 (Day7/Day1 accumulation ratios (AR) of 0.44 to 0.53). Further, no sex differences in brincidofovir TK parameters were observed. Overall, exposure to metabolite cidofovir increased less than proportional to the increase in brincidofovir dose, there were no apparent sex differences in TK parameters, and no change in TK parameters after repeat administration Under the conditions of the study, single dose administration of 1 or 15 mg/kg brincidofovir via 2-hour IV infusion on Days 1, 4 and 7 were well tolerated.

Concentrations of Brincidofovir in Plasma

[0544] The concentration-time data for brincidofovir following single and repeat administration of 1 mg/kg and 15 mg/kg are summarized in Table 59 and Table 60.

[0545] The concentrations of brincidofovir and cidofovir in all control plasma samples were BLQ for each analyte (<1.0 ng/mL and <5.0 ng/mL, respectively).

[0546] Following single IV infusion administration of brincidofovir over a 2 hour period, plasma concentrations of brincidofovir were detected in the first sample (1 hour) at both dose levels (1 mg/kg and 15 mg/kg). Following twice-weekly administration (3 doses on Days 1, 3 and 7), brincidofovir was BLQ in predose samples, but was detected in the first postdose sample (2 hour, end of the infusion) on Day 7. The median time of peak concentration (T.sub.max) of brincidofovir occurred at 2 hour following single and twice weekly 1 mg/kg dosing, and at 1 hour following single 15 mg/kg dosing and at 2 hours following twice weekly 15 mg/kg dosing. Concentrations of brincidofovir reached BLQ in all males and some females 8 hours following administration of 1 mg/kg and were at or near the LLOQ by 24 hours following administration of 15 mg/kg. The coefficient of variation (% CV) for mean plasma concentrations for brincidofovir ranged from 13.0 to 173% for female animals and from 9.98 to 28.1% for male animals. In general, the highest % CV was observed at later time points when concentrations were at or near the LLOQ.

Toxicokinetics of Brincidofovir in Plasma

[0547] Mean plasma brincidofovir concentration-time information used in the input file for Phoeneix WinNonlin analysis are contained in Table 63. TK parameters for brincidofovir following single and twice weekly administration of brincidofovir as an intravenous infusion to rats are summarized in Table 56. Terminal half-life (t1/2), CL, and V.sub.ss were not determinable due to insufficient data points to characterize the elimination phase.

[0548] On Days 1 and 7, brincidofovir C.sub.max increased in approximate proportion to dose (for female rats), or less than proportionally to dose (for male rats). For a 15-fold increase in dose from 1 mg/kg to 15 mg/kg, C.sub.max increased 17.3- to 18.2-fold for females and 9.2- to 18.6-fold for males. On Days 1 and 7, brincidofovir AUC.sub.last increased in approximate proportion (for female rats); for a 15-fold increase in dose, the AUC.sub.last increased 14.1- to 16.1-fold. Due to insufficient data points in male rats after 1 mg/kg administration, proportionality of AUC.sub.last could not be determined.

[0549] Between female and male animals, the brincidofovir C.sub.max on Days 1 and 7 for a 1 or 15 mg/kg dose were similar (difference within 54%). The brincidofovir AUC.sub.last for a 1 mg/kg dose could not be compared due to insufficient data points to calculate AUC in males. The brincidofovir AUC.sub.last on Days 1 and 7, for a 15 mg/kg dose, were similar (difference 16%) between female and male animals.

[0550] The brincidofovir C.sub.max and AUC.sub.last values on Day 7 for female animals following twice-weekly dosing of brincidofovir at 1 mg/kg, and for both female and male animals after 15 mg/kg, showed a trend toward lower C.sub.max and AUC.sub.last on Day 7 compared to Day 1 [Day7/Day1 accumulation ratios (AR) of 0.44 to 0.53].

TABLE-US-00060 TABLE 57 Brincidofovir TK Parameters Following Single and Multiple 2-Hour Intravenous Infusion Administrations of brincidofovir on Days 1, 3 and 7 to Rats Brincidofovir Dose (mg/kg) 1 15 Male Female Male Female Period Period TK Parameter Day 1 Day 7 Day 1 Day 7 Day 1 Day 7 Day 1 Day 7 T.sub.max (h) 2 2 2 2 1 2 1 2 T.sub.last (h) 2 2 8 8 24 24 24 24 C.sub.max (ng/mL) 235 208 191 95.4 4370 1910 3470 1660 C.sub.max/Dose 235 208 191 95.4 291 127 231 110 [(ng/mL)/(mg/kg)] AUC.sub.last (h*ng/mL) NR NR 459 244 8780 4090 7360 3420 AUC.sub.last/Dose NR NR 459 244 585 273 491 228 [(h*ng/mL)/(mg/kg)] AR: C.sub.max NA NR NA 0.50 NA 0.44 NA 0.48 Day 7/Day 1 AR: AUC.sub.last NA NR NA 0.53 NA 0.47 NA 0.46 Day 7/Day 1

Concentrations of Cidofovir in Plasma

[0551] The concentration-time data for cidofovir following single and repeat daily administration of 1 mg/kg and 15 mg/kg are summarized in Table 59 through Table 60.

[0552] In female animals following single IV infusion administration of brincidofovir, plasma concentrations of cidofovir were detected 2 hours following administration of 1 mg/kg (second time point) or 1 hour following administration of 15 mg/kg (first time point). Following twice-weekly administration of brincidofovir, plasma concentrations of cidofovir were BLQ in predose samples, but were detected in all of the 2 hour samples (first postdose time point) on Day 7. The median T.sub.max of cidofovir occurred 2 hours following single and twice-weekly 1 mg/kg brincidofovir dosing, and at 8 hours following single and twice-weekly 15 mg/kg brincidofovir dosing.

[0553] In male animals following single IV infusion administration of brincidofovir, detectable plasma concentrations of cidofovir were observed only in a single time point 8 hours following administration of 1 mg/kg. However, cidofovir was detected in the first sample (1 hour) following administration of 15 mg/kg. Following twice-weekly administration of brincidofovir, plasma concentrations of cidofovir were BLQ in the predose samples and in the first postdose sample (2 hours), but were detected in the 8 hour samples on Day 7 following administration of 1 mg/kg, and in the 2 hour samples (first postdose sample) on Day 7 following administration of 15 mg/kg. The median T.sub.max of cidofovir occurred at 8 hours following single or twice-weekly 1 mg/kg or 15 mg/kg brincidofovir dosing. In both male and female animals, concentrations of cidofovir reached BLQ by 24 hours following single and twice-weekly administration 1 mg/kg brincidofovir and did not reach BLQ by 24 hours (last time point) following single or twice-weekly administration of 15 mg/kg brincidofovir. The % CV for mean concentrations of cidofovir ranged from 4.82 to 88.4% for female animals and 5.49 to 173% for male animals. In general, the highest % CV was observed at early or later time points when cidofovir concentrations were at or near the LLOQ.

Toxicokinetics of Cidofovir in Plasma

[0554] Mean plasma cidofovir concentration-time information used in the input file for Phoenix WinNonlin analysis are contained in Table 64. TK parameters for cidofovir following single and twice weekly administration of brincidofovir as an intravenous infusion to rats are summarized in Table 58.

[0555] On Days 1 and 7, cidofovir C.sub.max generally increased less than proportional to dose. For a 15-fold increase in brincidofovir dose from 1 mg/kg to 15 mg/kg, cidofovir C.sub.max increased 2.9- to 4.1-fold for female animals and 5.7- to 16.8-fold for males. On Days 1 and 7, cidofovir AUC.sub.last increased less than proportionally to dose for female animals; for a 15-fold increase in brincidofovir dose, cidofovir AUC.sub.last increased 9.4- to 16.2-fold. Due to insufficient data points in male rats after 1 mg/kg administration, proportionality of AUC.sub.last could not be determined.

[0556] The cidofovir C.sub.max and AUC.sub.last on Days 1 and 7, after a 15 mg/kg brincidofovir dose, were similar (difference within 33%) between female and male animals.

[0557] The cidofovir C.sub.max and AUC.sub.last on Day 7 for female animals following twice-weekly dosing of brincidofovir at 1 mg/kg showed a trend toward higher C.sub.max and AUC.sub.last on Day 7 compared to Day 1 (Day7/Day1 AR ranged from 1.6 to 1.9). However, cidofovir C.sub.max and AUC.sub.last for both female and male animals administered 15 mg/kg (3 doses), were similar (Day7/Day1 AR ranged from 1.0 to 1.1).

[0558] The metabolite-to-parent (M/P) ratio of AUC.sub.last on Day 1 for female animals were 0.190 and 0.661 for doses of 1 mg/kg and 15 mg/kg of brincidofovir, respectively. The M/P ratios on Day 7 for female animals were 0.193 and 0.444 for doses of 1 mg/kg and 15 mg/kg of brincidofovir, respectively.

[0559] Due to insufficient data points in male rats after 1 mg/kg administration, the M/P ratio for AUC.sub.last could not be determined. The M/P ratios on Day 1 and 7 after 15 mg/kg administration of brincidofovir were 0.126 and 0.307, respectively.

TABLE-US-00061 TABLE 58 cidofovir TK Parameters Following Single and Multiple 2-Hour Intravenous Infusion Administrations of brincidofovir on Days 1, 3 and 7 to Rats Brincidofovir Dose (mg/kg) 1 15 Male Female Male Female Period Period TK Parameter Day 1 Day 7 Day 1 Day 7 Day 1 Day 7 Day 1 Day 7 T.sub.max (h) 8 8 2 2 8 8 8 8 T.sub.last (h) 8 8 8 8 24 24 24 24 C.sub.max (ng/mL) 1.95 5.73 9.35 15 32.8 32.8 38.6 43.6 C.sub.max/Dose 1.95 5.73 9.35 15 2.18 2.18 2.57 2.91 [(ng/mL)/(mg/kg)] AUC.sub.last (h*ng/mL) NR NR 43.4 80.1 548 624 705 756 AUC.sub.last/Dose NR NR 43.4 80.1 36.5 41.6 47 50.4 [(h*ng/mL)/(mg/kg)] AR: C.sub.max NA 2.94 NA 1.60 NA 1.00 NA 1.13 Day 7/Day 1 AR: AUC.sub.last NA NR NA 1.85 NA 1.14 NA 1.07 Day 7/Day 1 M/P Ratio: C.sub.max 0.017 0.056 0.099 0.316 0.035 0.015 0.022 0.053 (molar equivalents) M/P Ratio: AUC.sub.last NR NR 0.190 0.661 0.126 0.307 0.193 0.444 (molar equivalents)

TABLE-US-00062 TABLE 59 Plasma Concentrations of brincidofovir and cidofovir Following Single and Multiple 2-Hour Intravenous Infusion Administrations on Days 1, 3 and 7 at a Nominal brincidofovir Dose of 1 mg/kg to Female and Male Rats Sex Male Female Dose Time BCV cidofovir BCV cidofovir (mg/kg) Period (h) Subject Imputed Concentration.sup.a (ng/mL) 1 Day 1 1 2031 156 0.00 NS NS 2033 244 0.00 NS NS 2035 280 0.00 NS NS 2542 NS NS 109 0.00 2544 NS NS 126 0.00 2546 NS NS 177 0.00 Mean 227 0.00 137 0.00 SD 63.8 0.00 35.4 0.00 % CV 28.1 NC 25.8 NC N 3 3 3 3 1 Day 1 2 2032 220 0.00 NS NS 2034 283 0.00 NS NS 2036 203 0.00 NS NS 2543 NS NS 253 9.62 2547 NS NS 164 6.22 2572 NS NS 155 12.2 Mean 235 0.00 191 9.35 SD 42.1 0.00 54.2 3.00 % CV 17.9 NC 28.4 32.1 N 3 3 3 3 1 Day 1 8 2031 0.00 5.85 NS NS 2033 0.00 0.00 NS NS 2035 0.00 0.00 NS NS 2542 NS NS 2.30 7.12 2544 NS NS 1.45 5.60 2546 NS NS 0.00 0.00 Mean 0.00 1.95 1.25 4.24 SD 0.00 3.38 1.16 3.75 % CV NC 173 93.0 88.4 N 3 3 3 3 1 Day 1 24 2032 0.00 0.00 NS NS 2034 0.00 0.00 NS NS 2036 0.00 0.00 NS NS 2543 NS NS 0.00 0.00 2547 NS NS 0.00 0.00 2572 NS NS 0.00 0.00 Mean 0.00 0.00 0.00 0.00 SD 0.00 0.00 0.00 0.00 % CV NC NC NC NC N 3 3 3 3 1 Day 7 0 2031 0.00 0.00 NS NS 2033 0.00 0.00 NS NS 2035 0.00 0.00 NS NS 2542 NS NS 0.00 0.00 2544 NS NS 0.00 0.00 2546 NS NS 0.00 0.00 Mean 0.00 0.00 0.00 0.00 SD 0.00 0.00 0.00 0.00 % CV NC NC NC NC N 3 3 3 3 1 Day 7 2 2032 255 0.00 NS NS 2034 189 0.00 NS NS 2036 179 0.00 NS NS 2543 NS NS 115 13.5 2547 NS NS 82.1 13.8 2572 NS NS 89.2 17.6 Mean 208 0.00 95.4 15.0 SD 41.3 0.00 17.3 2.29 % CV 19.9 NC 18.1 15.3 N 3 3 3 3 1 Day 7 8 2031 0.00 5.67 NS NS 2033 0.00 5.45 NS NS 2035 0.00 6.07 NS NS 2542 NS NS 6.58 9.51 2544 NS NS 0.00 7.84 2546 NS NS 0.00 5.36 Mean 0.00 5.73 2.19 7.57 SD 0.00 0.314 3.80 2.09 % CV NC 5.49 173 27.6 N 3 3 3 3 1 Day 7 24 2032 0.00 0.00 NS NS 2034 0.00 0.00 NS NS 2036 0.00 0.00 NS NS 2543 NS NS 0.00 0.00 2547 NS NS 0.00 0.00 2572 NS NS 0.00 0.00 Mean 0.00 0.00 0.00 0.00 SD 0.00 0.00 0.00 0.00 % CV NC NC NC NC N 3 3 3 3 .sup.aImputations of zero were made for samples with bioanalytical result of BLQ.

TABLE-US-00063 TABLE 60 Plasma Concentrations of brincidofovir and cidofovir Following Single and Multiple 2-Hour Intravenous Infusion Administrations on Days 1, 3 and 7 at a Nominal brincidofovir Dose of 15 mg/kg to Female and Male Rats Sex Male Female Dose Time BCV cidofovir BCV cidofovir (mg/kg) Period (h) Subject Imputed Concentration.sup.a 15 Day 1 1 3053 4020 8.97 NS NS 3055 4030 0.00 NS NS 3057 5060 7.08 NS NS 3564 NS NS 3030 14.5 3566 NS NS 3930 20.5 3568 NS NS 3450 10.3 Mean 4370 5.35 3470 15.1 SD 598 4.73 450 5.13 % CV 13.7 88.4 13.0 33.9 N 3 3 3 3 15 Day 1 2 3054 2510 25.9 NS NS 3058 2990 13.1 NS NS 3074 3010 15.1 NS NS 3565 NS NS 2460 10.9 3567 NS NS 1860 41.2 3569 NS NS 3380 34.1 Mean 2840 18.0 2570 28.7 SD 283 6.89 766 15.8 % CV 9.98 38.2 29.8 55.2 N 3 3 3 3 15 Day 1 8 3053 10.7 35.8 NS NS 3055 8.23 29.5 NS NS 3057 9.85 33.0 NS NS 3564 NS NS 5.05 37.3 3566 NS NS 7.97 37.7 3568 NS NS 6.57 40.7 Mean 9.59 32.8 6.53 38.6 SD 1.25 3.16 1.46 1.86 % CV 13.1 9.63 22.4 4.82 N 3 3 3 3 15 Day 1 24 3054 1.20 20.5 NS NS 3058 1.10 14.0 NS NS 3074 1.36 15.5 NS NS 3565 NS NS 2.32 24.6 3567 NS NS 1.25 23.2 3569 NS NS 2.48 18.6 Mean 1.22 16.7 2.02 22.1 SD 0.131 3.40 0.669 3.14 % CV 10.7 20.4 33.2 14.2 N 3 3 3 3 15 Day 7 0 3053 0.00 0.00 NS NS 3055 0.00 0.00 NS NS 3057 0.00 0.00 NS NS 3564 NS NS 0.00 0.00 3566 NS NS 0.00 0.00 3568 NS NS 0.00 0.00 Mean 0.00 0.00 0.00 0.00 SD 0.00 0.00 0.00 0.00 % CV NC NC NC NC N 3 3 3 3 15 Day 7 2 3054 1640 38.8 NS NS 3058 1940 15.0 NS NS 3074 2150 18.3 NS NS 3565 NS NS 2220 20.2 3567 NS NS 995 49.1 3569 NS NS 1750 34.2 Mean 1910 24.0 1660 34.5 SD 256 12.9 618 14.5 % CV 13.4 53.7 37.3 41.9 N 3 3 3 3 15 Day 7 8 3053 9.82 38.4 NS NS 3055 7.60 31.1 NS NS 3057 8.73 28.8 NS NS 3564 NS NS 5.32 36.4 3566 NS NS 6.27 54.2 3568 NS NS 4.75 40.2 Mean 8.72 32.8 5.45 43.6 SD 1.11 5.01 0.768 9.37 % CV 12.7 15.3 14.1 21.5 N 3 3 3 3 15 Day 7 24 3054 1.85 27.2 NS NS 3058 1.32 17.9 NS NS 3074 1.17 19.9 NS NS 3565 NS NS 2.37 30.0 3567 NS NS 0.00 7.10 3569 NS NS 0.00 23.8 Mean 1.45 21.7 0.790 20.3 SD 0.357 4.90 1.37 11.8 % CV 24.7 22.6 173 58.3 N 3 3 3 3 .sup.aImputations of zero were made for samples with bioanalytical result of BLQ.

TABLE-US-00064 TABLE 61 Measured and Imputed brincidofovir Concentration Data for Mean Generation Period Dose Time Day 1 Day 7 Analyte (mg/kg) (h) Subject Sex C (ng/mL) IC (ng/mL) C (ng/mL) IC (ng/mL) BCV 0 2 1015 Male BLQ 0.00 BLQ 0.00 0 2 1016 Male BLQ 0.00 BLQ 0.00 0 2 1017 Male BLQ 0.00 BLQ 0.00 0 2 1523 Female BLQ 0.00 BLQ 0.00 0 2 1524 Female BLQ 0.00 BLQ 0.00 0 2 1525 Female BLQ 0.00 BLQ 0.00 1 0 2031 Male NS NS BLQ 0.00 1 0 2033 Male NS NS BLQ 0.00 1 0 2035 Male NS NS BLQ 0.00 1 0 2542 Female NS NS BLQ 0.00 1 0 2544 Female NS NS BLQ 0.00 1 0 2546 Female NS NS BLQ 0.00 1 1 2031 Male 156 156 NS NS 1 1 2033 Male 244 244 NS NS 1 1 2035 Male 280 280 NS NS 1 1 2542 Female 109 109 NS NS 1 1 2544 Female 126 126 NS NS 1 1 2546 Female 177 177 NS NS 1 2 2032 Male 220 220 255 255 1 2 2034 Male 283 283 189 189 1 2 2036 Male 203 203 179 179 1 2 2543 Female 253 253 115 115 1 2 2547 Female 164 164 82.1 82.1 1 2 2572 Female 155 155 89.2 89.2 1 8 2031 Male BLQ 0.00 BLQ 0.00 1 8 2033 Male BLQ 0.00 BLQ 0.00 1 8 2035 Male BLQ 0.00 BLQ 0.00 1 8 2542 Female 2.30 2.30 6.58 6.58 1 8 2544 Female 1.45 1.45 BLQ 0.00 1 8 2546 Female BLQ 0.00 BLQ 0.00 1 24 2032 Male BLQ 0.00 BLQ 0.00 1 24 2034 Male BLQ 0.00 BLQ 0.00 1 24 2036 Male BLQ 0.00 BLQ 0.00 1 24 2543 Female BLQ 0.00 BLQ 0.00 1 24 2547 Female BLQ 0.00 BLQ 0.00 1 24 2572 Female BLQ 0.00 BLQ 0.00 15 0 3053 Male NS NS BLQ 0.00 15 0 3055 Male NS NS BLQ 0.00 15 0 3057 Male NS NS BLQ 0.00 15 0 3564 Female NS NS BLQ 0.00 15 0 3566 Female NS NS BLQ 0.00 15 0 3568 Female NS NS BLQ 0.00 15 1 3053 Male 4020 4020 NS NS 15 1 3055 Male 4030 4030 NS NS 15 1 3057 Male 5060 5060 NS NS 15 1 3564 Female 3030 3030 NS NS 15 1 3566 Female 3930 3930 NS NS 15 1 3568 Female 3450 3450 NS NS 15 2 3054 Male 2510 2510 1640 1640 15 2 3058 Male 2990 2990 1940 1940 15 2 3074 Male 3010 3010 2150 2150 15 2 3565 Female 2460 2460 2220 2220 15 2 3567 Female 1860 1860 995 995 15 2 3569 Female 3380 3380 1750 1750 15 8 3053 Male 10.7 10.7 9.82 9.82 15 8 3055 Male 8.23 8.23 7.60 7.60 15 8 3057 Male 9.85 9.85 8.73 8.73 15 8 3564 Female 5.05 5.05 5.32 5.32 15 8 3566 Female 7.97 7.97 6.27 6.27 15 8 3568 Female 6.57 6.57 4.75 4.75 15 24 3054 Male 1.20 1.20 1.85 1.85 15 24 3058 Male 1.10 1.10 1.32 1.32 15 24 3074 Male 1.36 1.36 1.17 1.17 15 24 3565 Female 2.32 2.32 2.37 2.37 15 24 3567 Female 1.25 1.25 BLQ 0.00 15 24 3569 Female 2.48 2.48 BLQ 0.00

TABLE-US-00065 TABLE 62 Measured and Imputed Cidofovir Concentration Data for Mean Generation Period Dose_Level Time Day 1 Day 7 Analyte (mg/kg) (h) Subject Sex C (ng/mL) IC (ng/mL) C (ng/mL) IC (ng/mL) cidofovir 0 2 1015 Male BLQ 0.00 BLQ 0.00 0 2 1016 Male BLQ 0.00 BLQ 0.00 0 2 1017 Male BLQ 0.00 BLQ 0.00 0 2 1523 Female BLQ 0.00 BLQ 0.00 0 2 1524 Female BLQ 0.00 BLQ 0.00 0 2 1525 Female BLQ 0.00 BLQ 0.00 1 0 2031 Male NS NS BLQ 0.00 1 0 2033 Male NS NS BLQ 0.00 1 0 2035 Male NS NS BLQ 0.00 1 0 2542 Female NS NS BLQ 0.00 1 0 2544 Female NS NS BLQ 0.00 1 0 2546 Female NS NS BLQ 0.00 1 1 2031 Male BLQ 0.00 NS NS 1 1 2033 Male BLQ 0.00 NS NS 1 1 2035 Male BLQ 0.00 NS NS 1 1 2542 Female BLQ 0.00 NS NS 1 1 2544 Female BLQ 0.00 NS NS 1 1 2546 Female BLQ 0.00 NS NS 1 2 2032 Male BLQ 0.00 BLQ 0.00 1 2 2034 Male BLQ 0.00 BLQ 0.00 1 2 2036 Male BLQ 0.00 BLQ 0.00 1 2 2543 Female 9.62 9.62 13.5 13.5 1 2 2547 Female 6.22 6.22 13.8 13.8 1 2 2572 Female 12.2 12.2 17.6 17.6 1 8 2031 Male 5.85 5.85 5.67 5.67 1 8 2033 Male BLQ 0.00 5.45 5.45 1 8 2035 Male BLQ 0.00 6.07 6.07 1 8 2542 Female 7.12 7.12 9.51 9.51 1 8 2544 Female 5.60 5.60 7.84 7.84 1 8 2546 Female BLQ 0.00 5.36 5.36 1 24 2032 Male BLQ 0.00 BLQ 0.00 1 24 2034 Male BLQ 0.00 BLQ 0.00 1 24 2036 Male BLQ 0.00 BLQ 0.00 1 24 2543 Female BLQ 0.00 BLQ 0.00 1 24 2547 Female BLQ 0.00 BLQ 0.00 1 24 2572 Female BLQ 0.00 BLQ 0.00 15 0 3053 Male NS NS BLQ 0.00 15 0 3055 Male NS NS BLQ 0.00 15 0 3057 Male NS NS BLQ 0.00 15 0 3564 Female NS NS BLQ 0.00 15 0 3566 Female NS NS BLQ 0.00 15 0 3568 Female NS NS BLQ 0.00 15 1 3053 Male 8.97 8.97 NS NS 15 1 3055 Male BLQ 0.00 NS NS 15 1 3057 Male 7.08 7.08 NS NS 15 1 3564 Female 14.5 14.5 NS NS 15 1 3566 Female 20.5 20.5 NS NS 15 1 3568 Female 10.3 10.3 NS NS 15 2 3054 Male 25.9 25.9 38.8 38.8 15 2 3058 Male 13.1 13.1 15.0 15.0 15 2 3074 Male 15.1 15.1 18.3 18.3 15 2 3565 Female 10.9 10.9 20.2 20.2 15 2 3567 Female 41.2 41.2 49.1 49.1 15 2 3569 Female 34.1 34.1 34.2 34.2 15 8 3053 Male 35.8 35.8 38.4 38.4 15 8 3055 Male 29.5 29.5 31.1 31.1 15 8 3057 Male 33.0 33.0 28.8 28.8 15 8 3564 Female 37.3 37.3 36.4 36.4 15 8 3566 Female 37.7 37.7 54.2 54.2 15 8 3568 Female 40.7 40.7 40.2 40.2 15 24 3054 Male 20.5 20.5 27.2 27.2 15 24 3058 Male 14.0 14.0 17.9 17.9 15 24 3074 Male 15.5 15.5 19.9 19.9 15 24 3565 Female 24.6 24.6 30.0 30.0 15 24 3567 Female 23.2 23.2 7.10 7.10 15 24 3569 Female 18.6 18.6 23.8 23.8

TABLE-US-00066 TABLE 63 Mean and Imputed brincidofovir Concentration Data for NCA Period Day 1 Day 7 Dose Time MC IMC MC IMC Analyte (mg/kg) Sex (h) (ng/mL) (ng/mL) (ng/mL) (ng/mL) BCV 1 Female 0 NS NS 0.00 0.00 1 Female 1 137 137 NS NS 1 Female 2 191 191 95.4 95.4 1 Female 8 1.25 1.25 2.19 2.19 1 Female 24 0.00 missing 0.00 missing 1 Male 0 NS NS 0.00 0.00 1 Male 1 227 227 NS NS 1 Male 2 235 235 208 208 1 Male 8 0.00 missing 0.00 missing 1 Male 24 0.00 missing 0.00 missing 15 Female 0 NS NS 0.00 0.00 15 Female 1 3470 3470 NS NS 15 Female 2 2570 2570 1660 1660 15 Female 8 6.53 6.53 5.45 5.45 15 Female 24 2.02 2.02 0.790 0.790 15 Male 0 NS NS 0.00 0.00 15 Male 1 4370 4370 NS NS 15 Male 2 2840 2840 1910 1910 15 Male 8 9.59 9.59 8.72 8.72 15 Male 24 1.22 1.22 1.45 1.45

TABLE-US-00067 TABLE 64 Mean and Imputed cidofovir Concentration Data for NCA Period Day 1 Day 7 Dose Time MC IMC MC IMC Analyte (mg/kg) Sex (h) (ng/mL) (ng/mL) (ng/mL) (ng/mL) cidofovir 1 Female 0 NS NS 0.00 0.00 1 Female 1 0.00 0.00 NS NS 1 Female 2 9.35 9.35 15.0 15.0 1 Female 8 4.24 4.24 7.57 7.57 1 Female 24 0.00 missing 0.00 missing 1 Male 0 NS NS 0.00 0.00 1 Male 1 0.00 0.00 NS NS 1 Male 2 0.00 0.00 0.00 0.00 1 Male 8 1.95 1.95 5.73 5.73 1 Male 24 0.00 missing 0.00 missing 15 Female 0 NS NS 0.00 0.00 15 Female 1 15.1 15.1 NS NS 15 Female 2 28.7 28.7 34.5 34.5 15 Female 8 38.6 38.6 43.6 43.6 15 Female 24 22.1 22.1 20.3 20.3 15 Male 0 NS NS 0.00 0.00 15 Male 1 5.35 5.35 NS NS 15 Male 2 18.0 18.0 24.0 24.0 15 Male 8 32.8 32.8 32.8 32.8 15 Male 24 16.7 16.7 21.7 21.7

Example 9--Mass Balance, Pharmacokinetics and Tissue Distribution by Quantitative Whole-Body Autoradiography in Rats Following a Single Oral or Intravenous Dose of [.sup.14C]Brincidofovir

[0560] The objectives of this study were to characterize the tissue distribution of total radioactivity in male Sprague Dawley (SD) and Long-Evans (LE) rats following administration of a single intravenous (IV, 2-h infusion) or oral (PO, gavage) dose of [.sup.14C]brincidofovir. In addition, the rate and extent of excretion (mass balance) and pharmacokinetics (PK) of total radioactivity in male Sprague Dawley rats following a single intravenous (IV, 2-h infusion) dose of [.sup.14C]brincidofovir was examined. Residual plasma and excreta collected on this study were stored at -70.degree. C. for metabolite profile and identification experiment conducted under a different protocol.

Materials and Methods

[0561] The study utilized 4 groups of male SD rats (albino), and 3 groups of male LE rats (pigmented); a total of 39 rats. All animals were obtained from Hilltop Lab Animals, Inc. (Scottdale, Pa.). Rats weighed between 214 and 265 g at dosing. Rats in Groups 1, 2, 3, and 4 were administered a single 2-hour (h) IV infusion of [.sup.14C]brincidofovir at a target dose of 15 mg/kg. Rats in Group 5 were administered a single 2-h IV infusion of [.sup.14C]brincidofovir at a target dose of 2 mg/kg. Rats in Group 6, and 7 were administered a single oral gavage administration of [.sup.14C]brincidofovir at a target dose of 15 mg/kg. Three dosing formulations (one for high dose IV, one for low dose IV, and one for PO dose) were prepared at QPS on the day of dosing. The IV formulation for Groups 1, 2, 3, and 4 contained a vehicle of 10 mM sodium phosphate buffer in 5% dextrose solution at pH 8.0(.+-.0.1). The IV formulation for Group 5 contained a vehicle of 16 mM sodium phosphate buffer in 5% dextrose solution at pH 8.0(.+-.0.1), and the PO dosing formulation contained a vehicle of 12.5 mM sodium phosphate buffer at pH 8.0(.+-.0.1). The IV dose formulation for Group 5 and the PO dose formulation for Groups 6 and 7 contained higher concentration of buffer so that the final buffer concentration was approximately 10 mM in all dose formulations.

Test Animals

[0562] Sixteen adult male SD rats, and 23 adult male LE rats, which were obtained from Hilltop Lab Animals, Inc. (Scottdale, Pa.), were placed on this study. Rats in Groups 1-5 were surgically modified to have indwelling femoral vein cannulas (FVC) for IV infusion. The body weight range, source, vendor, and receipt date of the animals was documented in the raw data. Animals placed on study were assigned a permanent identification number using a permanent marker on the tail, while unused spare animals were returned to stock after successful dose administration of animals placed on study. Randomization was performed by cannula patency and QPS SOP.

[0563] Tables 65 and 66 give summaries of the dosing and sample collection protocols.

TABLE-US-00068 TABLE 65 In-Life Study Dosing Summary No. of Target Dose Target Target Dose Target Dose Target Dose Group Strain/Dose Animals, Level Radioactivity Date Volume Conc. No. Study route Sex (mg/kg) Level (.mu.Ci/kg) (mL/kg/g).sup.b (mL/kg) (mg/mL) 1 MB SD/IV.sup.a 3M 15 mg/kg 180 10 20 0.75 mg/mL 2 PK SD/IV.sup.a 7M 15 mg/kg 180 10 20 0.75 mg/mL 3 QWBA SD/IV.sup.a 3M 15 mg/kg 180 10 20 0.75 mg/mL 4 QWBA LE/IV.sup.a 9M 15 mg/kg 180 10 20 0.75 mg/mL 5 QWBA LE/IV.sup.a 4M 2 mg/kg 200 1.33 2.66 0.75 mg/mL 6 QWBA SD/PO 3M 15 mg/kg 200 -- 5 3 mg/mL 7 QWBA LE/PO 10M 15 mg/kg 200 -- 5 3 mg/mL .sup.aFemoral vein-cannulated; .sup.b2-h infusion time

TABLE-US-00069 TABLE 66 In-Life Sample Collection Summary Purpose, Group ROA, Cage Terminal Blood Number Strain Urine.sup.a Feces.sup.a Residues Carcass and Plasma 1 MB, IV, SD Pre-dose, 0-8, Pre-dose, 0- Daily Cage Frozen NA 8-24, and at 24 and at 24 Rinse following 24 h intervals h intervals beginning at euthanasia and through 168 h through 168 24 h, and retained for post-dose; h post-dose; cage wash potential N = 3 N = 3 and wipes at analysis termination; N = 3 2 PK, IV, SD NA NA NA NA At 0.5, 1, 2 (end of infusion), 4, 8, 24, and 72 h; N = 1/time point 3 QWBA, NA NA NA At 2 (end of At 2 IV, SD infusion), 24, (end of infusion), and 168 h; 24, and 168 h; N = 1/time point N = 1/time point 4 QWBA, NA NA NA At 1, 2 (end of At 1 ,2 IV, LE infusion), 4, 8, (end of infusion), 24, 72, 96, 168 4, 8, 24, 72, 96, h and TBD; 168 and 840 h; N = 1/time point N = 1/time point 5 QWBA, NA NA NA At 2 (end of At 2 IV, LE infusion), 8, 24, (end of infusion), and 72 h 8, 24, and 72 h 6 QWBA, NA NA NA At 2, 24, and At 2, 24, and PO, SD 168 h post- 168 h post-dose; dose; N = 1/time N = 1/time point point 7 QWBA, NA NA NA At 0.5, 1,2, 4, 8, At 0.5, 1, 2, 4, 8, PO, LE 24, 72, 96, 168 24, 72, 96, 168 h and TBD; and 840 h; N = 1/time point N = 1/time point ROA = Route of Administration NA = Not Applicable TBD = To be Determined .sup.aSamples collected over dry ice

Dose Administration

[0564] Animal body weights, which were used for determining the dosing volume, were measured before dosing on the day of dose administration. Dose assay results, animal body weights, and targeted dosing parameters were entered into the Debra LIMS and target dose volumes for each animal were determined by the Debra LIMS. Animals received a single 2-h IV infusion of [.sup.14C]brincidofovir at a target oral dose of 15 mg/kg (Groups 1-4) or 2 mg/kg (Group 5), or an oral gavage administration of [.sup.14C]brincidofovir at a target oral dose of 15 mg/kg (Groups 6 and 7). The actual dose administered to each rat was determined by the Debra LIMS, which used individual animal body weight data and by subtracting the weight of the emptied dose syringe/needle or needle/infusion line after dosing from the weight of the full syringe/needle or needle/infusion line prior to dosing. The mean pre-dose radioactivity concentration (dpm/g dosing solution) was multiplied by the net weight of the administered dosing solution to calculate the amount of radioactivity administered to each animal. Actual doses are presented in Table 67.

TABLE-US-00070 TABLE 67 Body Weights and Doses of [.sup.14C]brincidofovir Administered to Male Sprague Dawley Rats (Groups 1, 2, 3, and 6), and Male Long-Evans Rats (Groups 4, 5, and 7) Amount of mg Test Animal Animal Formulation .mu.Ci Article mg/kg No. Weight (g) Administered (g) Administered .mu.Ci/kg Administered.sup.a,b Administered.sup.a,b Gp. 1, Mass Balance, Male SD Rat, 2-h IV, 15 mg/kg 1 251.2 5.0069 50.607 201.459 3.729 14.847 2 250.2 5.0375 50.916 203.501 3.752 14.997 3 261.9 5.2909 53.477 204.189 3.941 15.048 MEAN 254.4 5.1120 51.667 203.050 3.807 14.964 SD 6.5 0.1560 1.575 1.420 0.116 0.104 Gp. 2, PK, Male SD Rats, 2-h IV, 15 mg/kg 4c 251.8 1.3636 13.782 54.736 1.016 4.034 5c 250.0 2.4727 24.992 99.970 1.842 7.367 6 242.3 4.7611 48.122 198.606 3.546 14.636 7 249.9 5.1748 52.304 209.298 3.855 15.424 8 261.6 5.2120 52.680 201.375 3.882 14.840 9 264.6 5.2057 52.616 198.851 3.878 14.654 10 254.0 5.0652 51.196 201.558 3.773 14.854 MEAN 254.5 5.084 51.384 201.938 3.787 14.882 SD 9.0 0.190 1.918 4.338 0.142 0.320 Gp. 3, QWBA, Male SD Rat, 2-h IV, 15 mg/kg 11 247.3 4.9350 49.880 201.698 3.676 14.864 12 256.6 5.0999 51.547 200.883 3.799 14.804 13 248.4 4.8006 48.521 195.336 3.576 14.395 MEAN 250.8 4.945 49.983 199.306 3.684 14.688 SD 5.1 0.150 1.516 3.462 0.112 0.255 Gp. 4, QWBA, Male LE Rats, 2-h IV 15 mg/kg 14c 240.5 2.3933 24.190 100.582 1.783 7.412 15 232.1 4.5816 46.308 199.517 3.413 14.703 16 234.9 4.6662 47.163 200.779 3.476 14.796 17 236.8 4.7293 47.801 201.861 3.523 14.876 18 214.4 4.2683 43.141 201.219 3.179 14.829 19 247.3 4.9154 49.682 200.897 3.661 14.805 20 237.9 4.7154 47.660 200.337 3.512 14.764 21 242.6 4.7892 48.406 199.531 3.567 14.705 22 235.0 4.5292 45.778 194.801 3.374 14.356 MEAN 235.1 4.649 46.992 199.868.sup.d 3.463 14.729 SD 9.7 0.195 1.969 2.198 0.145 0.162 Gp. 5, QWBA, Male LE Rat, 2-h IV 2 mg/kg 23 231.9 0.6395 52.998 228.538 0.495 2.133 24 244.6 0.6611 54.788 223.990 0.511 2.090 25 231.8 0.6303 52.235 225.347 0.487 2.103 26 243.5 0.7172 59.437 244.095 0.555 2.278 MEAN 238.0 0.662 54.865 230.493 0.512 2.151 SD 7.1 0.039 3.231 9.267 0.030 0.087 Gp. 6, QWBA, Male SD Rats, PO, 15 mg/kg 27 250.7 1.2270 51.798 206.613 3.630 14.480 28 231.2 1.1460 48.379 209.250 3.390 14.665 29 254.8 1.3077 55.205 216.659 3.869 15.184 MEAN 245.6 1.227 51.794 210.841 3.630 14.776 SD 12.6 0.081 3.413 5.208 0.240 0.365 Gp. 7, QWBA, Male LE Rats, PO, 15 mg/kg 30 229.4 1.1586 48.910 213.210 3.428 14.942 31 236.1 1.1913 50.291 213.007 3.525 14.928 32 254.6 1.2685 53.550 210.330 3.753 14.740 33 234.1 1.1959 50.485 215.656 3.538 15.114 34 223.1 1.1223 47.378 212.362 3.320 14.883 35 246.6 1.1773 49.700 201.541 3.483 14.124 36 231.3 1.1473 48.433 209.397 3.394 14.675 37 257.7 1.2626 53.301 206.833 3.735 14.495 38 234.7 1.2077 50.983 217.227 3.573 15.224 39 238.5 1.2192 51.469 215.802 3.607 15.124 MEAN 238.6 1.195 50.450 211.537 3.536 14.825 SD 11.1 0.047 1.987 4.729 0.139 0.332 .sup.aSpecific activity of the oral (PO) formulation was 14.2689 .mu.Ci/mg [.sup.14C]brincidofovir. Dose preparation activity of the PO formulation was 42.2151 .mu.Ci/g .sup.bSpecific activity of the intravenous (IV) infusion formulations were 13.5694 (at 15 mg/kg) and 101.5100 (at 2 mg/kg) .mu.Ci/mg [.sup.14C]brincidofovir. Dose preparation activity of the IV formulations were 10.1074 .mu.Ci/g (at 15 mg/kg) and 82.8740 .mu.Ci/g (at 2 mg/kg) .sup.cAnimal terminated during IV infusion for sample collection. These animals were omitted from the mean dose calculations. .sup.dThe mean for all animals dosed in Group 4, 188.836 .mu.Ci/kg was used for dosimetry estimate. Note: SD = Standard deviation; MB = Mass Balance; PK = pharmacokinetics; QWBA = Quantitative Whole-Body Autoradiography

Sample Collection

[0565] Urine, feces, blood/plasma, cage residue samples, and carcasses were collected during this study as described below for each group

Excreta Collection (Group 1)

[0566] Three male albino rats were assigned to Group 1 (IV infusion) for excreta collection.

[0567] Urine samples were collected from each animal into pre-labeled urine collection tubes at pre-dose (overnight) and at intervals of 0-8 h, 8-24 h, and at every subsequent 24 h interval until 168 h post-dose. All urine specimens were collected over dry ice. The total weight of each urine collection was documented, and the samples were stored frozen at approximately -70.degree. C. until LSC analysis. Samples were maintained at approximately -70.degree. C. following analysis, and were saved for further analysis, to be conducted under a separate protocol.

[0568] Feces samples were collected from each animal into a pre-labeled feces collection tube at pre-dose (overnight) and at 24 h intervals post-dose until 168 h post-dose. Feces specimens were collected over dry ice and the total weight of each feces sample was documented. Feces samples were stored at approximately -70.degree. C. until homogenization and LSC analysis, were maintained at approximately -70.degree. C. following analysis, and were saved for further analysis, to be conducted under a separate protocol.

[0569] Cage residue specimens were collected. Cages were rinsed with approximately 30 mL of deionized water following each daily post-dose excreta collection beginning at 24 h post-dose. The interior surfaces of the metabolism cage were sprayed with approximately 90 mL of Windex solution (or equivalent detergent solution) and wiped with gauze pads following the final excreta collection. Cage rinse and cage wash specimens were collected into tared and pre-labeled containers. The total weight of each cage rinse and wash were documented. Cage rinse, cage wash, and cage wipe collections were stored at approximately -20.degree. C. Carcasses of animals in Groups 1 and 2 were retained at approximately -20.degree. C. Carcasses and cage residues were not analyzed because the recovery of radioactivity in excreta was >90%.

Plasma Pharmacokinetic Sample Collection (Group 2)

[0570] Blood was collected from each animal in Group 2 (IV infusion) for plasma and blood analysis at the times listed below.

[0571] Terminal blood samples were collected by cardiac puncture into pre-labeled blood collection tubes that contained K.sub.2EDTA as an anticoagulant. A terminal sample (N=1 per time point) was collected at 0.5, 1, 2, 4, 8, 24, and 72 h after initiation of infusion and the volume was not limited. Triplicate weighed aliquots of blood (.about.0.100 g) were removed and analyzed for total radioactivity using combustion followed by LSC. Blood aliquots were maintained at approximately 4.degree. C. until analysis. The remaining blood samples were maintained on wet ice (approximately 4.degree. C.) and then centrifuged to obtain plasma within 1 h of the blood collection time. Centrifuge settings were recorded in the study notebook. Duplicate aliquots of plasma (0.050 mL) were analyzed for radioactivity using direct counting by LSC, and residual plasma was stored at approximately -70.degree. C. and was saved for possible future analysis under a separate study protocol. Residual red blood cells and the animal carcasses of Group 2 rats were discarded as radioactive waste.

QWBA (Groups 3-7)

[0572] Each animal, at their respective termination time points, was euthanized for QWBA analysis immediately after blood collection. One rat per time point per Group was euthanized as listed below.

Group 3 (IV)--2 (end of infusion), 24, and 168 h Group 4 (IV)--1, 2 (end of infusion), 4, 8, 24, 72, 96, 168, and 840 h Group 5 (IV)--2 (end of infusion), 8, 24, and 72 h

Group 6 (PO)--2, 24, and 168 h

Group 7 (PO)--0.5, 1, 2, 4, 8, 24, 72, 96, 168, and 840 h

[0573] Each rat was deeply anesthetized via isoflurane anesthesia, a blood sample was collected by cardiocentesis (approximately 2 mL) into tubes that contained K.sub.2EDTA as the anticoagulant, and the rat was euthanized by being frozen in a hexane/solid carbon dioxide bath for at least 15 min. The blood samples were maintained on wet ice (approximately 4.degree. C.) and then centrifuged to obtain plasma within 1 h of the blood collection time. Centrifuge settings were recorded in the study notebook. Duplicate aliquots of plasma (0.050 mL) were analyzed for radioactivity using direct counting by LSC, and residual plasma was stored at approximately -70.degree. C. and was saved for possible future analysis under a separate study protocol. Residual red blood cells were discarded as radioactive waste.

Sample Storage Conditions

[0574] Carcass and cage residue samples were stored frozen at -20.degree. C. until sample analysis, and residual plasma and excreta were stored frozen at -70.degree. C.

Sample Analysis

LSC Analysis of Blood, Plasma, and Excreta

[0575] Feces were homogenized and analyzed for total radioactivity content. Weighed feces specimens for each rat were homogenized in approximately 3 volumes of water (approximately 3.times. the weight of the feces specimen). The total weight of each homogenate was determined and triplicate weighed aliquots (.about.0.5 g) were combusted in a Packard Sample Oxidizer, followed by LSC analysis. The actual weights of the individual aliquots and the amount of solvent used in homogenization were recorded. Pre-weighed portions of each feces homogenate were placed into a CombustoCone.RTM. that contained a CombustoPad.RTM., allowed to dry over-night in a fume hood, and were burned completely in the sample oxidizer.

[0576] Triplicate weighed aliquots (0.100 g) of blood aliquots (Group 2 only) were combusted in the Packard Sample Oxidizer, followed by LSC analysis.

[0577] The .sup.14CO2 liberated in combustion was trapped in a solution of Carbo-Sorb.RTM., Permafluor.RTM. (scintillation fluid) was added, and the radioactivity was determined by LSC. Blank cones that contained a known amount of radioactivity (0.100 mL SpecChec.RTM.) were oxidized and compared to non-combusted standards to determine the percent recovery from the combustion process. These controls were analyzed in triplicate, and quantitative recovery of .sup.14C from these control samples was determined on each day of sample combustion. Blank cones (without added radioactivity) were burned to evaluate the radioactivity carryover between samples.

[0578] Fecal homogenates were maintained at approximately -70.degree. C. after analysis and were saved for further analysis under a separate study protocol.

[0579] Urine (0.300 mL), and plasma (0.050 mL) specimens were thawed, if necessary, then aliquoted in duplicate by volume and analyzed by LSC. Volumes or weights of sample aliquots were documented in the study records. Ultima Gold scintillation fluid (5 mL, PerkinElmer) was added to each urine, and plasma aliquot, aliquot were mixed thoroughly, and then analyzed by LSC for radioactivity.

[0580] The radioactivity (counts per minute) in each sample was converted to disintegrations per minute (dpm) by means of an external standardization and a quench curve. Radioactivity content was quantified by a Model 2800TR or Model 2900TR Liquid Scintillation Analyzer (PerkinElmer). All samples were counted for at least 5 minutes or at least 100,000 counts per minute (cpm). LSC results for duplicate samples that differed by more than 10% from the mean value, were re-aliquoted and re-analyzed, if sufficient volume was available. If the LSC results for triplicate samples had a % CV that was >10%, then the sample was re-homogenized and re-analyzed, if sufficient volume was available. These specifications were applied to all sample aliquots that had an average radioactivity greater than 500 dpm, and values that were under 500 dpm were accepted as is. A lower limit of quantification (LLOQ) was applied to the data equal to 2 times the counts per minute (CPM) value of a background sample.

QWBA

[0581] The pinna, distal limbs, hair, and tail were removed from each frozen carcass and each frozen carcass was embedded in an aqueous suspension of approximately 2% (w/v) carboxymethylcellulose and frozen into a block. The blocks were stored at approximately -20.degree. C. prior to sectioning. Each blocked carcass was mounted on the object stage of a cryomicrotome (Leica CM3600 Cryomacrocut, Nussloch, Germany and Vibratome 9800, St. Louis, Mo.) maintained at approximately -20.degree. C. and internal standards (3), which were plasma fortified with [.sup.14C]-glucose at one concentration (approximately 0.05 .mu.Ci/mL), were placed into the frozen blocks, prior to sectioning, and were used for section thickness quality control. A number of whole-body sections (approximately 40 .mu.m thick) were taken in the sagittal plane at various levels of interest using a whole-body cryomicrotome set at -20.degree. C. Sagittal sections (20-50 .mu.m thick) of frozen carcasses were exposed to phosphor imaging plates. Regions of images in tissues were converted to concentrations using standards. All of the major tissues, organs, and biological fluids were represented. The sections were collected on adhesive tape (Scotch 8210, 3M Corp., USA) and dehydrated in the cryomicrotome for at least 48 h prior to removal for mounting and exposure.

[0582] Tissue concentration data was determined for the following tissues and/or contents: adipose (brown and white), adrenal gland, bile (in duct), blood (cardiac), bone, bone marrow, brain (cerebrum, cerebellum, medulla), cecum (and contents), large intestine (and contents), epididymis, esophagus, eye (uvea and lens), Harderian gland, heart, kidney (cortex and medulla), liver, lung, lymph node, pancreas, pituitary gland, prostate gland, salivary gland, seminal vesicles, skeletal muscle, skin (pigmented and non-pigmented), small intestine (and contents), stomach (gastric mucosa and contents), spleen, spinal cord, testis, thymus, thyroid, and urinary bladder (and contents).

[0583] A set of whole-body sections for each rat was mounted on a cardboard backing, covered with a thin plastic wrap, and exposed along with calibration standards, which were .sup.14C-glucose mixed with blood at 10 different concentrations (0.0009595 to 7.806 .mu.Ci/g), to a .sup.14C-sensitive phosphor imaging plate (Fuji Biomedical, Stamford, Conn.). The imaging plate and sections were placed in light-tight exposure cassettes, in a copper-lined lead safe, for a 4-day exposure at room temperature. The imaging plate was scanned using the Typhoon 9410 image acquisition system (GE/Molecular Dynamics, Sunnyvale, Calif., USA) and the resultant image stored on a dedicated QPS computer server. Quantification was performed by image densitometry using MCD image analysis software (v. 7.0, Interfocus Imaging Ltd) and a standard curve constructed from the integrated response [i.e., Molecular Dynamics Counts per square millimeter (MDC/mm.sup.2)] and the nominal concentrations of the .sup.14C-calibration standards. The concentrations of radioactivity were expressed as the .mu.g equivalents of [.sup.14C]brincidofovir per gram sample (.mu.g equiv/g). A lower limit of quantification (LLOQ) was applied to the data (See section 6.7.4).

[0584] Tissue concentrations that fell below the LLOQ were identified as being below the quantification limit (BQL). Tissue areas that were not visualized on autoradiographic images were identified as no sample (NS) and reported as BQL. If no tissues were visualized on the autoradiograph(s) for an animal, then no calibration curves were generated and all tissue concentrations for that animal were reported as BQL.

[0585] The results are based on original, electronic, digital images that were selected from a complete set of autoradiographs.

Animal Observations

[0586] No observable abnormalities were observed in the rats on this study.

Group 1--Mass Balance in Rats after a 2-H Intravenous Infusion

[0587] A summary of the individual animal and mean excretion data for total radioactivity in Group 1 male SD rats is presented in Table 68.

TABLE-US-00071 TABLE 68 Excretion of Total Radioactivity Following an 2-Hour Intravenous Infusion of [.sup.14C]brincidofovir to Male Sprague Dawley Rats at a Target Dose of 15 mg/kg (Group 1) Percent Recovery (% Dose) Rat Numbers Sample Time Point 01 02 03 Mean SD Urine Pre-dose 0.0 0.0 0.0 0.0 0.0 0-8 h 33.0 29.3 30.7 31.0 1.9 8-24 h 10.7 11.5 16.2 12.8 3.0 24-48 h 4.3 3.6 4.5 4.1 0.5 48-72 h 1.6 1.6 1.9 1.7 0.2 72-96 h 0.8 0.8 0.8 0.8 0.0 96-120 h 0.4 0.3 0.5 0.4 0.1 120-144 h 0.2 0.2 0.3 0.2 0.0 144-168 h 0.2 0.1 0.2 0.2 0.0 Subtotal 51.2 47.5 55.1 51.2 3.8 Feces Pre-dose 0.0 0.0 0.0 0.0 0.0 0-24 h 37.3 37.6 35.1 36.7 1.4 24-48 h 3.4 4.6 3.5 3.8 0.7 48-72 h 0.9 1.0 0.8 0.9 0.1 72-96 h 0.4 0.4 0.4 0.4 0.0 96-120 h 0.2 0.2 0.2 0.2 0.0 120-144 h 0.1 0.1 0.1 0.1 0.0 144-168 h 0.1 0.1 0.1 0.1 0.0 Subtotal 42.4 44.2 40.2 42.2 2.0 Total 93.6 91.6 95.2 93.5 1.8 SD = Standard deviation;

[0588] The primary route of elimination of radioactivity after a 2-h IV infusion administration of 15 mg/kg [.sup.14C]brincidofovir to male rats was in the urine, which accounted for an average of 51.2% of the administered dose over a 168 h period. An average of 42.2% of the administered dose was recovered in feces. The total recovery of radioactivity in urine and feces combined in Group 1 male rats averaged 93.5% of the dose over the 168 h collection period. Most of the urinary excretion (43.8% of the dose or 86% of the radioactivity of the radioactivity excreted in the urine) and fecal excretion (36.7% of the dose or 87% of the radioactivity excreted in the feces) of radioactivity occurred in the first 24 h after dosing.

Group 2--Plasma Pharmacokinetics in Rats after IV Administration of 15 mg/kg

[0589] The plasma total radioactivity concentration versus time data, PK parameters, and blood to plasma ratios for Group 2 male SD rats are reported in Table 69.

TABLE-US-00072 TABLE 69 Blood and Plasma Concentration and Pharmacokinetic Parameters of Male Sprague Dawley Rats After a Single 2-Hour Intravenous Infusion of 15 mg/kg of [.sup.14C]brincidofovir (Group 2) Plasma Blood Blood to Time Point Concentration Concentration Plasma (h) (.mu.g/mL) (.mu.g equiv/mL) Ratio 0.5 2.557 1.666 0.65 1 4.609 3.017 0.65 2 10.297 6.393 0.62 4 5.363 3.845 0.72 8 1.961 1.584 0.81 24 0.234 0.294 1.26 72 0.045 0.057 1.27 t.sub.1/2 (h) 13.0 14.5 T.sub.max (h) 2 2 C.sub.max (.mu.g equiv/mL) 10.297 6.393 AUC.sub.last (.mu.g equiv- 64.448 50.836 h/mL) AU.sub.Call (.mu.g equiv- 64.448 50.836 h/mL) AUC.sub.inf.sub.--.sub.obs (.mu.g 65.292 52.028 equiv-h/mL)

[0590] The C.sub.max of [.sup.14C]brincidofovir total radioactivity in plasma after a 2-h IV infusion administration to male rats at 15 mg/kg was 10.3 .mu.g equiv/mL at a T.sub.max of 2 h (i.e., end of infusion), and the concentration decreased to 0.045 .mu.g equiv/mL at 72 h post-dose. The AUC.sub.last of [.sup.14C]brincidofovir in plasma was 64.4 .mu.g equiv. h/mL, and the t.sub.1/2 was 13.0 h.

[0591] Blood to plasma ratios for Group 2 SD rats showed that [.sup.14C]brincidofovir-derived radioactivity initially partitioned preferentially into plasma versus the cellular portion of blood of rats at 0. 5 to 8 h (ranged between 0. 62-0.8 l), but then showed more partitioning into the cellular aspects at 24 and 72 h post-dose with a ratios of 1.26 and 1.27, respectively.

Groups 3-7--Comparison of blood to plasma concentration ratios in albino and pigmented rats after IV and PO administration of 15 mg/kg (IV and PO) and 2 mg/kg (IV)

[0592] The blood to plasma concentration ratios for Group 3 to Group 7 male SD and LE rats are reported in Table 70. A comparison of blood and plasma concentration versus time profiles obtained from Groups 3-7 is presented in FIG. 3.

TABLE-US-00073 TABLE 70 Blood to Plasma Ratios in Male Rats After a Single 2-Hour Intravenous Infusion or Oral Gavage Administration of [.sup.14C]brincidofovir (Groups 3-7) Blood:Plasma Ratio Group/ Dose Dose Time After Dose Initiation (h) Strain Route (mg/kg) 0.5 1 2 4 8 24 72 3/SD IV 15 NS NS 0.96 NS NS 1.23 NS 4/LE IV 15 NS 1.10 1.08 0.92 1.39 1.46 3.06 5/LE IV 2 NS NS 0.89 NS 1.03 1.41 1.25 6/SD PO 15 NS NS 0.64 NS NS 0.78 NS 7/LE PO 15 ND 1.13 0.98 0.80 0.76 0.87 ND ND = not determined, blood and/or plasma BQL NS = no sample collected at this time point

[0593] Similar to Group 2, trends for higher blood:plasma ratios at later time-points were observed in other groups of animals administered IV [.sup.14C]brincidofovir (SD rats, Group 3; LE rats, Groups 4 and 5), though ratios at early (sparse) time-points were approximately unity. Following oral administration of [.sup.14C]brincidofovir, blood:plasma ratios ranged between 0.64-1.13, and no trends for higher blood:plasma ratios at later time-points (up to 24 h after dosing) were observed.

Group 3 and 4--QWBA Tissue Distribution in Albino and Pigmented Male Rats after IV Administration of 15 mg/kg

[0594] A summary of tissue distribution of [.sup.14C]brincidofovir in male albino (Group 3; n=3) and pigmented rats (Group 4; n=9) are presented in Table 71 and Table 72, respectively.

TABLE-US-00074 TABLE 71 Concentrations of Radioactivity in Tissues of Male Sprague Dawley (Albino, Group 3) Rats After a Single 2-Hour Intravenous Infusion of [.sup.14C]brincidofovir at a Target Dose of 15 mg/kg Concentration (.mu.g equiv./g) Rat# 11 Rat# 12 Rat# 13 Tissue Type Tissue 2 h 24 h 168 h Vascular/ Plasma (by LSC) 7.130 0.251 BQL Lymphatic Blood (cardiac) 6.865 0.308 BQL Bone Marrow 3.411 1.219 0.196 Lymph Node 4.055 1.004 0.247 Spleen 4.536 2.244 0.694 Thymus 2.373 0.556 0.105 Excretory/ Bile (in duct) NI 1.469 0.058 Metabolic Kidney Cortex 106.513 32.109 0.474 Kidney Medulla 33.444 4.271 0.186 Liver 229.371 13.907 0.453 Urinary Bladder 35.978 2.264 0.132 Urinary Bladder 47.034 21.237 0.121 (contents) Central Brain (cerebellum) 0.907 0.161 BQL Nervous Brain (cerebrum) 0.803 0.107 BQL System Brain (medulla) 0.864 0.132 BQL Spinal Cord 0.437 0.079 BQL Endocrine Adrenal Gland 4.308 1.680 0.647 Pituitary Gland 2.793 1.021 0.257 Thyroid 2.751 0.791 0.239 Secretory Harderian Gland 2.081 1.278 0.954 Pancreas 1.974 0.557 BQL Salivary Gland 2.049 0.395 0.047 Fatty Adipose (brown) 2.088 0.473 0.391 Adipose (white) 0.386 0.135 BQL Dermal Skin (non-pigmented) 2.277 0.602 0.121 Reproductive Epididymis 2.391 0.566 0.234 Prostate Gland 2.033 0.254 0.051 Seminal Vesicles 1.809 1.636 0.817 Testis 1.647 0.436 0.078 Skeletal/ Bone 0.449 0.095 BQL Muscular Heart 2.540 0.408 0.057 Skeletal Muscle 0.868 0.099 BQL Respiratory Lung 5.675 1.128 0.296 Alimentary Cecum 3.658 5.981 0.099 Canal Cecum (contents) 0.085 58.016 0.196 Esophagus 3.567 0.572 0.043 Large Intestine 3.581 2.019 0.129 Large Intestine BQL 35.809 0.374 (contents) Small Intestine 43.741 6.290 1.704 Small Intestine 95.869 66.664 0.039 (contents) Stomach (gastric 3.777 1.333 0.062 mucosa) Stomach (contents) 3.827 26.408 0.051 Ocular Eye (lens) BQL BQL BQL Eye (uveal tract) 2.342 0.469 0.095 BQL = Value is below the LLOQ; NI = Not identified on acceptable sections. LLOQ = 0.00048400 .mu.Ci/g/0.0135694 .mu.Ci/.mu.g = 0.036 .mu.g equivalent/g tissue ULOQ = 20.97300000 .mu.Ci/g/0.0135694 .mu.Ci/.mu.g = 1545.610 .mu.g equivalent/g tissue

TABLE-US-00075 TABLE 72 Concentrations of Radioactivity in Tissues of Male Long-Evans (Pigmented, Group 4) Rats After a Single 2-Hour Intravenous Infusion of [.sup.14C]brincidofovir at a Target Dose of 15 mg/kg Concentration (.mu.g equiv/g tissue) Rat #14 Rat #15 Rat #16 Rat #17 Rat #18 Rat #19 Rat #20 Rat #21 Rat #22 Tissue Type Tissue 1 h 2 h 4 h 8 h 24 h 72 h 96 h 168 h 840 h Vascular/ Plasma (by LSCa) 2.754 4.918 3.080 1.207 0.154 0.017 BQL BQL BQL Lymphatic Blood (cardiac) 3.040 5.291 2.839 1.679 0.225 0.052 BQL BQL BQL Bone Marrow 1.464 3.348 2.550 2.045 0.898 0.552 0.280 0.232 0.043 Lymph Node 1.682 4.002 2.244 1.936 0.745 0.710 0.270 0.247 0.062 Spleen 2.081 4.314 3.249 2.728 1.870 1.675 1.106 0.892 0.273 Thymus 1.223 2.125 1.205 1.107 0.403 0.359 0.215 0.100 BQL Excretory/ Bile (in duct) 243.705 508.358 178.597 BQL 1.010 0.526 0.044 BQL BQL Metabolic Kidney Cortex 36.713 97.276 76.678 56.528 19.453 4.070 1.041 0.330 BQL Kidney Medulla 8.092 26.987 25.330 22.989 7.117 1.478 0.648 0.138 BQL Liver 126.516 229.669 118.543 53.166 13.918 3.261 1.269 0.451 BQL Urinary Bladder 6.053 5.004 22.781 5.643 1.885 0.336 0.100 BQL BQL Urinary Bladder 19.057 26.805 814.445 82.440 22.258 1.911 0.357 0.104 BQL (contents) Central Brain (cerebellum) 0.569 1.144 0.422 0.284 0.099 0.073 BQL BQL BQL Nervous Brain (cerebrum) 0.391 0.887 0.341 0.196 0.070 0.062 BQL BQL BQL System Brain (medulla) 0.498 0.858 0.376 0.199 0.085 0.059 BQL BQL BQL Spinal Cord 0.462 0.940 0.277 0.116 0.060 BQL BQL BQL BQL Endocrine Adrenal Gland 2.523 4.802 3.282 3.179 1.253 1.186 0.776 0.490 0.091 Pituitary Gland 2.397 6.343 4.081 1.089 0.431 0.603 0.297 0.189 BQL Thyroid 1.486 2.454 1.884 1.532 0.567 0.376 0.197 0.459 BQL Secretory Harderian Gland 0.897 2.049 1.476 1.284 0.665 1.098 0.536 0.403 BQL Pancreas 0.864 1.896 1.051 0.834 0.315 0.194 0.082 BQL BQL Salivary Gland 1.335 2.508 1.153 0.821 0.314 0.190 0.134 0.056 BQL Fatty Adipose (brown) 0.839 1.680 1.123 0.755 0.306 0.402 0.187 0.132 BQL Adipose (white) 0.369 0.365 0.317 0.124 0.058 0.062 BQL BQL BQL Dermal Skin (non-pigmented) 0.979 2.556 1.925 1.273 0.312 0.443 0.269 0.226 BQL Skin (pigmented) 1.193 2.848 2.118 1.233 0.491 0.337 0.209 0.090 BQL Reproductive Epididymis 0.828 1.654 1.795 0.821 0.362 0.243 0.204 0.129 BQL Prostate Gland 0.561 0.901 0.715 0.542 0.157 0.104 0.127 BQL BQL Seminal Vesicles 0.938 1.834 2.285 1.717 1.127 1.374 1.038 0.361 BQL Testis 0.595 1.321 1.021 0.802 0.285 0.240 0.196 0.098 BQL Skeletal/ Bone 0.130 0.257 0.183 0.190 BQL 0.036 0.040 BQL BQL Muscular Heart 1.190 2.401 0.997 0.815 0.226 0.173 0.118 0.062 BQL Skeletal Muscle 0.427 0.611 0.343 0.152 0.063 0.070 0.040 BQL BQL Respiratory Lung 3.063 6.350 3.690 2.233 0.585 0.886 0.362 0.178 BQL Alimentary Cecum 1.781 3.052 2.443 6.189 7.077 0.527 0.208 0.080 BQL Canal Cecum (contents) 0.099 1.065 42.166 175.858 30.022 1.987 0.159 0.123 BQL Esophagus 1.750 2.456 1.803 1.747 0.377 0.357 0.098 0.041 BQL Large Intestine 1.455 5.497 2.338 3.071 3.064 0.993 0.545 0.278 BQL Large Intestine BQL 2.223 137.870 235.751 48.798 3.347 0.273 0.103 BQL (contents) Small Intestine 6.622 71.029 42.479 48.384 3.184 4.569 0.911 0.339 BQL Small Intestine 50.503 422.957 55.541 22.421 0.962 0.615 0.120 BQL BQL (contents) Stomach (gastric 1.446 2.518 1.430 1.223 0.295 0.225 0.110 0.069 BQL mucosa) Stomach (contents) 1.125 6.178 0.960 10.997 0.170 0.080 BQL BQL BQL Ocular Eye (lens) 0.039 0.077 BQL BQL BQL BQL BQL BQL BQL Eye (uveal tract) 0.875 1.411 1.412 0.925 0.452 0.322 0.188 BQL BQL

Group 3--QWBA Tissue Distribution in Albino Male Rats (Sparse Sampling)

[0595] [.sup.14C]Brincidofovir-derived radioactivity was well distributed into most tissues of albino male rats after a 2-h IV infusion at 15 mg/kg (n=1 animal at 2, 24, and 168 h after initiation of dosing), and most tissues had concentrations that were slightly lower than in blood/plasma at the end of the infusion, but were higher than blood/plasma for later time points. Excretory and alimentary canal tissue concentrations were much higher than blood/plasma at all time points. The highest observed concentration of [.sup.14C]brincidofovir-derived radioactivity in blood was 6.9 .mu.g equiv/g, which was observed at 2 h (end of infusion time and the first collection time point for this sparsely-sampled group). The C.sub.max of [.sup.14C]brincidofovir-derived radioactivity in most tissues (35 of 37 tissues) were found at 2 h post-dose (first sampled time point) when most of the tissues had concentrations that were between 1.0 and 6.0 .mu.g equiv/g.

[0596] The tissues with the highest concentrations (>20.0 .mu.g equiv/g) at the respective T.sub.max were: liver (229.4 .mu.g equiv/g), kidney cortex (106.5 .mu.g equiv/g), small intestine (43.7 .mu.g equiv/g), urinary bladder (36.0 .mu.g equiv/g), and kidney medulla (33.4 .mu.g equiv/g). The tissues with the lowest concentrations (<1.0 .mu.g equiv/g) at their respective T.sub.max were: brain, spinal cord, white adipose, bone, skeletal muscle, and eye lens.

[0597] The highest overall concentrations determined by QWBA were observed in the contents of the alimentary canal (C.sub.max ranged from 26.4 .mu.g equiv/g in stomach at 24 h to 95.9 .mu.g equiv/g in small intestine at 2 h), and urinary bladder contents (47.0 .mu.g equiv/g at 2 h), which reflected the routes of elimination for the [.sup.14C]brincidofovir-drug-derived radioactivity after a 2-h IV infusion.

[0598] At 168 h post-dose, elimination of drug related material was not complete in most tissues; however those tissues that did contain radioactivity had concentrations that were <1.0 .mu.g equiv/g.

Group 4--QWBA Tissue Distribution in Pigmented Male Rats

[0599] [.sup.14C]Brincidofovir-derived radioactivity was well distributed into most tissues of pigmented male rats after a 2-h IV infusion at 15 mg/kg (n=1 animal at 1, 2, 4, 8, 24, 72, 96, 168 and 840 h) after initiation of dosing, and most tissues had concentrations that were slightly lower than in blood/plasma at early time-points (up to 4 hours after infusion initiation), but were higher than blood/plasma for later time-points. Tissue:plasma (T/P) ratios based on AUC (AUC.sub.tissue:AUC.sub.plasma; Table 78) show values >1 for most tissues, indicating higher tissue concentrations relative to plasma. Excretory and alimentary canal tissue concentrations were much higher than blood/plasma at all time points. The highest observed concentration of [.sup.14C]brincidofovir-derived radioactivity in blood was 5.3 .mu.g equiv/g, which was observed at 2 h (end of infusion and the second collection time point for this intensively-sampled group). The C.sub.max of [.sup.14C]brincidofovir-derived radioactivity in most tissues (33 of 38 tissues) were found at 2 h post-dose when most of the tissues had concentrations that were between 1.0 and 6.0 .mu.g equiv/g.

[0600] The tissues with the highest concentrations (>20.0 .mu.g equiv/g) at the respective T.sub.max were: liver (229.7 .mu.g equiv/g), kidney cortex (97.3 .mu.g equiv/g), small intestine (71.0 .mu.g equiv/g), kidney medulla (27.0 .mu.g equiv/g), and urinary bladder (22.8 .mu.g equiv/g). The tissues with the lowest concentrations (<0.5 .mu.g equiv/g) at their respective T.sub.max were: brain (cerebrum and medulla), spinal cord, white adipose, prostate gland, bone, skeletal muscle, and eye lens.

[0601] The highest overall concentrations determined by QWBA were observed in the urinary bladder contents (814.4 .mu.g equiv/g at 4 h), bile (508.4 .mu.g equiv/g at 2 h), and contents of the alimentary canal (C.sub.max ranged from 11.0 .mu.g equiv/g in stomach at 8 h to 423.0 .mu.g equiv/g in small intestine at 2 h), which reflected the routes of elimination for the [.sup.14C]brincidofovir-drug-derived radioactivity after a 2-h IV infusion. Without wishing to be bound by theory, bile may have been the source for the high concentration of radioactivity in the intestinal contents with high concentrations of radioactivity present in early time points. The high concentration in the stomach would support this observation, since it is in close proximity to the entrance of bile into the digestive system, although gastric secretion may also possible, based on the observation that radioactivity was present in the gastric mucosa at 168 h.

[0602] Tissue concentrations in most tissues decreased steadily, but at 168 h post-dose, elimination of drug related material was not complete in most tissues. In tissues containing radioactivity the concentrations present were <1.0 .mu.g equiv/g. At 840 h post-dose (last sampled time point) radioactivity was still present in bone marrow, lymph node, spleen, and adrenal gland, but was approaching the LLOQ (0.036 .mu.g equiv/g) in these tissues except spleen (0.270 .mu.g equiv/g).

[0603] At sparse time-points examined in SD rats (Group 3), no substantial differences in tissue concentrations were observed relative to LE rats administered the same dose (Group 4). A comparison of tissue distribution of radioactivity between non-pigmented (SD rats Group 3) and pigmented tissues (LE rats Group 4) showed that concentrations in the pigmented tissues (i.e., eye uveal tract and pigmented skin) were similar to those concentrations observed in the non-pigmented tissues, which suggested that there was no specific association of [.sup.14C]brincidofovir-drug-derived radioactivity with melanin.

Group 5--QWBA Tissue Distribution in Pigmented Male Rats after IV Dosing at 2 mg/kg (Sparse Sampling)

[0604] A summary of tissue distribution of [.sup.14C]brincidofovir in male pigmented rats administered a single 2-h IV infusion at 2 mg/kg (n=1 animal at 2, 8, 24, and 72 h after initiation of dosing) is presented in Table 73.

TABLE-US-00076 TABLE 73 Concentrations of Radioactivity in Tissues of Male Long-Evans (Pigmented, Group 5) Rats After a Single 2-Hour Intravenous Infusion of [.sup.14C] brincidofovir at a Target Dose of 2 mg/kg Concentration (.mu.g equiv/g tissue) Rat# Rat# Rat# Rat# 23 24 25 26 Tissue Type Tissue 2 h 8 h 24 h 72 h Vascular/ Plasma (by LSC) 0.863 0.173 0.029 0.008 Lymphatic Blood (cardiac) 0.771 0.178 0.041 0.010 Bone Marrow 0.596 0.225 0.163 0.074 Lymph Node 0.453 0.219 0.149 0.085 Spleen 0.821 0.561 0.358 0.284 Thymus 0.331 0.136 0.078 0.050 Excretory/ Bile (in duct) 32.663 3.781 0.097 0.069 Metabolic Kidney Cortex 11.047 8.188 3.570 0.772 Kidney Medulla 3.359 1.362 0.854 0.257 Liver 34.158 10.631 4.037 1.393 Urinary Bladder 0.474 0.519 0.253 0.117 Urinary Bladder 22.905 3.544 1.348 0.249 (contents) Central Brain (cerebellum) 0.172 0.036 0.020 0.012 Nervous Brain (cerebrum) 0.150 0.030 0.016 0.006 System Brain (medulla) 0.101 0.027 0.019 0.006 Spinal Cord 0.151 0.024 0.011 0.006 Endocrine Adrenal Gland 0.685 0.440 0.245 0.167 Pituitary Gland 0.899 0.367 0.089 0.055 Thyroid 0.348 0.157 0.105 0.056 Secretory Harderian Gland 0.300 0.162 0.116 0.100 Pancreas 0.406 0.089 0.052 0.022 Salivary Gland 0.494 0.111 0.071 0.030 Fatty Adipose (brown) 0.356 0.099 0.050 0.039 Adipose (white) 0.120 0.015 0.033 0.005 Dermal Skin (non-pigmented) 0.288 0.268 0.108 0.068 Skin (pigmented) 0.460 0.186 0.116 0.047 Reproductive Epididymis 0.328 0.088 0.089 0.037 Prostate Gland 0.166 0.039 0.035 0.015 Seminal Vesicles 0.364 0.304 0.327 0.172 Testis 0.231 0.091 0.076 0.041 Skeletal/ Bone 0.022 0.017 0.019 0.010 Muscular Heart 0.373 0.109 0.040 0.025 Skeletal Muscle 0.167 0.023 0.017 0.007 Respiratory Lung 0.997 0.283 0.195 0.115 Alimentary Cecum 0.507 0.696 0.145 0.090 Canal Cecum (contents) 0.051 30.663 3.948 0.191 Esophagus 0.425 0.346 0.134 0.036 Large Intestine 0.545 1.028 0.504 0.128 Large Intestine 0.059 48.477 4.244 0.335 (contents) Small Intestine 1.710 2.462 0.704 0.076 Small Intestine 63.303 1.982 0.207 0.032 (contents) Stomach (gastric 0.553 0.122 0.077 0.031 mucosa) Stomach (contents) 0.014 0.051 0.006 0.008 Ocular Eye (lens) BQL BQL BQL BQL Eye (uveal tract) 0.352 0.144 0.092 0.061 BQL = Value is below the LLOQ LLOQ = 0.00048400 .mu.Ci/g/0.10151 .mu.Ci/.mu.g = 0.005 .mu.g equivalent/g tissue ULOQ = 20.97300000 .mu.Ci/g/0.10151 .mu.Ci/.mu.g = 206.610 .mu.g equivalent/g tissue

[0605] [.sup.14C]Brincidofovir-derived radioactivity was well distributed into most tissues of pigmented male rats after a 2-h IV infusion at 2 mg/kg, and most tissues had concentrations that were slightly lower than in blood/plasma at the end of infusion, but were higher than blood/plasma for later time points. Excretory and alimentary canal tissues were much higher than blood/plasma at all time points. The highest observed concentration of [.sup.14C] brincidofovir-derived radioactivity in blood was 0.771 .mu.g equiv/g, which was observed at 2 h (end of infusion and the first collection time point for this sparsely-sampled group) and was 7-fold lower than observed after a 15 mg/kg dose. The C.sub.max of [.sup.14C]brincidofovir-derived radioactivity in most tissues (35 of 38 tissues) were found at 2 h post-dose (first sampled time point) when most of the tissues had concentrations that were between 0.2 and 0.7 .mu.g equiv/g, which was about 10-fold lower than that observed in rats administered 15 mg/kg. Mean tissue concentration ratios after IV administration of 2 mg/kg (Group 5) compared to 15 mg/kg (Group 4) indicated that the increase in tissue concentrations were approximately dose proportional (i.e., with increase in dose of 7.5, a tissue concentration ratio between 4-15) in the vascular lymphatic, excretory/metabolic, central nervous, endocrine, secretory, fatty, dermal, reproductive, skeletal/muscular, and respiratory (lung) tissues (Table 74). Mean tissue concentration ratios were generally greater in the small intestine, indicating a greater than proportional increase in small intestine concentrations with an increase in dose from 2 to 15 mg/kg.

TABLE-US-00077 TABLE 74 [.sup.14C]Brincidofovir Ratio of Individual Tissue Concentration of Group 4 (15 mg/kg) to Group 5 (2 mg/kg) Concentration (.mu.g equiv/g tissue) G5 (2 G 4 (15 G 5 (2 G4 (15 G5 (2 G4 (15 G 5 (2 G 4 (15 mg/kg) mg/kg) mg/kg) mg/kg) mg/kg) mg/kg) mg/kg) mg/kg) Rat #23 Rat #15 Ratio Rat #24 Rat #17 Ratio Rat #25 Rat #18 Ratio Rat #26 Rat #19 Ratio Tissue Type Tissue 2 h 2 h 2 h 8 h Lo 8 h Hi 8 h 24 h Lo 24 h Hi 24 h 72 h Lo 72 h Hi 72 h Vascular/ Blood 0.771 5.291 7 0.178 1.679 9 0.041 0.225 5 0.010 0.052 5 Lymphatic (cardiac) Vascular/ Bone 0.596 3.348 6 0.225 2.045 9 0.163 0.898 6 0.074 0.552 7 Lymphatic Marrow Vascular/ Lymph 0.453 4.002 9 0.219 1.936 9 0.149 0.745 5 0.085 0.710 8 Lymphatic Node Vascular/ Spleen 0.821 4.314 5 0.561 2.728 5 0.358 1.870 5 0.284 1.675 6 Lymphatic Vascular/ Thymus 0.331 2.125 6 0.136 1.107 8 0.078 0.403 5 0.050 0.359 7 Lymphatic Excretory/ Kidney 11.047 97.276 9 8.188 56.528 7 3.570 19.453 5 0.772 4.070 5 Metabolic Cortex Excretory/ Kidney 3.359 26.987 8 1.362 22.989 17 0.854 7.117 8 0.257 1.478 6 Metabolic Medulla Excretory/ Liver 34.158 229.669 7 10.631 53.166 5 4.037 13.918 3 1.393 3.261 2 Metabolic Excretory/ Urinary 0.474 5.004 11 0.519 5.643 11 0.253 1.885 7 0.117 0.336 3 Metabolic Bladder Central Brain 0.172 1.144 7 0.036 0.284 8 0.020 0.099 5 0.012 0.073 6 Nervous (cerebellum) System Central Brain 0.150 0.887 6 0.030 0.196 7 0.016 0.070 4 0.006 0.062 10 Nervous (cerebrum) System Central Brain 0.101 0.858 8 0.027 0.199 7 0.019 0.085 4 0.006 0.059 10 Nervous (medulla) System Central Spinal Cord 0.151 0.940 6 0.024 0.116 5 0.011 0.060 5 0.006 BQL NC Nervous System Endocrine Adrenal 0.685 4.802 7 0.440 3.179 7 0.245 1.253 5 0.167 1.186 7 Gland Endocrine Pituitary 0.899 6.343 7 0.367 1.089 3 0.089 0.431 5 0.055 0.603 11 Gland Endocrine Thyroid 0.348 2.454 7 0.157 1.532 10 0.105 0.567 5 0.056 0.376 7 Secretory Harderian 0.300 2.049 7 0.162 1.284 8 0.116 0.665 6 0.100 1.098 11 Gland Secretory Pancreas 0.406 1.896 5 0.089 0.834 9 0.052 0.315 6 0.022 0.194 9 Secretory Salivary 0.494 2.508 5 0.111 0.821 7 0.071 0.314 4 0.030 0.190 6 Gland Fatty Adipose 0.356 1.680 5 0.099 0.755 8 0.050 0.306 6 0.039 0.402 10 (brown) Fatty Adipose 0.120 0.365 3 0.015 0.124 8 0.033 0.058 2 0.005 0.062 12 (white) Dermal Skin (non- 0.288 2.556 9 0.268 1.273 5 0.108 0.312 3 0.068 0.443 7 pigmented) Dermal Skin 0.460 2.848 6 0.186 1.233 7 0.116 0.491 4 0.047 0.337 7 (pigmented) Reproductive Epididymis 0.328 1.654 5 0.088 0.821 9 0.089 0.362 4 0.037 0.243 7 Reproductive Prostate 0.166 0.901 5 0.039 0.542 14 0.035 0.157 4 0.015 0.104 7 Gland Reproductive Seminal 0.364 1.834 5 0.304 1.717 6 0.327 1.127 3 0.172 1.374 8 Vesicles Reproductive Testis 0.231 1.321 6 0.091 0.802 9 0.076 0.285 4 0.041 0.240 6 Skeletal/ Bone 0.022 0.257 12 0.017 0.190 11 0.019 BQL NC 0.010 0.036 NC Muscular Skeletal/ Heart 0.373 2.401 6 0.109 0.815 7 0.040 0.226 6 0.025 0.173 7 Muscular Skeletal/ Skeletal 0.167 0.611 4 0.023 0.152 7 0.017 0.063 4 0.007 0.070 10 Muscular Muscle Respiratory Lung 0.997 6.350 6 0.283 2.233 8 0.195 0.585 3 0.115 0.886 8 Tract Alimentary Cecum 0.507 3.052 6 0.696 6.189 9 0.145 7.077 49 0.090 0.527 6 Canal Alimentary Esophagus 0.425 2.456 6 0.346 1.747 5 0.134 0.377 3 0.036 0.357 10 Canal Alimentary Large 0.545 5.497 10 1.028 3.071 3 0.504 3.064 6 0.128 0.993 8 Canal Intestine Alimentary Small 1.710 71.029 42 2.462 48.384 20 0.704 3.184 5 0.076 4.569 60 Canal Intestine Alimentary Stomach 0.553 2.518 5 0.122 1.223 10 0.077 0.295 4 0.031 0.225 7 Canal (gastric mucosa) Ocular Eye (lens) BQL 0.077 NC BQL BQL NC BQL BQL NC BQL BQL NC Ocular Eye 0.352 1.411 4 0.144 0.925 6 0.092 0.452 5 0.061 0.322 5 (uveal tract) NC = Not Calculated NI = Tissue not collected during sectioning NS = Not Sampled, tissues that could not be visualized on autoradioluminograph due to BQL radioactivity BQL = Value is below the LLOQ LLOQ = 0.00048400 .mu.Ci/g / 0.10151 .mu.Ci/.mu.g = 0.005 .mu.g equivalent / g tissue ULOQ = 20.97300000 .mu.Ci/g / 0.10151 .mu.Ci/.mu.g = 206.610 .mu.g equivalent / g tissue

[0606] The tissues with the highest concentrations >1.0 .mu.g equiv/g at the respective T.sub.max were: liver (34.2 .mu.g equiv/g), kidney cortex (11.0 .mu.g equiv/g), kidney medulla (3.4 .mu.g equiv/g), small intestine (2.5 .mu.g equiv/g), and large intestine (1.0 .mu.g equiv/g). The tissues with the lowest concentrations (<0.20 .mu.g equiv/g) at their respective T.sub.max were: brain, spinal cord, white adipose, prostate gland, bone, skeletal muscle, and eye lens.

[0607] The highest overall non-tissue concentrations determined by QWBA were observed in the contents of the alimentary canal (C.sub.max ranged from 0.051 .mu.g equiv/g in stomach at 8 h to 63.3 .mu.g equiv/g in small intestine at 2 h), bile (32.7 .mu.g equiv/g at 2 h), and urinary bladder contents (22.9 .mu.g equiv/g at 2 h), which reflected the routes of elimination for the [.sup.14C]brincidofovir-drug-derived radioactivity after a 2-h IV infusion at 2 mg/kg.

[0608] Tissue concentrations in most tissues decreased steadily, but at 72 h post-dose (last time point analyzed for this group), drug related material was detected in all tissues, but most tissues, not associated with organs of clearance, had concentrations that were <0.10 .mu.g equiv/g.

Group 6 and 7--QWBA Tissue Distribution in Albino and Pigmented Male Rats after PO Dosing at 15 mg/kg

[0609] A summary of tissue distribution of [.sup.14C]brincidofovir in male albino (Group 6; n=3) and pigmented rats (Group 7; n=10) are presented in Table 75 and Table 76, respectively. Pharmacokinetic parameters are given in Table 77.

TABLE-US-00078 TABLE 75 Concentrations of Radioactivity in Tissues of Male Sprague Dawley (Albino, Group 6) Rats After a Single Oral Dose of [.sup.14C]brincidofovir at a Target Dose of 15 mg/kg Concentration (.mu.g equiv./g) Rat# 27 Rat# 28 Rat# 29 Tissue Type Tissue 2 h 24 h 168 h Vascular/ Plasma (by LSC) 0.313 0.253 BQL Lymphatic Blood (cardiac) 0.200 0.197 BQL Bone Marrow 0.080 0.407 0.075 Lymph Node 0.095 0.329 0.124 Spleen 0.116 0.831 0.239 Thymus 0.045 0.142 BQL Excretory/ Bile (in duct) 9.639 0.465 0.045 Metabolic Kidney Cortex 2.752 18.379 0.588 Kidney Medulla 2.189 7.926 0.274 Liver 7.260 20.567 0.450 Urinary Bladder 4.367 17.321 0.087 Urinary Bladder 9.808 39.903 0.549 (contents) Central Brain (cerebellum) 0.035 BQL BQL Nervous Brain (cerebrum) BQL BQL BQL System Brain (medulla) BQL BQL BQL Spinal Cord BQL BQL BQL Endocrine Adrenal Gland 0.114 0.927 0.035 Pituitary Gland BQL 0.257 0.071 Thyroid 0.084 0.291 0.036 Secretory Harderian Gland 0.062 0.315 0.100 Pancreas 0.092 0.164 0.034 Salivary Gland 0.067 0.103 BQL Fatty Adipose (brown) 0.053 0.212 BQL Adipose (white) BQL 0.079 BQL Dermal Skin (non-pigmented) 0.088 0.189 0.063 Reproductive Epididymis 0.073 0.091 0.075 Prostate Gland 0.069 0.217 BQL Seminal Vesicles 0.048 0.333 0.097 Testis 0.039 0.118 BQL Skeletal/ Bone BQL 0.047 BQL Muscular Heart 0.080 0.116 BQL Skeletal Muscle 0.039 BQL BQL Respiratory Lung 0.184 0.296 0.078 Alimentary Cecum 0.109 7.533 0.064 Canal Cecum (contents) BQL 28.579 0.170 Esophagus 0.593 0.271 BQL Large Intestine 0.084 2.194 0.076 Large Intestine BQL 34.450 0.168 (contents) Small Intestine 236.279 84.063 2.680 Small Intestine 321.878 11.948 0.054 (contents) Stomach (gastric 11.183 1.075 0.041 mucosa) Stomach (contents) 877.490 5.664 BQL Ocular Eye (lens) BQL BQL BQL Eye (uveal tract) 0.145 0.290 BQL BQL = Value is below the LLOQ LLOQ = 0.00048400 .mu.Ci/g/0.01427 .mu.Ci/.mu.g = 0.034 .mu.g equivalent/g tissue ULOQ = 20.97300000 .mu.Ci/g/0.01427 .mu.Ci/.mu.g = 1469.727 .mu.g equivalent/g tissue

TABLE-US-00079 TABLE 76 Concentrations of Radioactivity in Tissues of Male Long-Evans (Pigmented, Group 7) Rats After a Single Oral Dose of [.sup.14C]brincidofovir at a Target Dose of 15 mg/kg Concentration (.mu.g equiv/g tissue) Rat #30 Rat #31 Rat #32 Rat #33 Rat #34 Rat #35 Rat #36 Rat #37 Rat #38 Rat #39 Tissue Type Tissue 0.5 h 1 h 2 h 4 h 8 h 24 h 72 h 96 h 168 h 840 h Vascular/ Plasma 0.040 0.248 0.446 1.216 1.499 0.184 0.017 BQL BQL BQL (by LSC) Lymphatic Blood (cardiac) BQL 0.280 0.435 0.978 1.132 0.160 BQL 0.035 BQL BQL Bone Marrow BQL 0.085 0.117 0.254 0.412 0.391 0.167 0.211 0.094 BQL Lymph Node 0.050 0.153 0.097 0.380 0.421 0.389 0.339 0.141 0.129 BQL Spleen 0.267 0.111 0.166 0.319 0.533 0.759 0.469 0.466 0.274 BQL Thymus 0.083 0.065 0.104 0.154 0.224 0.116 0.040 0.083 BQL BQL Excretory/ Bile (in duct) 0.572 8.660 20.537 36.265 17.878 0.614 2.425 0.317 0.140 BQL Metabolic Kidney Cortex 0.146 1.452 3.976 10.662 17.789 8.640 2.368 1.696 0.342 0.051 Kidney 0.096 0.602 3.593 5.104 9.070 2.724 1.023 0.653 0.201 BQL Medulla Liver 0.432 7.701 12.037 36.137 34.744 18.184 2.183 2.680 0.393 0.034 Urinary Bladder BQL 1.146 0.214 1.588 3.728 0.659 0.177 0.256 0.076 BQL Urinary Bladder 0.200 11.376 7.593 17.825 110.910 11.541 0.205 1.292 0.278 BQL (contents) Central Brain BQL BQL BQL 0.037 0.075 BQL BQL BQL BQL BQL (cerebellum) Nervous Brain BQL BQL BQL 0.035 0.066 BQL BQL BQL BQL BQL (cerebrum) System Brain BQL BQL BQL 0.049 0.043 BQL BQL BQL BQL BQL (medulla) Spinal Cord BQL BQL BQL 0.037 BQL BQL BQL BQL BQL BQL Endocrine Adrenal Gland BQL 0.180 0.372 0.749 1.331 0.578 0.278 0.172 0.058 BQL Pituitary Gland BQL 0.045 0.085 0.330 0.266 0.151 0.076 0.073 0.042 BQL Thyroid BQL 0.118 0.168 0.317 0.298 0.139 0.117 0.089 0.038 BQL Secretory Harderian BQL 0.046 0.090 0.181 0.244 0.150 0.155 0.194 0.110 BQL Gland Pancreas 0.066 0.166 0.193 0.255 0.214 0.096 0.051 0.044 BQL BQL Salivary Gland BQL 0.087 0.112 0.212 0.234 0.109 BQL 0.052 BQL BQL Fatty Adipose 0.040 0.099 0.084 0.302 0.240 0.099 0.050 0.044 0.037 BQL (brown) Adipose (white) BQL BQL 0.063 0.089 0.076 BQL BQL BQL BQL BQL Dermal Skin (non- BQL 0.102 0.100 0.261 0.429 0.107 0.058 0.067 BQL BQL pigmented) Skin BQL 0.086 0.156 0.416 0.429 0.157 0.069 0.077 BQL BQL Reproductive Epididymis BQL 0.084 0.064 0.294 0.229 0.146 0.064 0.108 0.042 BQL Prostate Gland BQL 0.077 0.120 0.148 0.128 0.100 BQL 0.052 BQL BQL Seminal BQL 1.018 0.109 0.221 0.244 0.259 0.156 0.185 0.063 BQL Vesicles Testis BQL 0.042 0.051 0.142 0.223 0.152 0.078 0.098 0.041 BQL Skeletal/ Bone BQL BQL BQL 0.099 0.043 0.035 BQL BQL BQL BQL Muscular Heart BQL 0.083 0.182 0.247 0.358 0.121 0.061 0.077 0.045 BQL Skeletal BQL 0.046 0.060 0.074 0.098 BQL BQL BQL BQL BQL Muscle Respiratory Lung BQL 0.226 0.386 0.768 1.025 0.178 0.136 0.105 0.065 BQL Alimentary Cecum BQL 0.127 0.289 0.931 7.561 0.688 0.189 0.327 0.137 BQL Canal Cecum BQL BQL 0.050 38.957 256.303 35.629 1.035 0.698 0.071 BQL (contents) Esophagus BQL BQL 0.185 4.636 0.458 0.179 BQL 0.071 BQL BQL Large Intestine BQL 0.107 0.136 0.265 3.338 1.681 0.276 0.413 0.060 BQL Large Intestine BQL BQL BQL BQL 378.695 69.825 1.236 1.252 0.108 BQL (contents) Small Intestine 72.672 374.71 295.445 458.485 178.32 74.587 6.889 16.979 5.867 0.070 Small Intestine 53.944 82.546 72.107 187.589 98.195 69.109 0.215 9.654 BQL BQL (contents) Stomach 2.769 7.730 11.955 0.833 0.880 0.937 0.108 0.101 0.042 BQL (gastric mucosa) Stomach 670.358 1004.619 328.988 274.917 17.792 0.056 BQL BQL BQL BQL (contents) Ocular Eye (lens) BQL BQL BQL BQL BQL BQL BQL BQL BQL BQL Eye (uveal BQL BQL 0.193 0.227 0.601 0.178 BQL 0.075 BQL BQL tract) BQL = Value is below the LLOQ LLOQ = 0.00048400 .mu.Ci/.mu.g = 0.034 .mu.g equivalent/g tissue ULOQ = 20.97300000 .mu.Ci/g/0.01427 .mu.Ci/.mu.g = 1469.727 .mu.g equivalent/g tissue

TABLE-US-00080 TABLE 77 Rat Tissue Pharmacokinetic Parameters After a Single Oral Dose of [.sup.14C]brincidofovir at a Target Dose of 15 mg/kg Rat PK No. of AUC.sub.all AUC.sub.inf_obs C.sub.max (.mu.g points (.mu.g (.mu.g Organ/Tissue (equiv/g) t.sub.1/2 (h) in t.sub.1/2 r.sup.2 T.sub.max (h) equiv h/g) equiv h/g) Adipose (brown) 0.302 233.5 3 0.96 4 24.3703 24.4043 Adipose (white) .sup.a 0.089 17.6 2 1.00 4 1.1215 2.4387 Adrenal Gland 1.331 43.2 3 1.00 8 74.5860 58.7136 Blood (cardiac) .sup.a 1.132 32.8 2 1.00 8 21.9165 22.3146 Bone .sup.a 0.099 53.9 2 1.00 4 1.8470 3.7274 Bone Marrow a 0.412 96.3 3 0.75 8 68.7413 50.2191 Brain (cerebellum) 0.075 ND 0 ND 8 0.8610 ND Brain (cerebrum) 0.066 ND 0 ND 8 0.7650 ND Brain (medulla) 0.049 ND 0 ND 4 0.5770 ND Cecum .sup.a 7.561 69.9 4 0.71 8 174.4118 142.1970 Epididymis .sup.a 0.294 89.0 4 0.72 4 31.1150 22.3936 Esophagus 4.636 37.2 3 0.88 4 27.9015 29.1559 Eye (lens) 0 ND ND ND ND ND ND Eye (uveal tract) .sup.a 0.601 57.7 2 1.00 8 16.2765 19.8244 Harderian Gland .sup.a 0.244 151.8 3 0.64 8 63.7645 50.8931 Heart .sup.a 0.358 110.8 4 0.81 8 31.1603 23.2365 Kidney Cortex .sup.a 17.789 244.8 2 1.00 8 804.4980 822.5076 Kidney Medulla 9.070 41.2 3 1.00 8 342.0130 286.4307 Large Intestine .sup.a 3.338 37.6 3 0.82 8 140.3313 123.4302 Liver .sup.a 36.137 190.3 2 1.00 4 1426.6340 1435.9700 Lung 1.025 93.0 3 0.99 8 53.1145 39.9962 Lymph Node .sup.a 0.421 86.4 3 0.56 8 85.04325 57.7701 Pancreas.sup.b 0.255 62.1 3 0.98 4 10.3720 12.7271 Pituitary Gland 0.330 106.2 3 0.97 4 30.5073 22.8331 Plasma (LSC) 1.499 ND ND ND 8 22.3885 ND Prostate Gland 0.148 70.1 3 0.99 4 7.6578 11.0471 Salivary Gland a 0.234 67.4 2 1.00 8 9.1933 12.3800 Seminal Vesicles 1.018 64.7 3 0.86 1 50.2500 34.9600 Skeletal Muscle 0.098 ND 0 ND 8 1.3265 ND Skin (non-pigmented) .sup.a 0.429 93.7 3 0.72 8 14.0275 20.6749 Skin (pigmented) .sup.a 0.429 63.4 3 0.81 8 17.0405 21.3144 Small Intestine 458.485 98.1 3 0.99 4 9575.1810 9585.0880 Spinal Cord 0.037 ND 0 ND 4 0.1110 ND Spleen 0.759 115.4 3 0.95 24 172.2208 125.7566 Stomach (mucosa) 11.955 66.8 3 0.97 2 90.7575 80.6917 Testis .sup.a 0.223 87.2 3 0.77 8 30.3920 21.7757 Thymus .sup.a 0.224 96.9 3 0.23 8 12.0843 20.6983 Thyroid 0.317 59.0 3 1.00 4 31.3395 21.8086 Urinary Bladder 3.728 49.7 4 0.87 8 111.2445 91.1627 ND = Not determined; unable to determine value from the data. .sup.a t.sub.1/2 values for these tissue/organs are obtained from insufficient data points and/or the resulting r.sup.2 values fall outside acceptance criteria (.gtoreq.0.85), thus, these values should be considered estimates.

Group 6--QWBA Tissue Distribution in Albino Male Rats after Dosing (Sparse Sampling)

[0610] [.sup.14C]Brincidofovir-derived radioactivity was well distributed into most tissues of albino male rats after a single oral dose at 15 mg/kg (n=1 at 2, 24, and 168 h after dosing), and most tissues had concentrations that were slightly lower than in blood/plasma at early time-point (2 hours after dosing), but were higher than blood/plasma for later time points. Excretory and alimentary canal tissue concentrations were much higher than blood/plasma at all time points. The highest observed concentration of [.sup.14C]brincidofovir-derived radioactivity in blood was 0.200 .mu.g equiv/g, which was observed at 2 h (the first collection time point for this sparsely-sampled group). The C.sub.max of [.sup.14C]brincidofovir-derived radioactivity in most tissues (31 of 37 tissues) were found at 24 h post-dose (second sampled time point) when most of the tissues had concentrations that were between 0.10 and 0.50 .mu.g equiv/g.

[0611] The tissues with the highest concentrations (>7.0 .mu.g equiv/g) at the respective T.sub.max were: small intestine (236.3 .mu.g equiv/g), liver (20.6 .mu.g equiv/g), kidney cortex (18.4 .mu.g equiv/g), urinary bladder (17.3 .mu.g equiv/g), stomach (11.2 .mu.g equiv/g), kidney medulla (7.9 .mu.g equiv/g), and cecum (7.5 .mu.g equiv/g). The tissues with the lowest concentrations (<0.10 .mu.g equiv/g) at their respective T.sub.max were: brain, spinal cord, white adipose, bone, skeletal muscle, and eye lens.

[0612] The highest overall concentrations determined by QWBA were observed in the contents of the alimentary canal (C.sub.max ranged from 28.6 .mu.g equiv/g in cecum at 24 h to 877.5 .mu.g equiv/g in stomach at 2 h), urinary bladder contents (39.9 .mu.g equiv/g at 24 h), and bile (9.7 .mu.g equiv/g at 2 h), which reflected the route of dose administration and/or the routes of elimination for the [.sup.14C]brincidofovir-drug-derived radioactivity after a single oral dose at 15 mg/kg.

[0613] At 168 h post-dose, elimination of drug related material was not complete, with detectable concentrations (<0.60 .mu.g equiv/g) in approximately half of the tissues. In small intestine, a higher residual concentration (2.7 .mu.g equiv/g) was observed relative to other tissues at 168 h.

Group 7--QWBA Tissue Distribution in Pigmented Male Rats after PO Dosing

[0614] [.sup.14C]Brincidofovir-derived radioactivity was well distributed into most tissues of pigmented male rats after a single oral dose at 15 mg/kg (n=1 animal at 0.5, 1, 2, 4, 8, 24, 72, 96, 168 and 840 h after dosing), and most tissues had concentrations that were slightly lower than in blood/plasma at time points up to 24 hours after dosing, but were higher than blood/plasma for later time points. Excretory and alimentary canal tissue concentrations were much higher than blood/plasma at all time points. Tissue:plasma (T/P) ratios based on AUC (Table 78) show values >1 for most tissues, indicating higher tissue concentration relative to plasma. The highest observed concentration of [.sup.14C]brincidofovir-derived radioactivity in blood was 1.1 .mu.g equiv/g, which was observed at 8 h. The C.sub.max of [.sup.14C]brincidofovir-derived radioactivity in most tissues (20 of 38 tissues) were found at 8 h post-dose when most of the tissues had concentrations that were between 0.20 and 0.40 .mu.g equiv/g.

TABLE-US-00081 TABLE 78 Tissue Distribution of Total Radioactivity in Male Long-Evans Rats Following a Single Two Hour Intravenous Infusion (Group 4) or Oral Dose (Group 7) of [.sup.14C]brincidofovir at a Target Dose of 15 mg/kg Tissue/Plasma Ratio Tissue C.sub.max AUC.sub.all AUC.sub.all Tissue/AUC.sub.all Concentration (.mu.g equiv/g) (.mu.g equiv h/g) Plasma .mu.g equiv/g (168 h) IV PO Ratio IV PO Ratio IV PO IV PO Tissue (Group 4) (Group 7) (PO/IV) (Group 4) (Group 7) (PO/IV) (Group 4) (Group 7) (Group 4) (Group 7) Plasma (by 4.918 1.499 0.30 36.3690 22.3885 0.62 1.0 1.0 BQL BQL LSC) Blood 5.291 1.132 0.21 45.3555 21.9165 0.48 1.2 1.0 BQL BQL (cardiac) Adipose 1.68 0.302 0.18 96.6220 24.3703 0.25 2.7 1.1 0.132 0.037 (brown) Adipose 0.369 0.089 0.24 7.1955 1.1215 0.16 0.2 0.1 BQL BQL (white) Adrenal 4.802 1.331 0.28 384.2580 74.5860 0.19 10.6 3.3 0.49 0.058 Gland Bone 0.257 0.099 0.39 6.1805 1.8470 0.30 0.2 0.1 BQL BQL Bone Marrow 3.348 0.412 0.12 197.3860 68.7413 0.35 5.4 3.1 0.232 0.094 Brain 1.144 0.075 0.07 12.1870 0.8610 0.07 0.3 0.04 BQL BQL (cerebellum) Brain 0.887 0.066 0.07 9.1765 0.7650 0.08 0.3 0.03 BQL BQL (cerebrum) Brain 0.858 0.049 0.06 9.7470 0.5770 0.06 0.3 0.0 BQL BQL (medulla) Cecum 7.077 7.561 1.07 360.7580 174.4118 0.48 9.9 7.8 0.08 0.137 Epididymis 1.795 0.294 0.16 95.0160 31.1150 0.33 2.6 1.4 0.129 0.042 Esophagus 2.456 4.636 1.89 73.1850 27.9015 0.38 2.0 1.2 0.041 BQL Eye (lens) 0.077 ND ND 0.1545 ND ND 0.0042 ND BQL BQL Eye (uveal 1.412 0.601 0.43 51.5575 16.2765 0.32 1.4 0.7 BQL BQL tract) Harderian 2.049 0.244 0.12 257.6905 63.7645 0.25 7.1 2.8 0.403 0.11 Gland Heart 2.401 0.358 0.15 58.1205 31.1603 0.54 1.6 1.4 0.062 0.045 Kidney 97.276 17.789 0.18 1919.6850 804.4980 0.42 52.8 35.9 0.33 0.342 Cortex Kidney 26.987 9.07 0.34 717.8445 342.0130 0.48 19.7 15.3 0.138 0.201 Medulla Large 5.497 3.338 0.61 310.7965 140.3313 0.45 8.5 6.3 0.103 0.06 Intestine Liver 229.669 36.137 0.16 2149.7645 1426.6340 0.66 59.1 63.7 0.451 0.393 Lung 6.35 1.025 0.16 180.1960 53.1145 0.29 5.0 2.4 0.178 0.065 Lymph Node 4.002 0.421 0.11 208.8530 85.04325 0.41 5.7 3.8 0.247 0.129 Pancreas 1.896 0.255 0.13 36.2010 10.3720 0.29 1.0 0.5 BQL BQL Pituitary 6.343 0.33 0.05 155.1085 30.5073 0.20 4.3 1.4 0.189 0.042 Gland Prostate 0.901 0.148 0.30 24.3415 7.6578 0.62 0.7 0.3 BQL BQL Gland Salivary 2.508 0.234 0.09 60.9180 9.1933 0.15 1.7 0.4 0.056 BQL Gland Seminal 2.285 1.018 0.45 297.3580 50.2500 0.17 8.2 2.2 0.361 0.063 Vesicles Skeletal 0.611 0.098 0.16 10.3485 1.3265 0.13 0.3 0.1 BQL BQL Muscle Skin (non- 2.556 0.429 0.17 146.2340 14.0275 0.10 4.0 0.6 0.226 BQL pigmented) Skin 2.848 0.429 0.15 95.5050 17.0405 0.18 2.6 0.8 0.09 BQL (pigmented) Small 71.029 458.485 6.45 1160.6505 9575.1810 8.25 31.9 427.7 0.339 5.867 Intestine Spinal Cord 0.94 0.037 0.04 5.7830 0.1110 0.02 0.2 0.005 BQL BQL Spleen 4.314 0.759 0.18 642.3590 172.2208 0.27 17.7 7.7 0.892 0.274 Stomach 2.518 11.955 4.75 70.2310 90.7575 1.29 1.9 4.1 0.069 0.042 (mucosa) Testis 1.321 0.223 0.17 77.2835 30.3920 0.39 2.1 1.4 0.098 0.041 Thymus 2.125 0.224 0.11 92.4355 12.0843 0.13 2.5 0.5 0.1 BQL Thyroid 2.454 0.317 0.13 238.0230 31.3395 0.13 6.5 1.4 0.459 0.038 Urinary 22.781 3.728 0.16 215.5480 111.2445 0.52 5.9 5.0 BQL 0.076 Bladder BQL = Below quantification limit LLOQ = 0.036 ug equiv/g (Group 4 IV) LLOQ = 0.034 ug equiv/g (Group 7 PO) ND = Not determined, insufficient data

[0615] FIG. 4 and FIG. 5 show plots of the concentration of brincidofovir in various tissue as a function of time and route of administration. FIG. 4 compares the concentration of brincidofovir in the small intestine for oral and IV dosing as a function of time. FIG. 5 compares the concentration of brincidofovir in the kidney cortex for oral and IV dosing as a function of time.

[0616] The tissues with the highest concentrations (>2.0 .mu.g equiv/g) at the respective T.sub.max were: small intestine (458.5 .mu.g equiv/g), liver (36.1 .mu.g equiv/g), kidney cortex (17.8 .mu.g equiv/g), stomach (12.0 .mu.g equiv/g), kidney medulla (9.1 .mu.g equiv/g), cecum (7.6 .mu.g equiv/g), urinary bladder (3.7 .mu.g equiv/g), and large intestine (3.3 .mu.g equiv/g). The tissues with the lowest concentrations (<0.10 .mu.g equiv/g) at their respective T.sub.max were: brain, spinal cord, white adipose, bone, skeletal muscle, and eye lens.

[0617] The highest overall concentrations determined by QWBA were observed in the contents of the alimentary canal (C.sub.max ranged from 187.6 .mu.g equiv/g in small intestine at 4 h to 1004.6 .mu.g equiv/g in stomach at 1 h), urinary bladder contents (110.9 .mu.g equiv/g at 8 h), and bile (36.3 .mu.g equiv/g at 4 h), which reflected route of dose administration and/or the routes of elimination for the [.sup.14C]brincidofovir drug-derived radioactivity after a single oral dose.

[0618] Tissue concentrations in most tissues decreased steadily, but at 168 h post-dose, drug related material was detected in approximately half of the tissues, with most concentrations <0.40 .mu.g equiv/g, except for small intestine (5.9 .mu.g equiv/g). At 840 h post-dose (last sampled time point) radioactivity was still present in small intestine, kidney cortex, and liver, but concentration were near the LLOQ (0.034 .mu.g equiv/g) in all tissues.

[0619] The primary route of elimination of radioactivity after a 2-h IV infusion of [.sup.14C]brincidofovir at 15 mg/kg in male SD rats was in urine, which accounted for an average of 51.2% of the administered dose over the 168 h collection period. An average of 42.2% of the administered dose was recovered in feces. Most (.about.86%) of the urinary and fecal excretion of radioactivity occurred in the first 24 h after dosing.

[0620] Following a 2-h IV infusion, radioactivity in most tissues (except small intestine) increased in proportion to dose (from 2 to 15 mg/kg) across each time point. In small intestinal tissue, the increase was greater than proportional to dose. Following oral administration radioactivity was readily absorbed. Radioactivity was well distributed with peak concentrations of radioactivity in most tissues (34 of 38) occurring at 4 to 8 h after oral administration, and at the end of the IV infusion (2-h). The qualitative distribution patterns of the radioactivity were similar after IV or oral administration; quantitatively, tissue radioactive exposure, adjusted for the radioactive bioavailability (approximately 50%), generally was greater after IV administration than after oral gavage. Tissues with highest concentrations of radioactivity were associated with excretory organs (i.e., liver, kidney, and intestine). The tissue to plasma ratios were high (>30) for each of these tissues, and in kidney cortex and liver the ratios were similar between the IV and oral routes of administration. Small intestinal tissue to plasma ratio was highest after oral administration (T/P=428) as expected based on the route of administration. However, notable radioactivity was associated with the small intestine (tissue to plasma ratio 32) after IV infusion. High concentration (C.sub.max) was associated with bile in the duct, which was 14 times greater after IV (C.sub.max 508 .mu.g equiv/g) than oral (C.sub.max=36 .mu.g equiv/g) administration, suggesting a possible source for the high small intestinal tissue concentrations after IV administration. Tissues with lowest concentrations of radioactivity were brain, spinal cord, skeletal muscle, white adipose tissue and bone. Association of radioactivity in the brain and spinal cord was higher after IV administration (.about.20% of plasma C.sub.max concentration and .about.5% of plasma C.sub.max concentration after oral administration). No evidence of specific association with melanin containing tissues (eye, uvea) was detected.

[0621] The predicted exposure in humans following a 100 .mu.Ci oral dose was estimated using four different methods (including single tissue and whole body exposure estimates), all of which resulted in a human exposure that was 4% or less of the FDA and ICRP allowed limits.

[0622] There were no observed SOP and/or protocol deviations that occurred on the study that had a detrimental effect on the quality or integrity of the study.

IV BCV Single Ascending Dose Study in Healthy Subjects

[0623] Brincidofovir was administered to healthy subjects at 10 mg and 25 mg. IV brincidofovir at 10 mg was found to provide similar exposure as orally administered brincidofovir at 100 mg. It was found that both IV brincidofovir doses (10 mg and 25 mg) were generally safe and well-tolerated. No drug related adverse events, no gastrointestinal adverse events, and no graded lab abnormalities (e.g., no hematologic toxicity and no kidney toxicity) were observed.

[0624] A single dose of oral BCV at 100 mg and 200 mg resulted in about 5% instance of diarrhea, and a 350 mg dose resulted in about 20% instance of diarrhea.

[0625] FIG. 6 shows a plot of the plasma brincidofovir concentration (AUC.sub.inf (ng*h/mL)) at different 100 mg oral administration, 10 mg IV administration, and 25 mg IV administration. As shown in FIG. 6, IV administration of 10 mg brincidofovir provided substantially the same plasma concentration as oral administration of 100 mg brincidofovir, while IV administration of 25 mg brincidofovir provided higher plasma concentration than both the IV 10 mg dose and the oral 100 mg dose.

Example 10--Brincidofovir: A 28-Day Intravenous Infusion Toxicity Study in Rats with a 14-Day Recovery Period

Summary

[0626] Sprague-Dawley CD.RTM. rats (15/sex/group) were administered 0 (2.times. Sodium Phosphate Buffer Solution), 1, 4 or 15 mg/kg/dose brincidofovir twice per week via 2-hour intravenous infusion on a total of 9 occasions over 29 days. The dose rate was 10 mL/kg/hr for all dose groups. At the end of the treatment phase, up to 10 animals/sex/group were euthanized and necropsied. The remaining animals (up to 5 animals/sex/group) were held for a 14-day recovery phase in order to determine progression or reversibility of any brincidofovir-related effects. Satellite animals (up to 3/sex/group) were similarly dosed and serial blood samples were collected from each animal on Days 1 and 29 for toxicokinetic analysis of brincidofovir and the brincidofovir metabolite (Cidofovir, CDV). Parameters evaluated during the study were: viability, clinical observations, ophthalmology, body weights, food consumption, clinical pathology (termination of dosing and end of recovery), organ weights, macroscopic observations and microscopic pathology.

[0627] Brincidofovir exposure increased with increasing dose on Day 1 and Day 29 with a trend towards a greater than proportional increase in AUC.sub.last versus dose. Without wishing to be bound by theory, in general, there was no accumulation of brincidofovir over the course of the 29 day study. Male to female ratios for brincidofovir indicated there were no sex differences in exposures (based on C.sub.max and AUC.sub.last).

[0628] On Days 1 and 29, following intravenous administration of brincidofovir, CDV C.sub.max and AUC.sub.last values increased with increasing brincidofovir dose (from 1 to 15 mg/kg). In general, there was little to no accumulation of CDV over the course of the study and male to female ratio generally indicated there were no sex differences in CDV exposures.

[0629] There were no brincidofovir-related mortalities. There were 3 unscheduled deaths on the study, 2 deaths in toxicity animals were determined not to be brincidofovir-related and 1 death in a TK animal for which the cause could not be determined.

[0630] There were no brincidofovir-related effects on ophthalmology, body weights, hematology, coagulation, clinical chemistry, or urinalysis.

[0631] Administration of brincidofovir resulted in intermittent (generally following infusion) clinical signs in some but not all animals (rapid breathing and hunched posture with and without piloerection, partially closed eyes, decreased activity irregular breathing, and/or flattened posture) in both sexes at .gtoreq.4 mg/kg/dose and minimally decreased food consumption at .gtoreq.1 mg/kg/dose in males and at 15 mg/kg/dose in females.

[0632] At the end of dosing, brincidofovir-related findings were present in the male reproductive tract (testes, epididymides and seminal vesicles) and the intestinal tract (duodenum, jejunum, and/or ileum) at .gtoreq.4 mg/kg/dose and skin (sebaceous glands) of males and females at 15 mg/kg/dose. In the male reproductive tract, testicular changes were characterized by decreased testicular weights and slight to marked germ cell depletion at .gtoreq.4 mg/kg, with minimal spermatid retention and Sertoli cell vacuolation at 15 mg/kg/dose. In addition, males at 15 mg/kg/dose had minimal luminal cell debris in the epididymides and minimally increased epithelial apoptosis in the seminal vesicles. In the intestinal tract, single cell necrosis was observed in the small intestine (crypts) at .gtoreq.4 mg/kg/dose and in the large intestine (glands) at 15 mg/kg/dose in both sexes. Sebaceous gland atrophy in the skin was present in males and females at 15 mg/kg.

TABLE-US-00082 TABLE 79 Comparison of toxicity of orally and intravenously administered brincidofovir ORAL BRINCIDOFOVIR (Single Dose) IV BRINCIDOFOVIR (Repeat Dose) Exposure Exposure DOSE (ng/mL or DOSE (ng/mL or Preliminary (mg/kg) ng hr/mL) Findings (mg/kg) ng hr/mL) Findings 30 C.sub.max-426 Single cell necrosis of 1 C.sub.max-298 No GI findings AUC-5564 the epithelial cells lining AUC-410 mucosal crypts or glands at the base of small intestinal villi 100 C.sub.max-1086 Diarrhea, dehydration, 4 C.sub.max-1200 Minimal to slight AUC-13552 .dwnarw. Body weight AUC- 2759 small intestine Gross - distended necrosis crypt; stomach minimal crypt Enteritis with epithelial hyperplasia loss and inflammation 38% mortality 300 C.sub.max-1186 Clinical: diarrhea, 15 C.sub.max-4500 Slight to minimal AUC-20506 dehydration, .dwnarw. Body AUC-9893 small intestine crypt weight necrosis; minimal Gross - distended crypt hyperplasia stomach, jejunum, Minimal epithelial cecum, colon gland necrosis large Extensive loss of intestine mucosal epithelial cells with inflammatory infiltrate into lamina propria 100% mortality 1000 C.sub.max-1375 Clinical: Diarrhea, Note: Infusion of BCV resulted in AUC-26449 dehydration, .dwnarw. body intermittent clinical signs of weight rapid breathing, hunched posture- Gross - distended with or without piloerection in stomach, jejunum, some animals during infusion; and cecum, colon fully resolved shortly following GI toxicity similar to infusion. 300 mg/kg 100% mortality

[0633] Following a 14-day recovery period, all findings completely recovered with the exception of testicular weight decreases and germ cell depletion in males at .gtoreq.4 mg/kg/dose; however, without wishing to be bound by theory, germ cell depletion would not be expected to completely recover in 2 weeks. In addition, a few animals at 15 mg/kg/dose (3/5 males; females) had bone marrow decreased cellularity and soft and/or small testes and epididymides (males). Without wishing to be bound by theory, germ cell depletion in the testes and decreased sperm in the head region of the epididymides were reflective of maturation depletion secondary to the loss of germ cell populations observed at the end of the dosing period.

[0634] Under the conditions of the study, based on the testicular germ cell depletion in males at .gtoreq.4 mg/kg/dose and moderate intestinal single cell crypt necrosis in 1 female at 15 mg/kg/dose, the no-observed-adverse-effect level (NOAEL) for brincidofovir was considered to be 1 mg/kg/dose in males and 4 mg/kg/dose for in females.

Introduction

[0635] The following abbreviations are used in this study: [0636] M Male(s) [0637] F Female(s) [0638] .about.approximately [0639] mg/kg milligrams of test item per kilogram of body weight [0640] mL/kg milliliters of dose formulation per kilogram of body weight [0641] mL/kg/hr milliliters of dose formulation per kilogram of body weight per hour [0642] mg/mL milligrams of test item per milliliter of dose formulation [0643] IV Intravenous [0644] MTD Maximum tolerated dose [0645] DRF Dose range finding

[0646] The purpose of this study was to assess the toxicity and toxicokinetics of brincidofovir when administered via intravenous (2-hour) infusion to rats 2 times per week for a total of 9 doses over the course of 29 days, and to evaluate the progression or reversibility of brincidofovir effects during a drug-free post-treatment recovery period of 14 days.

[0647] The study design incorporates elements of general regulatory guidelines for toxicity studies.

[0648] This study was conducted in compliance with the principles of Good Laboratory Practice as set forth in: United States Food and Drug Administration's Good Laboratory Practice Regulations (Part 58 of 21 CFR); and OECD Principles of Good Laboratory Practice (as revised in 1997), ENV/MC/CHEM(98)17

[0649] These principles are compatible with Good Laboratory Practice regulations specified by regulatory authorities throughout the European Community, the United States (EPA and FDA), and Japan (MHLW, MAFF and METI), and other countries that are signatories to the OECD Mutual Acceptance of Data Agreement.

[0650] This study complied with all appropriate parts of the Animal Welfare Act Regulations: 9 CFR Parts 1 and 2 Final Rules, Federal Register, Volume 54, No. 168, Aug. 31, 1989, pp. 36112-36163 effective Oct. 30, 1989 and 9 CFR Part 3 Animal Welfare Standards; Final Rule, Federal Register, Volume 56, No. 32, Feb. 15, 1991, pp. 6426-6505 effective Mar. 18, 1991.

[0651] Pre-clinical studies in two or more species (one rodent, one non-rodent) are recommended by regulatory agencies such as the FDA to support administration in humans. The rat is an animal model commonly utilized in toxicity studies. In addition, a historical data base is available for comparative evaluation.

[0652] The test item was administered intravenously up to 2 times per week.

[0653] The number of animals in this study was considered to be the minimum necessary for statistical, regulatory and scientific reasons. The purpose of this study was to monitor for toxicity and progression or reversibility of effects of the test item. Historical control data indicate that clinical laboratory data, organ weight data and microscopic examination of tissues vary among individual animals. The number of rats utilized in the conduct of subchronic studies (10 animals/sex/group) is recommended by regulatory guidance documents and was considered the minimum number that would account for the expected variability among animals. To determine progression or reversibility of potential effects of the test item, additional post-treatment (recovery) animals (5 animals/sex/group) were included in the study. Three test item-treated groups receiving low, intermediate and high multiples of the proposed human dose and a negative control group were considered the minimum number of groups necessary to provide a range of effects and allowed for extrapolation of results to humans.

[0654] The number of animals selected for toxicokinetic evaluations (3/sex/treated group) was considered the minimum number necessary to provide meaningful data, given the inherent variability in absorption, distribution, metabolism and excretion processes. A control group with 3 animals/sex was necessary to evaluate the absence of the test item.

[0655] The doses for this study (0, 1, 4 and 15 mg/kg/dose) were 2, 4, 10 or 15 mg/kg/dose over the course of 2 hours in the maximum tolerated dose (MTD) phase, and a second set of animals were then administered 3 doses of 1 or 15 mg/kg/dose over the course of 2 hours in the dose range finding (DRF) phase over the course of 9 days. The high dose (15 mg/kg) administered on this 2-phase study, was determined to be the maximum feasible dose (MFD) based on hemolysis observed at higher concentrations.

[0656] Because the infusion catheter receded or was not usable on the scheduled day of dosing and insufficient time to reposition or repair the line, the following animals were dosed via intravenous injection into the tail vein on Day 29; Animal Nos. 1511 and 1519 were found on Day 28 with receded catheters, and Animal No. 4581 was found on Day 29 with an ulceration near the exteriorization site. These animals were restrained into individual plastic cones for no more than 2.25 hours (15 minutes for dose site preparation activities and 2 hours for dose administration) during this procedure.

TABLE-US-00083 TABLE 80 Experimental Outline Number of animals Dose Toxicity Recovery Satellite Treat- (mg/ study phase study.sup.b Group ment kg).sup.a Male Female Male Female Male Female 1 Control 0 15 15 5 5 3 3 2 BCV 1 15 15 5 5 3 3 3 BCV 4 15 15 5 5 3 3 4 BCV 15 15 15 5 5 3 3 .sup.aDoses represent active ingredient. .sup.bToxicokinetic samples were collected on Day 1 and Day 29.

[0657] The first day of dosing was defined as Day 1 of the study. Brincidofovir was provided as a white powder and was stored at room temperature. The purity was determined to be 99.8%.

[0658] The diluent was a 5% dextrose solution for injection (USP) and was supplied as a clear liquid (96.8% pure). It was stored at room temperature.

[0659] 2.times. Sodium Phosphate buffer solution was used as a control. It was prepared from monobasic sodium phosphate (anhydrous), supplied as a white crystalline powder and stored at room temperature (100% pure). The phosphate buffer (control) solution was dissolved in sterile water and pH was adjusted with sodium hydroxide.

Preparation of Dose Formulations

[0660] A vehicle solution of 2.times. sodium phosphate buffer solution (400 mM, pH 8.0.+-.0.04) was prepared by mixing the appropriate amounts of monobasic sodium phosphate solution (400 mM) and dibasic sodium phosphate solution (400 mM). The pH of the solution was adjusted with monobasic sodium phosphate solution (400 mM), when necessary. The solution was filtered through a 0.22 .mu.m Millex.RTM.-GP (PES) filter within a laminar flow hood into a sterile vessel.

[0661] The vehicle (control item) was stored refrigerated at 2-8.degree. C. Fresh vehicle solution was formulated twice; once prior to Dose 1 and again prior to Dose 4, and each formulation was used within one month of preparation.

[0662] A brincidofovir stock of 15 mg/mL was prepared by mixing the appropriate amount of brincidofovir with 2.times. sodium phosphate buffer solution, sterile water for injection, USP and 1N sodium hydroxide (NaOH). The solution was filtered through a 0.22 .mu.m Millex.RTM.-GP filter under a laminar flow hood into a sterile vessel. Brincidofovir stock was prepared once prior to Dose 1 and again prior to Dose 4, and stored refrigerated at 2-8.degree. C.

[0663] Dosing formulations were prepared by diluting the appropriate amounts of the brincidofovir stock (15 mg/mL) or stock vehicle (2.times. sodium phosphate buffer solution) with the appropriate amounts of 5% dextrose for injection, USP, into a sterile vial under a laminar flow hood and inverting 10 times to mix. Fresh dosing formulations were prepared weekly and were stored refrigerated 2-8.degree. C. when not in use.

TABLE-US-00084 TABLE 81 Dosing Dose Concentration Volume dose Group Treatment (mg/kg) (mg/mL) (mL/kg/hr) 1 Control 0 0 10 2 BCV 1 0.05 10 3 BCV 4 0.2 10 4 BCV 15 0.75 10

[0664] Detailed records of brincidofovir usage were maintained. The amount of brincidofovir necessary to prepare the formulations and the amount actually used were determined at each preparation.

Stability

[0665] Stability analyses demonstrated that brincidofovir formulations at concentrations bracketing those used in this study (0.05 mg/mL and 2.0 mg/mL) are stable for 24 hours when stored at room temperature, and for 8 days when stored refrigerated at 2 to 8.degree. C. The brincidofovir stock solution of 15.0 mg/mL was stable for 24 hours when stored at room temperature (nominally 20.degree. C.) and for 8, 14 and 28 days when stored at 2 to 8.degree. C.

Confirmation Analysis

[0666] Two samples (5 mL each) were taken on the day of preparation from the middle region of each dosing formulation (including control) prepared for each week. One sample was analyzed, in duplicate, for dose confirmation analysis and one sample was retained refrigerated (2 to 8.degree. C.) (Due to initial results outside the study acceptance plan criteria (.+-.10% of nominal), the Group 2 retained samples were re-analyzed for Weeks 1 and 2).

Storage Conditions

[0667] All dose formulation samples were stored refrigerated (2 to 8.degree. C.) until analysis or final disposition.

Brincidofovir Administration

[0668] Brincidofovir was administered as an intravenous infusion over 2 hours. Animals were treated at constant doses in 20 mL/kg/2 hours.

[0669] Infusion Catheter Implantation Procedure

[0670] Catheters for infusion were implanted approximately 1-3 weeks prior to dose administration. All animals, including the spares, were surgically implanted with a catheter.

[0671] The surgical site was prepared as per Testing Facility's SOP for aseptic, recovery surgical procedures. Animals received analgesics pre-emptively (flunixin meglumine (USP) 2 mg/kg, subcutaneously).

[0672] All animals were anesthetized (isoflurane) and shaved in the inguinal and dorsal regions. A small incision was made and the femoral vein isolated. A small incision in the vessel was performed and a sterile catheter was placed into the vessel. The catheter was inserted via the femoral vein and passed retrograde into the inferior vena cava, just caudal to the kidneys. The catheter was secured with sutures and passed subcutaneously so as to exit at the nape of the neck. A small pocket was made at the exteriorization site in which a loop of catheter remained. The inguinal region was closed by sutures.

[0673] The animals were placed in jackets and the implanted catheters were attached to pins with capped septum connectors. The catheters were locked with taurolidine citrate locking solution. Each animal received enrofloxacin 5.0 mg/kg intramuscularly on the day of surgery.

[0674] Surgical repairs (1 repair per animal limit) to restore catheter patency were performed as necessary, during the pretest period and at the discretion of the Study Director during the on-test period. Analgesics were administered preemptively (flunixin meglumine 2 mg/kg (USP), subcutaneously). Animals undergoing a surgical repair received an appropriate dose of antibiotics (as per the Standard Operating Procedures of the Testing Facility) on the day of surgery.

[0675] Catheters were tied off after the completion of dose administration just prior to recovery (a knot was placed in the catheter and the catheter was receded under the skin) and jackets were removed.

Infusion Catheter Maintenance Procedure

[0676] Implanted femoral vein catheters were assessed for patency as per Testing Facility's SOP prior to placing animals on study.

Volume Dose (Rate)

[0677] 20 mL/kg/2 hours (10 mL/kg/hour).

Individual Dose Concentration

[0678] The individual animal concentrations were calculated from the most recently recorded scheduled bodyweight.

Frequency and Duration

[0679] The test/control item was administered via 2 hours intravenous infusion twice weekly for a total of 9 individual doses over 29 days. Test/control item administration continued through the day prior to terminal necropsy. Recovery animals were held for a 14 day observation period after the last dose.

Dose Site

[0680] Surgically implanted cannula in the femoral vein/vena cava or via peripheral tail vein.

Dosing Procedure

[0681] One to two days prior to dose initiation, the lock solution was withdrawn from each animal's implanted catheter (if possible) and the catheter was flushed with saline before connecting to a tether and infusion dosing set. After connecting the animals' catheters, the animals were infused with sterile saline (0.9% NaCl, USP) at a rate of 0.5 mL/hr by a calibrated Medfusion syringe pump until dose administration on Day 1.

[0682] A saline flush was infused to deliver the test item in the infusion lines and to ensure the delivery of a complete dose (an additional -0.5 to 1 mL volume, at the same rate as the test item, was administered to flush the catheter line).

[0683] Animals were maintained on saline in between dose administrations. Saline syringes were changed at least every 4 days.

[0684] Animals with an unusable/non-functioning catheter were surgically repaired when possible. If repair was not possible, these animals were dosed via a peripheral tail vein in a restrainer, at the discretion of the Study Director. Animals were placed into individual plastic cones for no more than 2.25 hours (15 minutes for dose site preparation activities and 2 hours for dose administration via tail vein injection).

[0685] On Day 29, Animal Nos. 1511, 1519 and 4581 were administered Dose 9 via peripheral tail vein.

Dosing Accuracy

[0686] The accountability (confirmation of actual dose administered) for each day of infusion was calculated based on the weight of the each syringe predose and postdose, and reviewed for adjustments daily; the data is retained in the study files. Pumps were checked for accuracy prior to dose initiation with saline using the same length of dose at rate as the study, and post dose accountability was used to monitor pump performance throughout the study.

Animals

Strain/Species

[0687] Albino Rats (Outbred) VAF/Plus.RTM. CD.RTM. (Sprague-Dawley derived) [Crl:CD.RTM. (SD)BR]

Number of Animals

[0688] 170 Total received (85 males and 85 females); 144 placed on test (72 males and 72 females).

Pretest Period

[0689] The pretest period was approximately 4 weeks. All animals were examined during the pretest period to confirm suitability for study. Pretest procedures were not performed until animals had been allowed to stabilize for 5 days.

Age of the Main Study Animals at Start of Treatment

[0690] Approximately 10 weeks

Weight Range of the Main Study Animals at the Start of Treatment

[0691] Males: 290.6 g to 422.4 g. Females: 203.8 g to 294.7 g

Assignment

[0692] More animals than required for the study were purchased and stabilized. Animals considered unsuitable for the study on the basis of pretest physical examinations and catheter patency were eliminated prior to random selection for group assignment. Animals considered suitable for study were distributed into 4 groups of 18 animals per sex by a computerized random sort program so that body weight means for each group were comparable. Due to pretest ophthalmology findings, several toxicity animals were swapped with satellite animals but remained within the same group. Individual weights of animals placed on test were within .+-.20% of the mean weight for each sex.

Identification of Animals

[0693] Each rat was implanted with a BMDS IMI-1000 Implantable Radio Frequency Transponder (microchip) programmed with a unique number. This number was cross referenced with an animal number assigned by the Testing Facility; this number plus the study number comprised a unique identification number for each animal. In addition, each cage was provided with a cage card that was color-coded for dose level identification and contained study number and facility-assigned animal number information.

Veterinary Care

[0694] Animals were monitored by the technical staff for any conditions requiring possible veterinary care and treated as necessary. Miscellaneous, non-test item-related veterinarian evaluations for individual animals were reviewed by the study director and are documented in the study file.

[0695] Serial observations performed on the animals from the satellite groups were confined to clinical observations, body weights, ophthalmology and food consumption; these data are not reported.

[0696] Blood samples were obtained for the determination of plasma concentrations of brincidofovir and the brincidofovir metabolite, cidofovir.

[0697] On Days 1 and 29, blood samples were obtained for toxicokinetic determinations at the timepoints shown in the table below:

TABLE-US-00085 TABLE 82 Collection times and number of animals Number of Toxicokinetic Intervals Timepoints Animals Day 1 2 and 24 hours post initiation of 3 animals/sex/group dose administration (Group 1) Predose, 1, 2, 8, 24, and 48 hours 3 animals/sex/group post initiation of dose administration (Groups 2, 3, 4) Day 29 2 and 24 hours post initiation of 3 animals/sex/group dose administration (Group 1) Predose, 1, 2, 8, 24, and 48 hours 3 animals/sex/group post initiation of dose administration (Groups 2, 3, 4)

Collection Procedures

[0698] Approximately 0.40 mL of whole blood was obtained via the tail vein from each animal at each time point. Animals were not fasted prior to blood collection. Blood was collected into tubes containing K.sub.2EDTA anticoagulant and placed on wet ice in an upright position. Plasma was separated by centrifugation (for 10 minutes at approximately 2000 g, at approximately 2-8.degree. C.). Approximately 0.150 mL of plasma was transferred into a single cryotube appropriately labeled as to study number, animal number, time point, date of sampling and sample type. Remaining plasma was transferred into a second cryotube and retained as a backup sample. Plasma was obtained and frozen at approximately -80.+-.10.degree. C. within approximately 2 hours after collection of each blood sample until analysis. Animals were euthanized (CO.sub.2 inhalation) after the final blood collection.

Storage and Disposition of Samples

[0699] All sample tubes were stored frozen (-80.+-.10.degree. C.) and shipped (frozen, on dry ice) to Pyxant Labs, Colorado Springs, Colorado for analysis.

Analysis/Reporting

[0700] Bioanalytical samples were analyzed using a validated liquid chromatographic-triple quadruple mass spectrometric (LC-MS/MS) assay at Pyxant Labs.

[0701] Upon receipt of the plasma concentration data, noncompartmental toxicokinetic analysis was performed by Nuventra, Inc. Durham, N.C.

Daily Observations

[0702] Animals were observed in their cages twice daily for mortality and signs of severe toxic or pharmacologic effects. Animals in extremely poor health or in a possible moribund condition were identified for further monitoring and possible euthanasia.

Dosing Observations

[0703] Observations were made immediately and 2 hours after the completion of dosing for any signs of poor health or toxic or pharmacologic effects (e.g., abnormalities in general condition, appearance, activity, behavior, respiration, etc.).

Signs

[0704] Animals were removed from their cages and examined once pretest and weekly during the study period. Examinations included observations of general condition, skin and fur, eyes, nose, oral cavity, abdomen and external genitalia as well as evaluations of respiration.

Ophthalmoscopic Examination

[0705] All animals were examined pretest. All surviving toxicity animals were examined at end of dosing and end of recovery. Only animals which were within normal limits at the pretest examination were placed on test.

[0706] Lids, lacrimal apparatus and conjunctiva were examined visually. The cornea, anterior chamber, lens, iris, vitreous humor, retina and optic disc were examined by indirect ophthalmoscopy.

[0707] The pupils of each animal were dilated prior to examination using tropicamide ophthalmic solution.

Body Weight

[0708] Animals were removed from their cages and weighed twice pretest, on the day prior to each dose during the treatment period and weekly during the recovery period. Terminal, fasted body weights were obtained just prior to the scheduled necropsy intervals. Animals euthanized for welfare reasons were weighed prior to necropsy.

Food Consumption

[0709] Food consumption was measured (weighed) weekly, beginning one week prior to treatment.

Clinical Pathology

[0710] Blood obtained via the orbital sinus (retrobulbar venous plexus) under light isoflurane anesthesia was used to analyze hematology, coagulation and clinical chemistry parameters for up to 10 toxicity animals/sex/group at the termination of dosing and up to 5 recovery animals/sex/group at the end of recovery. Animals were fasted overnight prior to each blood collection interval.

[0711] Urine obtained via a 16-hour overnight collection period was analyzed for up to 10 toxicity animals/sex/group at the termination of dosing and up to 5 recovery animals/sex/group at the end of recovery. Animals were fasted but not water-deprived during the collection period.

Hematology

[0712] Blood samples (approximately 0.25 mL) were collected into tubes containing K.sub.2EDTA anticoagulant and analyzed for the following using a Siemens ADVIA 120 Hematology Analyzer: Hemoglobin concentration (HGB); Hematocrit (HCT); Erythrocyte count (RBC); Platelet count (PLT); Mean corpuscular volume (MCV); Mean corpuscular hemoglobin (MCH); Mean corpuscular hemoglobin concentration (MCHC); Red cell distribution width (RDW); Total leukocyte count (WBC); Reticulocyte count (RETIC); Differential leukocyte count (Manual differential leukocyte counts were performed for verification and absolute values were calculated if necessary).; Neutrophils (ANEU); Lymphocytes (ALYM); Eosinophils (AEOS); Basophils (ABASO); Monocytes (AMONO); and Large unstained cells (ALUC).

[0713] A peripheral blood smear was prepared for each animal at each blood collection interval and was available for confirmation of automated results and/or other evaluations deemed necessary.

Coagulation

[0714] Blood samples (approximately 1.0 mL) were collected into tubes containing sodium citrate anticoagulant and analyzed for the following using a Diagnostica Stago Products STA Compact.RTM. mechanical clot detection system: Prothrombin time (PT): and Activated partial thromboplastin time (APTT).

Clinical Chemistry

[0715] Blood samples (approximately 1.0 mL) were collected into tubes with no anticoagulant, allowed to clot, centrifuged to obtain serum and analyzed for the following using a Siemens ADVIA 1800 Chemistry Analyzer:

[0716] Aspartate aminotransferase (AST) (Kinetic--Modified Bergmeyer); Alanine aminotransferase (ALT) (Kinetic--Modified Bergmeyer); Alkaline phosphatase (ALKP) (Kinetic Tietz AMP Buffer); Blood urea nitrogen (BUN) (Enzymatic--Roch-Ramek with Urease); Creatinine (CREAT) (Jaffe Picric Acid in Alkaline Medium); Glucose (GLU) (Glucose Hexokinase II Method); Cholesterol (CHOL) (Enzymatic esterase/oxidase--Trinder Endpoint); Triglycerides (TRIG) (Fossati Three Step Enzymatic--Trinder Endpoint); Total protein (TP) (Biuret Technique); Albumin (ALB) (Bromocresol Green Method); Total bilirubin (TBILI) (Oxidation with Vandate); Sodium (NA.sup.+) (Ion Selective Electrode) Potassium (K.sup.+) (Ion Selective Electrode); Chloride (Cl.sup.-) (Ion Selective Electrode); Calcium (Ca.sup.++) (Michaylova & Ilkova, Arsenazo III); and Inorganic phosphorus (PHOS) (Phosphomolybdate--UV Method)

Urinalysis

[0717] Urine was collected into ice-chilled containers overnight (approximately 16 hours) from toxicity study animals housed in metabolism cages.

[0718] Urine samples were analyzed for the following using Multistix reagent strips, interpreted using a Siemens Clinitek Advantus: pH; Protein (PRO); Glucose (GLU); Ketones (KET); Bilirubin (BIL); and Occult blood (BLD).

[0719] The samples were also analyzed for the following using manual methods:

[0720] Appearance (APP); Specific gravity (Sp.G.) (Clinical Refractometer, Atago Uricon-N); Volume (VOL).

Postmortem

Method of Euthanasia

[0721] Animals sacrificed for welfare reasons or at the scheduled sacrifice intervals were euthanized by exsanguination following isoflurane inhalation. Toxicokinetic animals were euthanized via CO.sub.2 inhalation.

Scheduled Necropsy

[0722] Necropsy was performed on up to 10 toxicity study animals/sex/group after animals had been treated for 29 days and on up to 5 toxicity study animals/sex/group after a 14 day treatment-free recovery period. Animals were fasted overnight prior to necropsy. Necropsy schedules were established to ensure that examination of animals of both sexes from each group were performed at similar times of the day throughout the necropsy periods.

[0723] Toxicokinetic animals were euthanized following the final blood collection on Day 29 and discarded without examination.

Macroscopic Examinations

[0724] Complete macroscopic examinations were performed on all animals. The macroscopic examination included examination of the external surface and all orifices; the external surfaces of the brain and spinal cord; the organs and tissues of the cranial, thoracic, abdominal and pelvic cavities and neck; and the remainder of the carcass for the presence of macroscopic morphologic abnormalities.

[0725] A macroscopic examination was performed for TK Animal No. 4107, which was sacrificed for welfare reasons, tissues were preserved but not examined microscopically.

Organ Weights

[0726] Organs indicated below were weighed for all surviving toxicity and recovery study animals at the scheduled sacrifice intervals. Prior to weighing, the organs were carefully dissected and properly trimmed to remove adipose and other contiguous tissues in a uniform manner. Organs were weighed as soon as possible after dissection in order to avoid drying. Paired organs were weighed together.

Tissues Preserved and Examined Histopathologically

[0727] The tissues listed below were obtained from all animals and preserved. In addition, slides of the indicated tissues were prepared and examined microscopically. Any abnormalities not noted during macroscopic examinations which were seen during histology processing were recorded.

TABLE-US-00086 TABLE 83 Tissues weighed, preserved and examined microscopically EXAMINED PRE- MICRO- ORGAN NAME WEIGHED SERVED SCOPICALLY animal identity X adrenal glands X X X aorta (thoracic) X X bone marrow smear X (femur) bone and marrow X X (femur with joint) bone and marrow X X.sup.a (sternum) brain (medulla, X X X pons, cerebrum and cerebellum) epididymides X X X esophagus X X eyes X X Harderian gland X X heart X X X infusion site X X kidneys X X X lacrimal glands X X large intestine X X.sup.b (cecum, colon, rectum) liver X X X lungs (with X X mainstem bronchi) lymph nodes X X (axillary, mesenteric) mammary gland X X.sup.c (inguinal) nerve (sciatic) X X optic nerve X ovaries X X X pancreas X X pituitary gland X.sup.d X X prostate gland X.sup.e X X salivary glands X X (mandibular) seminal vesicles X.sup.e X X skeletal muscle X X {rectus femoris) skin (dorsal - X X base of tail) small intestine X X.sup.c (duodenum, ileum, jejunum, Peyer's patches\GALT) spinal cord X X (cervical, thoracic, lumbar) spleen X X X stomach X X testes X X X thymus X X X thyroid/ X.sup.d X X parathyroid glands tongue X trachea X X urinary bladder X X uterus (body/homs) X X X with cervix vagina X X Zymbal's gland X tissues with X X macroscopic findings including tissue masses .sup.aQualitative examination (no differential count). .sup.bRectum was not examined microscopically. .sup.cMammary gland for males and GALT were evaluated only if present in routine sections. .sup.dWeighed post-fixation. .sup.eProstate and seminal vesicles were weighed together.

Preservatives

[0728] All tissues were preserved in 10% neutral buffered formalin (NBF). The eyes and testes were placed in Modified Davidson's solution initially and then retained in 10% NBF. Lungs were infused with 10% NBF prior to their immersion into a larger volume of the same fixative. Smear preparations of the marrow from the femur were air dried and fixed in absolute methanol.

Processing

[0729] After fixation, the tissues and organs from all animals were routinely processed, embedded in paraffin, cut at a microtome setting of 4-7 microns, mounted on glass slides, stained with hematoxylin and eosin and examined by light microscopy. The bones were decalcified in Decalcifier II.TM..

[0730] The data collection system used for collecting the in-life and post-life data divides the study into phases. The start of each phase begins with Day 1/Week 1. The phases presented in this report are:

[0731] Pre-Treatment: Begins the day the animals arrive. Phase days/weeks are labeled P1, P2, P3, etc. During this phase, animals are not being dosed and they have not been assigned their permanent identification numbers.

[0732] Randomization: Begins the day the animals are sorted into groups and assigned their permanent identification numbers. Phase days/weeks are labeled Ra1, Ra2, Ra3, etc. Animals are not being dosed during this phase.

[0733] Treatment: Begins for each animal when it receives its first administration of test/control item as specified in the study plan.

[0734] Recovery: Begins the day after each designated animal stops receiving its specified dose and continues until study completion. Phase days/weeks are labeled R1, R2, R3, etc.

[0735] The death codes used have the following meanings:

[0736] "FD": Found dead; "S1": Scheduled euthanasia 1; "WE": euthanized for welfare reasons; "Z1": Recovery euthanasia 1.

Clinical Observations

[0737] Clinical observations are presented for each animal that showed signs, providing detail of the type of sign, day or week of occurrence and information on the duration of the sign applicable. "AM": Morning Observation; "PM": Afternoon Observation; "Unsched": Unscheduled Observation; "Post Dose": Immediately After Completion of Dosing; "2 Hr PD": 2 Hours After Completion of Dosing.

Ophthalmic Examination

[0738] Observations are presented for each animal that showed findings. Observations were bilateral unless otherwise indicated.

Food Consumption

[0739] Food consumption (g/day) was calculated as: grams of food consumed/#days

Food consumption (g/animal/day) was calculated as: grams of food consumed/animal days.sup.a

.sup.aSum of whole days each animal is alive in the cage for each consumption period. If an animal dies the day of death is not counted as an animal day. For example: 4 animals in a cage for 5 days equals 20 animal days; if one animal died on the 3.sup.rd day then the animal days equals 1

Hematology

[0740] The following abbreviations were used: "ACMO": Automated count invalidated by manual observations; "CS": Clotted specimen; "NSAD": No sample animal died.

[0741] Sample comments (SCM1) were described using the following abbreviations: "CLSE": Severe platelet clumping noted; "CLSL": Slight platelet clumping noted; "NCLP": No clumping.

Coagulation

[0742] The following abbreviations were used: "CS": Clotted specimen; "NS": No specimen; "NSAD": No sample animal dead; "NVIR": Not valid due to irreproducible result.

[0743] Sample comments (SCM1) were described using the following abbreviations: "SLH": Slightly hemolyzed.

Blood Chemistry

[0744] BUN values below the analytical limit (<10) are excluded from calculations. TBILI values below the analytical limit (<0.2) are excluded from calculations.

[0745] The following abbreviations were used: "NS": No specimen; "NSAD": No sample animal dead; "1+H": Minimal hemolysis.

Globulin (GLOB) was calculated as: total protein-albumin

Albumin/globulin ratio (A/G) was calculated as: albumin/globulin

Urinalysis

[0746] Group means are presented for volume, pH, and specific gravity only.

[0747] The following other abbreviations were used: "NS": No specimen.

[0748] The appearance and color were described using the following abbreviations: "CLOG": Cloudy; "CLR": Clear; "SLCL": Slightly cloudy; "STR": Straw; "YELL": Yellow.

TABLE-US-00087 TABLE 84 Qualitative results of Multistix diagnostic reagents are reported according to the following convention: NEG Negative Protein TR Trace levels detected 30 30 mg/dL 100 100 mg/dL .gtoreq.300 .gtoreq.300 mg/dL Glucose 100 100 mg/dL 250 250 mg/dL 500 500 mg/dL .gtoreq.1000 .gtoreq.1000 mg/dL Ketones TR Trace levels detected 15 15 mg/dL 40 40 mg/dL .gtoreq.80 .gtoreq.80 mg/dL Bilirubin SM Small amount detected MOD Moderate amount detected LG Large amount detected Occult Blood TR Trace levels detected SM Small amount detected MOD Moderate amount detected LG Large amount detected

Organ Weights

[0749] Organ weights were presented as absolute, relative to terminal body weight using the weight recorded on the day of necropsy and relative to brain weight.

Pathology

[0750] Unless otherwise specified in a histopathology note, the organ/tissue examined was the required (routine) section.

[0751] The following abbreviations were used: "B-": Benign; "Cath": Catheter; "GALT": Gut-associated lymphoid tissue; "Infuse": Infusion; "LS": Longitudinal; "M-": Malignant; "TS": Transverse.

[0752] Histopathology findings were graded according to the following scale:

TABLE-US-00088 Minimal The change is barely discernible and/or very few/very small foci or areas are affected. Slight The change is more noticeable but only evident as few/small foci or areas affected. Moderate The change is obviously present, and of appreciable size and/or number. Marked The change is abundant in many areas of the section and/or is of prominent size. Severe The change affects a large proportion of the tissue and/or is of a large size.

[0753] All statistical analyses were carried out separately for males and females using the individual animal as the basic experimental unit.

[0754] The following data types were analyzed at each timepoint separately: body weight; body weight change from interval to interval; food consumption; hematology; coagulation; clinical chemistry; urinalysis; and organ weights.

[0755] The parameters to analyze were identified as continuous, discrete or binary. Test-item treated groups were then compared to the control using the following procedures.

Study Plan Deviations

[0756] The following study plan deviations occurred during the study, but without wishing to be bound by theory, were not considered to have compromised the validity or integrity of the study.

[0757] On Day 1, the processed 8 hour post-dose toxicokinetic samples for Groups 2 to 4 were left on dry ice overnight. Samples were found frozen the following morning. However, the required temperature (-80.degree. C. (.+-.10.degree. C.)) was not monitored overnight. Samples were immediately transferred to freezer SL301 at nominal -80.degree. C. (.+-.10.degree. C.).

[0758] On Pretest Day 13, Animal No. 1015 did not have surgery performed due to significantly low body weight.

[0759] On Day 13, viability checks were not performed on the spare animals. However, animals were within normal limits the day prior to and the day after Day 13.

[0760] Surgical repairs for Animal Nos. 1592 (Day 21) and 4078 (Day 10) were not acknowledged by the Study Director until 7 and 2 days after repair, respectively.

[0761] On Day 29, low blood volumes were collected for toxicokinetic samples from Animal Nos. 2036 (8 hour post dose timepoint) and 3050 (2 hour post dose timepoint) because the animals were "difficult to bleed" and "slow to bleed", respectively.

[0762] The pituitary gland tissue for Animal No. 4074 was collected at the recovery necropsy, but was later found to be missing at the post-fix trimming; a weight was not obtained.

[0763] On Day 29, the immediate post-dose observation was not performed on Animal No. 4580.

[0764] On Day 1, the 24 hour toxicokinetic samples for Animal Nos. 1016 and 2036 were processed 1 minute over the "within the 2 hours" specified in the study plan.

[0765] At the terminal necropsy, the collection site for clinical pathology samples was not documented for Animal No. 1004.

[0766] On Day 23, toxicokinetic Animal No. 4107 was transferred to necropsy due to welfare reasons, however, the animal was no documented on the appropriate records but examination was performed.

[0767] Spare Animal No. 1015 was euthanized rather than transferred to the in-house colony as per study plan.

[0768] There is no documentation present on the surgery records documenting the catheterized animals were individually housed post-surgery.

[0769] On Day 19, the dose infusion time was interrupted for 14 minutes for female TK Animal No. 1528 due to changing of vehicle syringes.

[0770] Between Days 4 and 12, the saline syringe change was not documented on the Saline and Injection Cap Change Record for the toxicokinetic animals.

[0771] On Day 29, toxicokinetic samples were collected from only 2 males, not 3 males/group/time-point in Group 4, due to the euthanasia of the male Animal No. 4107 for welfare reasons on Day 23.

[0772] On Days 29, the following sample was collected more than .+-.3 minutes (the time permitted per SOP) after the indicated time point:

TABLE-US-00089 Animal Scheduled Actual Deviation Number Day/Interval Time Time in Minutes 2029 29-1 hr 10:59 11:04 2

[0773] Various tissues were noted missing at the time of trimming or microscopic examination for various animals and necropsy intervals.

[0774] Due to the timing of the validation study, it was anticipated that stability might not have been established before the first day of dosing, therefore the formulation preparation was changed to daily preparations rather than weekly as per original study plan. The Day 1 dose formulation was prepared on the day before dosing thereby deviating from the study plan in place at that time. Once stability was established, formulation preparation was changed back to weekly preparations.

[0775] Some dose intervals had inconsistencies in the dose accountability, resulting in deviations from the study plan in that animals received more or less than the intended dose (Table 84A). These may or may not have resulted in out of range dose accountability criteria which are .+-.10.0%.

TABLE-US-00090 TABLE 84A Samples with inconsistencies in dose accountability. Infusion Theoretical Actual Percent Animal Rate Volume Volume Difference Number Dose No. (ml/hr) (ml) (ml) (%) Code *3566 Dose 1 2.51 5.05 5.00 -0.40 A 4061 Dose 8 4.08 8.16 9.76 19.61 B 3046 Dose 5 4.04 8.08 10.25 26.86 B 3551 Dose 5 2.47 4.94 4.99 1.01 A 2037 Dose 6 3.55 7.10 8.21 15.63 B 2030 Dose 6 4.04 8.08 7.21 -10.77 B 1511 Dose 9 2.67 5.34 1.07 -79.96 C 1519 Dose 9 2.79 5.58 6.22 11.47 B 4063 Dose 9 4.00 8.00 10.96 37.00 B *TK animal A = Catheter found leaking for an indeterminable amount of time B = No apparent reason C = Tail vein catheter difficulties; dosing stopped due to welfare reasons

[0776] On one occasion for some animals dosing was interrupted or pump settings needed to be adjusted affecting the accountability documentation. However, variations were less than 10% deviation from desired dose. Additionally, Animal No. 1017 (TK) did not receive the last 2 minutes of saline catheter flush.

[0777] On the day of terminal necropsy, Animal Nos. 1010, 3047, 3048, 3049, 3057, 2538, 2539 and 2594 were given feed for welfare reasons after being fasted close to 26 hours.

Analytical Chemistry

[0778] Analysis of preliminary mixes confirmed that the test item was stable in the vehicle for at least 24 hours at room temperature (nominally 20.degree. C.) and for at least 8 days at 2 to 8.degree. C. (under storage conditions used in this study). Analyses conducted during the treatment period confirmed that with the exception of the Day 1, 1 mg/mL (Group 2) formulation, dose solutions of appropriate concentration were administered. The Day 1, 1 mg/mL dose solution was 86.7% of nominal. Without wishing to be bound by theory, it was concluded that due to interactions at the meniscus, the volume collected for dose formulation analysis may have lowered the analysis results. The volume collected for analysis was increased for subsequent weekly preparations and confirmed to be of appropriate concentration.

TABLE-US-00091 TABLE 85 Mean analytical concentrations Day 1 NOMINAL ANALYTICAL CONCENTRATION CONCENTRATION GROUP (ppm) OR (mg/mL) (% of Nominal) 2 1 86.7 2 Back-up 1 76.9 2 Repeat 1 75.5 Preparation 3 4 95.7 4 15 100.7

TABLE-US-00092 TABLE 86 Mean analytical concentrations Week 2 NOMINAL ANALYTICAL CONCENTRATION CONCENTRATION GROUP (ppm) OR (mg/mL) (% of Nominal) 2 1 90.1 2 Back-up 1 86.8 3 4 95.4 4 15 102.9

TABLE-US-00093 TABLE 87 Mean analytical concentrations Week 3 NOMINAL ANALYTICAL CONCENTRATION CONCENTRATION GROUP (ppm) OR (mg/mL) (% of Nominal) 2 1 93.2 3 4 94.8 4 15 100.2

TABLE-US-00094 TABLE 88 Mean analytical concentrations Week 4 NOMINAL ANALYTICAL CONCENTRATION CONCENTRATION GROUP (ppm) OR (mg/mL) (% of Nominal) 2 1 92.8 3 4 96.9 4 15 101.0

TABLE-US-00095 TABLE 89 Mean analytical concentrations Week 5 NOMINAL ANALYTICAL CONCENTRATION CONCENTRATION GROUP (ppm) OR (mg/mL) (% of Nominal) 2 1 94.9 3 4 97.7 4 15 101.1

Brincidofovir

[0779] In the brincidofovir-treated animals on Day 1 peak plasma concentrations of brincidofovir occurred at 1 to 2 hours post start of infusion with mean concentrations ranging from 215 ng/mL to 3670 ng/mL for doses ranging from 1 to 15 mg/kg. Plasma concentrations of brincidofovir remained quantifiable at the 8-hour time point for the 4 and 15 mg/kg dose groups, but only through the 2-hour time point for the 1 mg/kg dose group. Plasma concentrations of brincidofovir increased as the dose increased from 1 to 15 mg/kg on Day 1.

TABLE-US-00096 TABLE 90 Summary of combined male and female plasma brincidofovir toxicokinetic parameters by treatment on Day 1 in rats (TK parameter population) AUC.sub.last/Dose C.sub.max C.sub.max/Dose T.sub.max C.sub.last T.sub.last AUC.sub.last ((hr*ng/mL)/ Treatment Statistic (ng/mL) (kg*ng/mL/mg) (hr) (ng/mL) (hr) (hr*ng/mL) (mg/kg)) 1 mg/kg N 6 6 6 6 6 6 6 Mean 219 219 NC 209 NC 319 319 SD 39.8 39.8 NC 39.5 NC 58.4 58.4 CV % 18.2 18.2 NC 18.9 NC 18.3 18.3 Min 171 171 1.00 157 2.00 249 249 Median 210 210 1.00 206 2.00 305 305 Max 271 271 2.00 271 2.00 398 398 4 mg/kg N 6 6 6 6 6 6 6 Mean 1010 252 NC 2.20 NC 2215 554 SD 159 39.8 NC 0.819 NC 717 179 CV % 15.8 15.8 NC 37.2 NC 32.3 32.3 Min 825 206 1.00 1.26 8.00 1340 335 Median 1000 250 2.00 2.31 8.00 2317 579 Max 1220 305 2.00 3.38 8.00 3056 764 15 mg/kg N 6 6 6 6 6 6 6 Mean 3800 254 NC 9.68 NC 8815 588 SD 565 37.7 NC 3.56 NC 1260 83.8 CV % 14.9 14.9 NC 36.7 NC 14.3 14.3 Min 2950 197 1.00 5.06 8.00 7368 491 Median 3760 251 1.00 10.3 8.00 8777 585 Max 4460 297 2.00 13.6 8.00 10565 704 NC = Not calculated; For the 1 mg/kg dose group, AUC.sub.last values were calculated using only 2 quantifiable data points.

[0780] In the Day 29 brincidofovir-treated animals, peak plasma concentrations of brincidofovir occurred at 1 to 2 hours post start of infusion with mean concentrations ranging 297 ng/mL to 4500 ng/mL for doses ranging from 1 to 15 mg/kg. Plasma concentrations of brincidofovir remained quantifiable at the 8 hour time point for the 4 and 15 mg/kg dose groups and for 1 of 6 animals in the 1 mg/kg dose group. For the 1 mg/kg dose group, plasma concentrations were quantifiable at the 2-hour time point for all animals. For the majority of the animals, there were no quantifiable concentrations at the 24 or 48-hour time points at any dose administered; exclusive of one animal in the 4 mg/kg dose group that showed measurable concentrations out to 48 hours. Plasma concentrations of brincidofovir increased as the dose increased from 1 to 15 mg/kg on Day 29.

TABLE-US-00097 TABLE 91 Summary of combined male and female plasma brincidofovir toxicokinetic parameters by treatment on Day 29 in rats (TK parameter population) AUC.sub.last/Dose C.sub.max C.sub.max/Dose T.sub.max C.sub.last T.sub.last AUC.sub.last ((hr*ng/mL)/ Treatment Statistic (ng/mL) (kg*ng/mL/mg) (hr) (ng/mL) (hr) (hr*ng/mL) (mg/kg)) 1 mg/kg N 6 6 6 6 6 6 6 Mean 298 298 NC 126 NC 410 410 SD 130 130 NC 88.2 NC 190 190 CV % 43.7 43.7 NC 69.9 NC 46.4 46.4 Min 52.6 52.6 1.00 1.12 2.00 73.9 73.9 Median 324 324 1.00 144 2.00 434 434 Max 421 421 2.00 229 8.00 653 653 4 mg/kg N 6 6 6 6 6 6 6 Mean 1200 301 NC 2.06 NC 2759 690 SD 297 74.3 NC 0.519 NC 851 213 CV % 24.7 24.7 NC 25.2 NC 30.8 30.8 Min 935 234 1.00 1.38 8.00 2119 530 Median 1160 290 1.00 2.14 8.00 2549 637 Max 1700 425 2.00 2.67 48.00 4415 1104 15 N 5 5 5 5 5 5 5 mg/kg Mean 4500 300 NC 10.9 NC 9893 660 SD 742 49.5 NC 4.14 NC 1840 122 CV % 16.5 16.5 NC 37.9 NC 18.6 18.6 Min 3550 237 1.00 7.75 8.00 8001 533 Median 4460 297 1.00 8.90 8.00 9528 635 Max 5600 373 1.00 18.0 8.00 12928 862 NC = Not calculated; For the 1 mg/kg dose group, AUC.sub.last values were calculated using only 2 quantifiable data points.

Brincidofovir Metabolite Cidofovir

[0781] In the brincidofovir-treated animals on Day 1, individual plasma concentrations of the metabolite CDV were generally not quantifiable with the first sample collected after initiation of dosing (1-hour post-start of the infusion) in the 1 mg/kg (0 of 6 rats) and 4 mg/kg (1 of 6 rats) dose groups. However, individual plasma concentrations of CDV were quantifiable at the 1-hour sample collection time point in 5 of 6 rats in the 15 mg/kg dose group.

[0782] On Day 29, individual plasma concentrations of the metabolite CDV were generally not quantifiable with the first sample collected after initiation of dosing (1-hour post-start of the infusion) in the 1 mg/kg (1 of 6 rats) dose group. However, individual plasma concentrations of CDV were quantifiable at the 1-hour sample collection time point in 4 of 6 rats in the 4 mg/kg and 5 of 5 rats in the 15 mg/kg dose group.

[0783] Plasma concentrations of CDV generally remained quantifiable at the 24-hour time point for the 4 mg/kg dosed animals, and at the 48-hour time point for the 15 mg/kg dosed animals, but only through the 8-hour time point for the 1 mg/kg dose group. Plasma concentrations of CDV generally increased as the brincidofovir dose increased from 1 to 15 mg/kg.

TABLE-US-00098 TABLE 92 Summary of combined male and female plasma CDV toxicokinetic parameters by treatment on Day 1 in rats (TK parameter population) AUC.sub.last Treat- Sta- C.sub.max T.sub.max C.sub.last T.sub.last T.sub.lag (hr*ng/ ment tistic (ng/mL) (hr) (ng/mL) (hr) (hr) mL) 1 N 6 5 5 5 5 6 mg/kg Mean 5.64 NC 5.66 NC NC 27.3 SD 2.96 NC 0.486 NC NC 20.2 CV % 52.4 NC 8.6 NC NC 74.1 Min 0 2.00 5.01 2.00 1.00 0 Median 6.06 2.00 5.87 8.00 1.00 37.8 Max 8.60 8.00 6.18 8.00 1.00 45.2 4 N 6 6 6 6 6 6 mg/kg Mean 18.0 NC 7.69 NC NC 280 SD 5.85 NC 1.12 NC NC 68.7 CV % 32.5 NC 14.5 NC NC 24.5 Min 11.2 2.00 6.52 24.00 0 204 Median 16.7 8.00 7.54 24.00 1.00 258 Max 27.8 8.00 9.62 24.00 1.00 394 15 N 6 6 6 6 6 6 mg/kg Mean 32.1 NC 7.29 NC NC 825 SD 7.86 NC 1.32 NC NC 122 CV % 24.5 NC 18.0 NC NC 14.8 Min 23.5 8.00 5.53 48.00 0 699 Median 33.0 8.00 7.42 48.00 0 817 Max 39.6 8.00 9.18 48.00 1.00 973 NC = Not calculated; For the 1 mg/kg dose group, AUC.sub.last values were calculated using only 2 quantifiable data points.

TABLE-US-00099 TABLE 93 Summary of combined male and female plasma CDV toxicokinetic parameters by treatment on Day 29 in rats (TK parameter population) AUC.sub.last Treat- Sta- C.sub.max T.sub.max C.sub.last T.sub.last T.sub.lag (hr*ng/ ment tistic (ng/mL) (hr) (ng/mL) (hr) (hr) mL) 1 N 6 5 5 5 5 6 mg/kg Mean 7.81 NC 8.31 NC NC 38.6 SD 4.18 NC 1.90 NC NC 28.9 CV % 53.5 NC 22.9 NC NC 75.0 Min 0 2.00 5.24 2.00 0 0 Median 8.41 2.00 8.61 8.00 1.00 43.0 Max 12.5 8.00 10.4 8.00 1.00 74.8 4 N 6 6 6 6 6 6 mg/kg Mean 20.4 NC 8.76 NC NC 399 SD 9.41 NC 1.69 NC NC 248 CV % 46.2 NC 19.3 NC NC 62.3 Min 10.6 8.00 6.47 24.00 0 195 Median 20.0 8.00 8.92 24.00 0 347 Max 37.3 8.00 10.7 48.00 1.00 883 15 N 5 5 5 5 5 5 mg/kg Mean 41.3 NC 8.05 NC NC 1068 SD 16.0 NC 1.04 NC NC 327 CV % 38.8 NC 13.0 NC NC 30.6 Min 23.6 2.00 6.99 48.00 0 732 Median 44.0 8.00 7.81 48.00 0 1117 Max 60.9 8.00 9.40 48.00 0 1473 NC = Not calculated; For the 1 mg/kg dose group, AUC.sub.last values were calculated using only 2 quantifiable data points. N = 5 in for the 15 mg/kg dose group because animal 4107 was euthanized prior to the collection of TK samples on Day 29.

Mortality

Dosing Phase

[0784] There were three unscheduled deaths on the study; none of which were considered to be brincidofovir-related. The causes of death in two animals (Tox Animal Nos. 2595 and 1005) were determined not to be brincidofovir-related. The cause of death in the third animal (TK Animal No. 4107) could not be determined.

[0785] Animal No. 2595 (toxicity female at 1 mg/kg/dose) was euthanized for welfare reasons on Day 23 because of a swollen cranium which was likely related to accidental trauma. Dark areas on the cranium observed macroscopically correlated microscopically with multifocal hemorrhage. On Day 23, this rat was abnormally cold to touch and displayed decreased activity and abnormal (labored and irregular) breathing.

[0786] Animal No. 4107 (toxicokinetics male at 15 mg/kg/dose) was euthanized for welfare reasons on Day 23. On Day 23, this rat was abnormally cold to touch and exhibited decreased activity, partially closed eyelids and dull eyes, and irregular breathing. Enlarged liver and spleen, and correct catheter tip placement were noted at gross necropsy. The cause of death could not be determined from macroscopic data and microscopic evaluations were not conducted to further determine cause of death.

[0787] Animal No. 1005 (toxicity male control) was found dead on Day 27. On Day 27, this animal had decreased activity and a red aqueous nasal discharge. Macroscopic and microscopic findings were consistent with bacterial infection that originated at the catheter site and disseminated to multiple other organs.

Recovery Phase

[0788] There was no brincidofovir-related mortality.

Clinical Observations

Dosing Phase

[0789] Administration of brincidofovir at 4 and 15 mg/kg/dose resulted in intermittent clinical signs in some animals (both sexes). The intermittent clinical signs consisted of rapid breathing and hunched posture with and without piloerection, partially closed eyes, decreased activity, irregular breathing, and/or flattened posture. Manual differential leukocyte counts were performed for verification and absolute values were calculated if necessary, these signs were generally seen following infusion between Days 22 and 29 and were dose-related in incidence.

Recovery Phase

[0790] All clinical signs observed during the treatment phase completely recovered by the first day of the recovery period. There were no brincidofovir-related clinical signs during the recovery phase.

Opthalmology

[0791] There were no brincidofovir-related effects on ophthalmology at the end of dosing recovery phases.

Body Weights

[0792] There were no brincidofovir-related effects on body weight or body weight change.

Food Consumption

Dosing Phase

[0793] There were minimal, statistically significant brincidofovir-related decreases in food consumption in males administered .gtoreq.1 mg/kg/dose on Weeks 1, 2, 3 and/or 4 (to -16% concurrent controls on Week 4), and in females administered 15 mg/kg/dose on Weeks 3 and 4 (to -9.8% concurrent controls on Week 3). The decreases in food consumption had no effect on body weight or body weight gain.

Recovery Phase

[0794] The brincidofovir-related decreases in food consumption observed during the treatment phase were fully reversible as there were no brincidofovir-related effects on food consumption in any animals during the recovery period.

Clinical Pathology

Hematology: Dosing Phase

[0795] There were no test item-related hematology changes.

[0796] Without wishing to be bound by theory, all hematologic abnormalities were considered to be related to the infusion procedure and exhibited no distinct dose-related trends. These included minimal to slight decreases in red cell mass (hemoglobin, hematocrit and red blood cells) accompanied by increases in reticulocytes, increases in platelet counts, and increases in neutrophil and/or monocyte counts. These findings were considered to reflect an appropriate hematopoietic and immune response to catheter vein infection, inflammation, and increased blood loss or destruction.

Hematology: Recovery Phase

[0797] There were no test item-related hematology findings after a 14-day recovery.

Coagulation: Dosing Phase

[0798] There were no test item-related effects on coagulation times.

[0799] Without wishing to be bound by theory, all differences from controls, statistically significant or otherwise, were not considered to be test item-related due to their direction, small magnitude, infrequent occurrence, lack of relation to dose, lack of concordance between related analytes, and/or because values were comparable to the study control range.

Coagulation: Recovery Phase

[0800] After a 14-day recovery, all values were comparable to controls.

Clinical Chemistry

Dosing Phase

[0801] There were no test item-related clinical chemistry changes.

[0802] Without wishing to be bound by theory, all abnormalities were considered to be related to the infusion procedure and exhibited no distinct dose-related trends. These included minimal to moderate increases in aspartate aminotransferase (AST), alanine aminotransferase (ALT), alkaline phosphatase (ALKP), blood urea nitrogen (BUN), creatinine, and globulins and decreases in albumin and albumin to globulin ratio. These findings correlated with microscopic evidence of inflammation, necrosis, abscesses, and infarction in the liver, inflammation, abscesses, urothelial hyperplasia, tubular basophilia and eosinophilic tubular globules in the kidneys, and general inflammation in a variety of other tissues.

Recovery Phase

[0803] There were no test item-related clinical chemistry findings after a 14-day recovery.

Urinalysis

Dosing Phase

[0804] There were no test item-related effects on urinalysis.

[0805] All differences from controls, statistically significant or otherwise, were not considered to be test item-related due to their direction, small magnitude, infrequent occurrence, lack of relation to dose, lack of concordance between related analytes, and/or because values were comparable to the study control range.

Recovery Phase

[0806] After a 14-day recovery, all values were comparable to controls.

Organ Weights

[0807] Brincidofovir-related, minimal to mild, dose-dependent lower testes weights were present in males administered .gtoreq.4 mg/kg/dose compared to controls (Table 94). Lower testes weights correlated microscopically with germ cell depletion. The 15 mg/kg/dose males had minimally lower epididymal weights. Without wishing to be bound by theory, however, since there were no correlative microscopic findings, the lower weights were considered incidental and unrelated to the microscopic changes in the testes.

TABLE-US-00100 TABLE 94 Brincidofovir-related organ weight changes (% difference relative to controls) in rats dosed twice weekly for a 29 day period Group/sex 2M 3M 4M 2F 3F 4F Dose (mg/kg/dose) 1 4 15 1 4 15 Testes Absolute weight (%) -- -9.sup.a -24.sup.a NA NA NA vs. body weight (%) -- -3 -16.sup.a NA NA NA vs. brain weight (%) -- -9.sup.a -22.sup.a NA NA NA Epididymides Absolute weight (%) -- -- -14.sup.a NA NA NA vs. body weight (%) -- -- -6 NA NA NA vs. brain weight (%) -- -- -13.sup.a NA NA NA .sup.aStatistically significant difference between mean values for brincidofovir-treated and control groups. -- = not CMX00-related; NA = Not applicable

[0808] All other organ weight differences at the end of dosing, statistically significant or otherwise, had no microscopic correlations and were considered to be incidental due to biologic variability.

[0809] At the end of the recovery period, testes weights in males administered .gtoreq.4 mg/kg/dose (Table 95) were still lower than controls and differences were of greater magnitude than at the end of dosing. Without wishing to be bound by theory, these changes were, without wishing to be bound by theory, reflective of maturation depletion secondary to the loss of germ cell stages observed microscopically at the end of the dosing period.

TABLE-US-00101 TABLE 95 Brincidofovir-related organ weight changes (% difference relative to controls) in rats dosed twice weekly for a 29 day period and following a 14-day recovery period Group/sex 2M 3M 4M 2F 3F 4F Dose (mg/kg/dose) 1 4 15 1 4 15 Testes Absolute weight (%) -- -21.sup.a -42.sup.a NA NA NA vs. body weight (%) -- -26.sup.a -40.sup.a NA NA NA vs. brain weight (%) -- -18.sup.a -41.sup.a NA NA NA .sup.aStatistically significant difference between mean values for brincidofovir-treated and control. -- = not CMX00-related; NA = Not applicable

[0810] All other organ weight differences had no microscopic correlations and were considered to be incidental and due to biologic variability.

Pathology: Macroscopic

[0811] There were three unscheduled deaths during the dosing phase of the study. Two animals, a control male and a female administered 1 mg/kg/dose, died or was euthanized during the dosing phase from causes unrelated to brincidofovir. A third animal, a toxicokinetics male administered 15 mg/kg/dose, was euthanized during the dosing phase; the cause of death could not be determined. There was no brincidofovir-related mortality during the recovery phase.

[0812] A control male (Animal No. 1005) was found dead on Day 27. The cause of death was sepsis. Macroscopic and microscopic findings were consistent with bacterial infection that originated at the catheter site and disseminated to multiple other organs, resulting in masses in many organs that correlated microscopically with inflammation, abscessation, thrombosis, infarction and necrosis. Inflammation was also present in a swollen extremity (left femorotibial joint). Increased cellularity in the bone marrow was considered to be an adaptive reactive response to infection and decreased cellularity and/or necrosis in the thymus, spleen and lymph nodes, adrenal cortical hypertrophy were considered to be related to physiologic stress.

[0813] One female administered 1 mg/kg/dose (Animal No. 2595) was euthanized on Day 23 because of a swollen cranium which was likely related to accidental trauma. Dark areas on the cranium were observed macroscopically and correlated microscopically with multifocal hemorrhage.

[0814] One toxicokinetics male administered 15 mg/kg/dose (Animal No. 4107) was euthanized on Day 23. Enlarged liver and spleen, and correct catheter tip placement were noted at gross necropsy. The cause of death could not be determined from macroscopic data and microscopic evaluations were not conducted to further determine the cause of death.

Terminal Sacrifice

Brincidofovir-Related Findings

[0815] No brincidofovir-related macroscopic findings were observed at the end of the dosing period.

Procedure-Related Findings

[0816] Most macroscopic findings were related to inflammation in the infusion vein and were present across all groups including the controls. These findings included scabs or thickening of the catheter exteriorization site and abnormal contents, masses, thickening, and/or edema associated with the infusion vein, abnormal color and masses in the kidneys, irregular surface or masses in the liver and dark or pale areas and masses in the lung. Microscopically, these findings correlated with inflammation and necrosis. Enlargement or abnormal color of lymph nodes and spleen correlated microscopically with reactive lymphoid processes (increased follicle cellularity in lymph nodes and spleen), drainage of blood from sites of inflammation (erythrocytosis/erythrophagocytosis in lymph nodes) or extramedullary hematopoiesis (spleen). All these changes were considered to be systemic reactions associated with inflammation related to the infusion site/procedure.

[0817] Without wishing to be bound by theory, all other macroscopic findings occurred sporadically or in control groups and were considered incidental and due to biological variability. Abnormal color (blue) and soft left testis and small left epididymis in one 15 mg/kg/dose male (Animal No. 4063) were considered incidental and spontaneous background findings unrelated to the brincidofovir-related change of testicular germ cell depletion. Sporadic unilateral testicular seminiferous tubular atrophy is not uncommon in rodents.

Recovery Sacrifice

Test Item-Related Findings

[0818] At the end of the recovery period, brincidofovir-related macroscopic findings were limited to the testes and epididymides in the 15 mg/kg/dose males.

[0819] Two of 5 males had bilaterally soft and/or small testes and epididymides. Without wishing to be bound by theory, these findings correlated with germ cell depletion in the testes and decreased sperm in the head region of the epididymides. These findings were reflective of maturation depletion secondary to the loss of germ cell populations observed microscopically at the end of the dosing period. One male (Animal No. 4071) had a unilaterally (left) soft and/or small testis and epididymis which correlated with tubular atrophy in the testis and decreased sperm throughout the epididymis; these unilateral findings in this male were considered to be incidental spontaneous background changes.

Procedure-Related Findings

[0820] Major macroscopic findings related to the infusion procedure included abnormal contents, masses, thickening, and/or edema of the infusion vein, enlarged spleens and an enlarged renal lymph node. These findings correlated microscopically with inflammation and abscess formation of the infusion vein and increased follicular cellularity and plasmacytosis in the lymphoid organs. An enlarged adrenal gland correlated with cortical hypertrophy (related to physiologic stress).

Pathology: Microscopic

[0821] Brincidofovir-related microscopic findings were present in the testes, epididymides, and/or seminal vesicles in males at .gtoreq.4 mg/kg/dose, in the intestinal tract in males and females at .gtoreq.4 mg/kg/dose, and in the sebaceous glands in males and females at 15 mg/kg/dose.

Terminal Sacrifice

Brincidofovir-Related Findings

Male Reproductive Tract (Testes, Epididymides, Seminal Vesicles)

[0822] Brincidofovir-related germ cell depletion was present in the testes in males at .gtoreq.4 mg/kg/dose (Table 96). These changes were bilateral and minimal to slight at 4 mg/kg/dose and slight to moderate at 15 mg/kg/dose with the severity grading generally reflecting the number of cell types affected by the depletion. At 4 mg/kg/dose, germ cell depletion was characterized by a variable loss of spermatogonia through preleptotene spermatocytes. At 15 mg/kg/dose, the germ cell depletion was characterized by a variable loss of spermatogonia through pachytene spermatocytes, often with some sparing of pachytene spermatocytes in late stage tubules and occasional active degeneration of germ cells in late stage tubules.

[0823] Brincidofovir-related findings at 15 mg/kg/dose also included minimal luminal cell debris in the epididymides and minimally increased epithelial apoptosis in the seminal vesicles compared to controls. The latter change was occasionally accompanied by an increase in mitotic figures. Minimally increased epithelial apoptosis was also present in 4 male administered 4 mg/kg/dose.

[0824] Unilateral tubular atrophy observed in one 15 mg/kg/dose male (Animal No. 4063) was considered an incidental background finding that occurs sporadically in laboratory rodents.

TABLE-US-00102 TABLE 96 Brincidofovir-related findings in the male reproductive tract in rats dosed with Brincidofovir twice weekly for a 29 day period Group/sex 1M 2M 3M 4M 1F 2F 3F 4F Dose (mg/kg/dose) 0 1 4 15 0 1 4 15 Number of tissues examined 10 10 10 10 NA NA NA NA Testes Depletion, Germ Cell Minimal -- -- 6 -- NA NA NA NA Slight -- -- 3 5 NA NA NA NA Moderate -- -- -- 5 NA NA NA NA Epididymides Cell Debris, Luminal Minimal -- -- -- 9 NA NA NA NA Seminal Vesicles Apoptosis, Epithelial Minimal -- -- 4 8 NA NA NA NA Mitotic Figures, Epithelial Minimal -- -- -- 4 NA NA NA NA Total Animals Represented 0 0 10 10 NA NA NA NA -- = Finding not present. NA = not applicable

Intestinal Tract

[0825] Single cell necrosis of the crypt epithelium (minimal to moderate) was present in the small intestine (duodenum, jejunum, and/or ileum) in males and females at .gtoreq.4 mg/kg/dose and was dose-dependent with respect to severity and/or incidence (Table 97). This change was occasionally accompanied by minimal crypt epithelial hyperplasia. Minimal single cell necrosis was also present in the glandular epithelium of the large intestine (cecum and/or colon) in males and females at 15 mg/kg/dose.

TABLE-US-00103 TABLE 97 Brincidofovir-related findings in the intestinal tract in rats dosed with brincidofovir twice weekly for a 29 day period Group/sex 1M 2M 3M 4M 1F 2F 3F 4F Dose (mg/kg/dose) 0 1 4 15 0 1 4 15 Number of tissues examined 10.sup.c 10 10 10 10 11.sup.d 10 10 Small intestine.sup.a Necrosis, Epithelium, Crypts Minimal -- -- 8 2 -- -- 9 3 Slight -- -- 1 8 -- -- -- 5 Moderate -- -- -- -- -- -- -- 1 Hyperplasia, Epithelium, Crypts Minimal -- -- 5 2 -- -- -- 5 Large intestine.sup.b Necrosis, Epithelium, glands Minimal -- -- -- 9 -- -- -- 4 Total Animals Represented 0 0 9 10 0 0 9 9 .sup.a, bIncidence of severity grades represent highest grade recorded for duodenum, jejunum and ileum.sup.a and for cecum and colon..sup.b .sup.cDuodenum was autolytic in one animal and not readable. .sup.dIncidence includes one unscheduled death (Animal No. 2595). -- = Finding not present.

Skin (Sebaceous Gland)

[0826] Sebaceous gland atrophy (slight to marked) was present in males and females at 15 mg/kg/dose (Table 98). Marked severity indicated absence of sebaceous glands microscopically with no change in remaining adnexa (hair follicles).

TABLE-US-00104 TABLE 98 Brincidofovir-related findings in the skin in rats dosed with brincidofovir twice weekly for a 29 day period Group/sex 1M 2M 3M 4M 1F 2F 3F 4F Dose (mg/kg/dose) 0 1 4 15 0 1 4 15 Number of tissues examined 10 10 10 10 10 11.sup.a 10 10 Atrophy, Sebaceous Glands Slight -- -- -- 3 -- -- -- 5 Moderate -- -- -- 3 -- -- -- 3 Marked -- -- -- 4 -- -- -- 2 Total Animals Represented 0 0 0 10 0 0 0 10 .sup.aIncidence includes one unscheduled death (Animal No. 2595). -- = Finding not present.

Procedure-Related Findings

[0827] Microscopic findings related to the infusion procedure were observed in the vehicle control group and in animals administered brincidofovir. Without wishing to be bound by theory, many findings were those commonly associated with catheterization, including pericatheteral thrombi, suture granulomas, and intimal proliferation in the catheter vein (Weber, 2011) and perivascular eosinophilic inflammatory cell infiltrates in the lung (Morton, 1997). Some animals had chronic-active inflammation, abscess formation and thrombosis with bacterial colonies in the catheter vein and epidermal necrosis and/or inflammatory tracts and abscesses in the skin at the catheter exteriorization site. Chronic-active inflammation, abscess formation, infarction and/or necrosis were occasionally present in other organs, primarily the kidney and liver. Without wishing to be bound by theory, these changes were likely the result of bacterial infection of the catheter vein. Findings indicative of an immune response to infection included overall increased cellularity or increased granulocytes in the bone marrow, increased follicular cellularity and plasmacytosis in lymph nodes and increased follicular cellularity, increased macrophages and/or extramedullary hematopoiesis in the spleen. Other findings in the lymphoid organs, such as decreased cellularity and necrosis, as well as cortical hypertrophy in the adrenal glands and vaginal mucification were considered related to stress.

[0828] Additional procedure-related microscopic findings were also observed in the stomach (serosal inflammation), heart (valvular endothelial hypertrophy), and lung (granulomas, emboli, macrophage aggregates, alveolar and vascular inflammation, and/or vascular intimal proliferation). Instances of minimal focal/multifocal myocardial inflammation were also observed and, without wishing to be bound by theory, could have been secondary to the infusion procedures or could have represented common background findings in laboratory rodents.

Miscellaneous Findings

Kidney

[0829] Eosinophilic globules were present in the cortical tubular epithelium of the kidney in a male (Animal No. 3048) and a female (Animal No. 3560) dosed at 4 mg/kg/dose. There were no associated inflammatory or degenerative changes. These globules exhibited negative staining with Prussian blue for iron and Martius Scarlet Blue as a general stain for erythrocytes.

[0830] All other microscopic findings occurred sporadically or at similar incidence and severity in control and brincidofovir groups and without wishing to be bound by theory were considered incidental and due to biological variability. These included a poorly differentiated carcinoma of the salivary gland in one 4 mg/kg/dose male (Animal No. 3044) and a renal tubular adenoma in a 15 mg/kg/dose male (Animal No. 4068). Without wishing to be bound by theory, these tumors have been reported to occur spontaneously in rats and were considered incidental (Nishikawa 2010, Hardisty 2013) and unrelated to brincidofovir.

[0831] Following the 14-day recovery period, microscopic findings remained present in the testes and/or epididymides in males at .gtoreq.4 mg/kg/dose, and a new finding noted in the bone marrow in males and females at 15 mg/kg/dose. There was complete recovery of all microscopic findings in the intestinal tract and sebaceous glands observed at the end of dosing. Infusion procedure-related findings were also present at the end of the recovery period.

Male Reproductive Tract (Testes, Epididymides)

[0832] At the end of the recovery period, there was continued depletion of the cell populations identified in terminal necropsy animals along with maturation depletion in the testes in males at .gtoreq.4 mg/kg/dose (Table 99). At 15 mg/kg/dose, this change was characterized by a depletion of most germ cell populations generally sparing only elongating spermatids often with multifocal tubular atrophy. At 4 mg/kg/dose, there was variable loss of spermatogonia, spermatocytes, and round spermatids in early stage tubules through about stage XIII tubules.

[0833] In the males at 15 mg/kg/dose, maturation depletion was accompanied by reduced sperm and luminal cell debris in the head of the epididymides. One 15 mg/kg/dose male (Animal No. 4071) had unilateral tubular atrophy in the testes accompanied by reduced sperm with cribiform change (a finding related to reduced sperm) in the epididymis. Without wishing to be bound by theory, these unilateral findings in this male were considered spontaneous background changes and unrelated to brincidofovir.

TABLE-US-00105 TABLE 99 Test item-related findings in the male reproductive tract in rats dosed with brincidofovir twice weekly for a 29 day period and following a 14-day recovery period Group/sex 1M 2M 3M 4M 1F 2F 3F 4F Dose (mg/kg/dose) 0 1 4 15 0 1 4 15 Number of tissues examined 5 5 5 5 NA NA NA NA Testes Depletion, Germ Cell Minimal -- -- 1 -- NA NA NA NA Slight -- -- 4 -- NA NA NA NA Moderate -- -- -- 1 NA NA NA NA Marked -- -- -- 4 NA NA NA NA Epididymides Sperm, Reduced, Luminal Marked -- -- -- 4 NA NA NA NA Severe -- -- -- 1 NA NA NA NA Cell Debris, Luminal Minimal -- -- -- 5 NA NA NA NA Total Animals Represented 0 0 5 5 NA NA NA NA -- = Finding not present. NA = not applicable

Bone Marrow

[0834] At the 15 mg/kg/dose, a few recovery males and females had minimal to slight decreased cellularity (Table 100) in the bone marrow, a finding which was not observed at the end of the dosing period.

TABLE-US-00106 TABLE 100 Test item-related findings in the bone marrow in rats dosed with brincidofovir twice weekly for a 29 day period and following a 14-day recovery period Group/sex 1M 2M 3M 4M 1F 2F 3F 4F Dose (mg/kg/dose) 0 1 4 15 0 1 4 15 Number of tissues examined 5 5 5 5 5 4 5 5 Cellularity, Decreased.sup.a Minimal -- -- -- -- -- -- -- 2 Slight -- -- -- 3 -- -- -- -- Total Animals Represented 0 0 0 3 0 0 0 2 .sup.aIncidence of severity grades represent highest grade recorded for sternal or femoral bone marrow. -- = Finding not present.

Procedure-Related Findings

[0835] At the end of the recovery period, a range of procedural-related microscopic observations were present similar to those observed at the end of the dosing period and described above These were related to infection of the infusion catheter and without wishing to be bound by theory were not considered to be related to brincidofovir.

[0836] All other microscopic findings occurred sporadically or at similar incidence and severity in control and brincidofovir groups and were considered incidental and due to biological variability.

[0837] Brincidofovir was administered twice weekly for 29 days via 2-hour intravenous infusion to Sprague-Dawley CD.RTM. rats (15/sex/group) at 0 (2.times. Sodium Phosphate Buffer Solution), 1, 4 or 15 mg/kg/dose. Up to 5 animals/sex/group were held for a 14-day post-dosing recovery.

[0838] Administration of brincidofovir resulted in intermittent (generally following infusion) clinical signs in some animals (primarily abnormal breathing and hunched posture) in both sexes at .gtoreq.4 mg/kg/dose and minimally decreased food consumption in males and females at .gtoreq.1 mg/kg/dose and 15 mg/kg/dose, respectively.

[0839] Brincidofovir-related findings were present in the male reproductive tract (testicular germ cell depletion, luminal cell debris in the epididymides and increased epithelial apoptosis in the seminal vesicles) and intestinal tract (single cell necrosis) at .gtoreq.4 mg/kg/dose and sebaceous glands (atrophy) at 15 mg/kg/dose. All brincidofovir-related findings completely recovered with the exception of testicular changes in males at .gtoreq.4 mg/kg/dose, which would not be expected to fully recover in a 2 week period. In addition, a few animals administered 15 mg/kg/dose had decreased bone marrow cellularity and unilaterally or bilaterally soft and/or small testes and epididymides at the end of recovery.

[0840] Under the conditions of the study, based on the testicular germ cell depletion in males at .gtoreq.4 mg/kg/dose and moderate intestinal single cell necrosis in 1 female at 15 mg/kg/dose, the no-observed-adverse-effect level (NOAEL) for brincidofovir was considered to be 1 mg/kg/dose in males and 4 mg/kg/dose for in females.

Example 11--IV BCV Ascending Dose Study in Healthy Human Subjects

Summary

[0841] Brincidofovir was administered to healthy subjects at 10 mg, 25 mg and 50 mg. IV brincidofovir at 10 mg was found to provide similar exposure as orally administered brincidofovir at 100 mg. It was found that both IV brincidofovir doses (10 mg and 25 mg) were generally safe and well-tolerated. No drug related adverse events, no gastrointestinal adverse events, and no graded lab abnormalities (e.g., no hematologic toxicity and no kidney toxicity) were observed.

[0842] This study also evaluated the peripheral blood mononuclear cell (PBMC) PK of CDV-PP following IV and oral suspension BCV administration. BCV is converted intracellularly to active cidofovir diphosphate (CDV-PP).

[0843] As background, a single dose of oral BCV at 100 mg and 200 mg resulted in about 5% instance of diarrhea, and a 350 mg dose resulted in about 20% instance of diarrhea.

Materials and Methods

[0844] In this double-blind study, subjects were randomized 3:1 to receive IV BCV or placebo in sequential single ascending dose cohorts (Tables 101 and 102, below). Plasma PK samples were collected over 7 days and assayed by HPLC-MS. Plasma BCV PK parameters were determined by non-compartmental analysis and dose proportionality was assessed. Safety assessments were collected over 14 days.

TABLE-US-00107 TABLE 101 Study Design IV Infusion Cohort N Actual Doses Duration 1 6 active and 2 placebo BCV 10 mg or placebo 2 hours 2 6 active and 2 placebo BCV 25 mg or placebo 2 hours 3 9 active and 3 placebo BCV 50 mg or placebo 2 hours 4 9 active and 3 placebo BCV 50 mg or placebo 4 hours

TABLE-US-00108 TABLE 102 Subject Demographics Cohort Male Sex Age (years) 1 8/8 (100%) 26 (18-34) 2 8/8 (100%) 23 (20-37) 3 12/12 (100%) 21 (18-26) 4 12/12 (100%) 26 (18-46) Male sex presented as n/N (%) Age presented as mean (minimum-maximum)

PBMC PK of CDV-PP

[0845] Two single dose BCV studies in healthy subjects contributed data for this analysis. The first study was a 2-period crossover bioequivalence study that enrolled 24 subjects who received two formulations of BCV oral suspension 100 mg fasted, one in each period. Data from 12 subjects who had PBMCs collected in Period 1 were included in this analysis; these subjects were 100% female, 31-59 years, and 58-88 kg. The second study, outlined above, enrolled 40 subjects who received IV BCV or placebo. Data from 18 subjects who received BCV 50 mg as either a 2-h or 4-h IV infusion were included in this analysis; these subjects were 100% male, 18-46 years, 64-106 kg. Serial PBMC samples were collected over 14 days and assayed by HPLC-MS-MS. PBMC CDV-PP PK parameters were determined by non-compartmental analysis.

[0846] Forty healthy male subjects (18-46 y, 83% White) were enrolled and completed the study. Plasma BCV C.sub.max and AUC.sub..infin. increased in proportion to dose (Table 103, below). Adverse events and alanine aminotransferase (ALT) elevations were dose- and infusion duration-related. Gastrointestinal adverse events were mild. All adverse events and ALT elevations were transient and no serious adverse events occurred. No significant differences between placebo and IV BCV in other chemistries or hematologic parameters were observed.

[0847] FIG. 6 shows a plot of the plasma brincidofovir concentration (AUC.sub.inf (ng*h/mL)) at different 100 mg oral administration, 10 mg IV administration, and 25 mg IV administration. As shown in FIG. 6, IV administration of 10 mg brincidofovir provided substantially the same plasma concentration as oral administration of 100 mg brincidofovir, while IV administration of 25 mg brincidofovir provided higher plasma concentration than both the IV 10 mg dose and the oral 100 mg dose.

[0848] A dose-related, reversible ALT increase was observed with IV BCV. In prior studies, oral BCV had reversible dose-related ALT increases in humans and preclinical studies. In the IV BCV SAD study, at therapeutic IV exposures (cohorts 1 and 2), no grade 2 or higher ALT elevations were observed. At supratherapeutic exposures (cohorts 3 and 4), transient grade 2-3 ALT elevations were noted in cohort 4 only. No Grade 2 or higher bilirubin was observed in any cohort.

TABLE-US-00109 TABLE 103 Preliminary Summary of Plasma BCV PK Data following IV infusion Cohort 1 Cohort 2 Cohort 3 Cohort 4 BCV 10 mg BCV 25 mg BCV 50 mg BCV 50 mg Pooled 2 h Infusion 2 h Infusion 2 h Infusion 4 h Infusion Placebo (n = 6) (n = 6) (n = 9) (n = 9) (n = 10) PK C.sub.max (ng/mL) 613 (25%) 1412 (27%) 2952 (19%) 1586 (14%) NA [439-799] [973-1900] [2200-3940] [1360-2180] AUC.sub..infin. (ng h/mL) 1312 (26%) 2889 (37%) 5948 (19%) 6570 (15%) NA [875-1710] [1742-4790] [4446-7274] [5347-9048] t.sub.1/2 (h) 3.42 (27%) 5.89 (23%) 6.54 (49%) 5.10 (13%) NA [2.70-5.76] [4.06-8.00] [3.98-18.09] [4.23-6.33] CL (L/h) 7.62 (26%) 8.65 (37%) 8.41 (19%) 7.61 (15%) NA [5.85-11.4] [5.22-14.4] [6.87-11.3] [5.53-9.35] V.sub.SS (L) 16.4 (24%) 19.8 (25%) 19.5 (21%) 10.8 (19%) NA [12.0-22.2] [15.0-30.3] [13.9-24.9] [7.67-14.5] Drug-Related AEs Diarrhea 0 0 1 (11%) 3 (33%) 0 Nausea 0 0 0 2 (22%) 0 Decreased appetite 0 0 0 1 (11%) Headache 0 0 2 (22%) 2 (22%) 0 Pain, phlebitis at infusion site 0 0 1 (11%) 0 0 Elevated liver transaminases.sup.1 0 0 0 1 (11%) 0 C.sub.max and AUC.sub..infin. presented as geometric mean (% CVb) [minimum-maximum] T.sub.max was 2 hours for Cohorts 1-3 (2-hour infusion) and 2.5 to 4 hours (4-hour infusion) .sup.1ALT >2x ULN in 2 BCV 50 mg 4 h infusion and 1 placebo subjects; 1 ALT elevation considered an AE 2. In Cohort 3, 5 drug-related AEs in 4 subjects. 3. In Cohort 4, 9 drug-related AEs in 5 subjects.

TABLE-US-00110 TABLE 104 IV BCV PK and Safety: Grade 3 or 4 Drug-Related Adverse Events Cohort 1 Cohort 2 Cohort 3 Cohort 4 BCV 10 mg BCV 25 mg BCV 50 mg BCV 50 mg Pooled 2 h Infusion 2 h Infusion 2 h Infusion 4 h Infusion Placebo (n = 6) (n = 6) (n = 9) (n = 9) (n = 10) Grade 3 or 4 Drug-Related AEs Diarrhea 0 0 0 0 0 Nausea 0 0 0 0 0 Decreased appetite 0 0 0 0 Headache 0 0 0 0 0 Pain, phlebitis at 0 0 0 0 0 infusion site Elevated liver 0 0 0 1 (11%) 0 transaminases.sup.1

[0849] FIG. 9 shows a mean plasma BCV concentration as a function of time for subjects in cohorts 1-4. FIG. 9 also shows mean plasma BCV concentration as a function of time for subjects administered BCV orally.

[0850] Table 105 shows a summary of plasma BCV PK data following IV and oral dosing (Single dose administration).

TABLE-US-00111 TABLE 105 Summary of Plasma BCV PK Data Following IV and Oral Dosing 10 mg 25 mg 50 mg 50 mg 100 mg 200 mg 200 mg 350 mg Plasma 2 h IV 2 h IV 2 h IV 4 h IV Oral Oral Oral Oral BCV PK infusion infusion infusion infusion Tablet Tablet Tablet Tablet Parameter N = 6 N = 6 N = 9 N = 9 N = 52 N = 15 N = 63 N = 70 C.sub.max 613 1412 2952 1586 258 622 802 1482 (ng/mL) (25%) (27%) (19%) (14%) (42%) (33%) (38%) (34%) [439- [973- [2200- [1360- [97.0- [384- [344- [732- 799] 1900] 3940] 2180] 586] 1030] 1980] 3600] AUC.sub.inf 1312 2889 5948 6570 1380 3307 3744 6938 (ng h/mL) (26%) (37%) (19%) (15%) (41%) (35%) (35%) (31%) [875- [1742- [4446- [5347- [555- [1741- [1605- [3293- 1710] 4790] 7274] 9048] 3070] 5190] 8227] 14132] t1/2 (h) 3.42 5.89 6.54 5.10 7.62 12.0 -- -- (27%) (23%) (49%) (13%) (47%) (27%) [2.70- [4.06- [3.98- [4.23- [3.05- [3.93- 5.76] 8.00] 18.09] 6.33] 24.1] 17.7] Data presented as geometric mean (CVb %) [minimum, maximum] AUC.sub.last presented in place of AUC.sub.inf for Study CMX001-108

[0851] Table 106 shows a summary of preliminary plasma CDV PK data cohorts 1-4.

TABLE-US-00112 TABLE 106 Preliminary Summary of Plasma CDV PK Data following IV infusion 10 mg Single 25 mg Single 50 mg Single 50 mg Single Plasma Dose 2 h Dose 2 h Dose 2 h Dose 4 h CDV PK IV infusion IV infusion IV infusion IV infusion Parameter N = 6 N = 6 N = 9 N = 9 C.sub.max ND 5.94 (22%) 12.7 (19%) 12.1 (14%) (ng/mL) [4.62-7.83] [9.44-16.6] [9.83-15.6] AUC.sub.last ND 189 (81%) 652 (38%) 654 (30%) (ng h/mL).sup.5 [77.6-419] [341-1367] [477-1151] T.sub.max (h) ND 9.0 10.0 10.0 [8.0-10.0] [8.00-12.0] [8.00, 10.00] Data presented as geometric mean (CVb %) [minimum-maximum] ND: not done because majority of CDV concentrations were below the lower limit of quantification AUC.sub.inf and t1/2 not reported due to high AUC percent extrapolated

[0852] FIG. 10 shows median plasma CDV concentration as a function of time following IV and Oral BCV Doses.

PBMC PK of CDV-PP

[0853] Single doses of BCV 50 mg administered as 2- or 4-h IV infusions delivered intracellular CDV-PP exposures comparable to BCV 100 mg oral suspension (Table 107). PBMC CDV-PP exposure was inversely related to infusion duration (Table 107).

[0854] FIG. 8 shows the mean (+/-SE) ALT levels observed in the IV BCV SAD trial.

TABLE-US-00113 TABLE 107 Summary of PBMC CDV-PP PK following BCV Oral and IV Dosing BCV 100 mg BCV 50 mg IV PBMC CDV-PP PK Oral suspension 2 h Infusion 4 h Infusion Parameter (n = 12) (n = 9) (n = 9) C.sub.max (pg/10.sup.6 cells) 7.68 (41%) 14.3 (63%) 8.41 (57%) AUC.sub.last (pg h/ 1107 (50%) 1409 (36%) 963 (36%) 10.sup.6 cells)

[0855] Single doses of BCV 10-50 mg administered as a 2 h IV infusion were well tolerated and not associated with significant clinical or laboratory abnormalities. BCV IV 10 mg and BCV IV 50 mg achieved geometric mean plasma BCV AUC.sub..infin. similar to and 4.5-fold, respectively, values achieved with BCV oral 100 mg tablets (C.sub.max=251 ng/mL and AUC.sub..infin.=1394 ngh/mL). These data support evaluation of repeat dose administration in healthy subjects and virally-infected patients.

[0856] Additionally, IV BCV provided comparable PBMC CDV-PP exposures with lower doses when compared with oral BCV. Without wishing to be bound by theory, in some embodiments this can allow for improvements in BCV tolerability and efficacy.

EQUIVALENTS

[0857] While the present disclosure has been described in conjunction with the specific embodiments set forth above, many alternatives, modifications and other variations thereof will be apparent to those of ordinary skill in the art. All such alternatives, modifications and variations are intended to fall within the spirit and scope of the present disclosure.

* * * * *

US20200138835A1 – US 20200138835 A1

uspto.report is an independent third-party trademark research tool that is not affiliated, endorsed, or sponsored by the United States Patent and Trademark Office (USPTO) or any other governmental organization. The information provided by uspto.report is based on publicly available data at the time of writing and is intended for informational purposes only.

While we strive to provide accurate and up-to-date information, we do not guarantee the accuracy, completeness, reliability, or suitability of the information displayed on this site. The use of this site is at your own risk. Any reliance you place on such information is therefore strictly at your own risk.

All official trademark data, including owner information, should be verified by visiting the official USPTO website at www.uspto.gov. This site is not intended to replace professional legal advice and should not be used as a substitute for consulting with a legal professional who is knowledgeable about trademark law.

© 2024 USPTO.report | Privacy Policy | Resources | RSS Feed of Trademarks | Trademark Filings Twitter Feed