Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
COMPOSITIONS AND METHODS FOR THE TREATMENT OF FUNGAL INFECTIONS
Document Type and Number:
WIPO Patent Application WO/2019/014333
Kind Code:
A9
Abstract:
The disclosure features non-irritating pharmaceutical compositions containing CD101 in pharmaceutical acceptable salt (e.g., CD101 acetate) or neutral form. The pharmaceutical compositions can be intravenously administered to a subject to treat fungal infections (e.g., candidiasis) in the subject.

Inventors:
BARTIZAL KENNETH (US)
DARUWALA PAUL (US)
HUGHES DAVID (US)
HUGHES MARTIN (US)
MALKAR NAVDEEP (US)
RADHAKRISHNAN BALASINGAM (US)
VAIDYA ANURADHA (US)
Application Number:
PCT/US2018/041617
Publication Date:
March 28, 2019
Filing Date:
July 11, 2018
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
CIDARA THERAPEUTICS INC (US)
International Classes:
A61K41/00
Attorney, Agent or Firm:
BELLIVEAU, Michael J. (US)
Download PDF:
Claims:
CLAIMS

1 . A pharmaceutical composition for intravenous injection comprising an aqueous solution comprising at least 85% (w/w) water, between 0.4 mg/mL and 10 mg/mL CD101 , and an intravenous solubility promoter,

wherein the weight to weight (w/w) ratio of the intravenous solubility promoter to the CD101 in the pharmaceutical composition is at least 2,

the CD101 is in its salt or neutral form;

the pharmaceutical composition has a pH of between 5 and 7, and

the pharmaceutical composition exhibits reduced local irritation upon intravenous administration to a subject.

2. The pharmaceutical composition of claim 1 , wherein the w/w ratio of the intravenous solubility promoter to the CD101 in the pharmaceutical composition is between 2 and 8.

3. The pharmaceutical composition of claim 1 or 2, wherein the intravenous solubility promoter is selected from the group consisting of polysorbate 20 (Tween 20; polyoxyethylene (20) sorbitan monolaurate), polysorbate 40 (Tween40; polyoxyethylene (40) sorbitan monopalmitate), polysorbate 60 (Tween 60; polyoxyethylene (60) sorbitan monostearate), polysorbate 80 (Tween 80; polyoxyethylene (80) sorbitan monooleate), β-cyclodextrin, polyoxyl 35 castor oil (Cremophor EL), polyoxyl 40 hydrogenated castor oil (Cremophor RH 40), polyoxyl 60 hydrogenated castor oil (Cremophor RH 60), D- a-tocopheryl polyethylene glycol 1000 succinate (TPGS), sorbitan monooleate (Span 20), polyoxyl 8 stearate (PEG 400 monosterate), polyoxyl 40 stearate (PEG 1 750 monosterate), PEG 400 caprylic/capric glycerides (Labrasol), PEG 300 oleic glycerides (Labrafil M-1944CS), phosphatidylcholine (lecithin), alkylglucoside, sucrose monolaurate, sucrose monooleate, and polyoxyethylene-polyoxypropylene block copolymer (Poloxamer).

4. The pharmaceutical composition of claim 3, wherein the alkylglucoside is an alkyl monoglucoside.

5. The pharmaceutical composition of claim 4, wherein the alkyl monoglucoside is selected from the group consisting of hexylglucoside, heptaglucoside, octylglucoside, nonaglucoside, decylglucoside, dodecylglucoside, and tetradecylglucoside.

6. The pharmaceutical composition of claim 3, wherein the alkylglucoside is an alkyl diglucoside.

7. The pharmaceutical composition of claim 6, wherein the alkyl diglucoside is selected from the group consisting of hexylmaltoside, heptamaltoside, octylmaltoside, nonamaltoside, decylmaltoside, dodecylmaltoside, and tetradecylmaltoside.

8. The pharmaceutical composition of claim 3, wherein the intravenous solubility promoter is polysorbate 80 (Tween 80).

9. The pharmaceutical composition of any one of claims 1 -8, wherein the concentration of CD101 in the pharmaceutical composition is between 0.4 mg/mL and 4 mg/mL.

10. The pharmaceutical composition of claim 9, wherein the concentration of CD101 in the

pharmaceutical composition is about 0.8 mg/mL.

1 1 . The pharmaceutical composition of claim 9, wherein the concentration of CD101 in the

pharmaceutical composition is about 1 .6 mg/mL.

12. The pharmaceutical composition of any one of claims 1 -1 1 , further comprising a buffer.

13. The pharmaceutical composition of claim 12, wherein the buffer is histidine, citrate, succinate, lactate, propanoate, arginine, tris(hydroxymethyl)aminomethane (tris), glycine, acetate, or formate.

14. The pharmaceutical composition of any one of claims 1 -13, wherein the pharmaceutical composition further comprises between 0.12% to 0.6% (w/w) of a saccharide.

15. The pharmaceutical composition of claim 14, wherein the saccharide is mannitol, sucrose, trehalose, maltose, dextrose, or lactose.

16. The pharmaceutical composition of claim 15, wherein the saccharide is mannitol.

17. The pharmaceutical composition of any one of claims 1 -16, wherein the CD101 in salt form is CD101 acetate.

18. A pharmaceutical composition comprising an effective amount of CD101 and an intravenous solubility promoter in a lyophilized composition, wherein the weight to weight (w/w) ratio of the intravenous solubility promoter to the CD101 in the lyophilized composition is between 2 and 8, the CD101 is in its salt or neutral form, and the lyophilized composition, once reconstituted, provides an aqueous solution having a pH of between 5 and 7.

19. The pharmaceutical composition of claim 18, wherein the intravenous solubility promoter is selected from the group consisting of polysorbate 20 (Tween 20; polyoxyethylene (20) sorbitan monolaurate), polysorbate 40 (Tween40; polyoxyethylene (40) sorbitan monopalmitate), polysorbate 60 (Tween 60; polyoxyethylene (60) sorbitan monostearate), polysorbate 80 (Tween 80; polyoxyethylene (80) sorbitan monooleate), β-cyclodextrin, polyoxyl 35 castor oil (Cremophor EL), polyoxyl 40 hydrogenated castor oil (Cremophor RH 40), polyoxyl 60 hydrogenated castor oil (Cremophor RH 60), D-a-tocopheryl polyethylene glycol 1000 succinate (TPGS), sorbitan monooleate (Span 20), polyoxyl 8 stearate (PEG 400 monosterate), polyoxyl 40 stearate (PEG 1750 monosterate), PEG 400 caprylic/capric glycerides (Labrasol), PEG 300 oleic glycerides (Labrafil M-1944CS), phosphatidylcholine (lecithin), alkylglucoside, sucrose monolaurate, sucrose monooleate, and polyoxyethylene-polyoxypropylene block copolymer (Poloxamer).

20. The pharmaceutical composition of claim 19, wherein the alkylglucoside is an alkyl monoglucoside.

21 . The pharmaceutical composition of claim 20, wherein the alkyl monoglucoside is selected from the group consisting of hexylglucoside, heptaglucoside, octylglucoside, nonaglucoside, decylglucoside, dodecylglucoside, and tetradecylglucoside.

22. The pharmaceutical composition of claim 19, wherein the alkylglucoside is an alkyl diglucoside.

23. The pharmaceutical composition of claim 22, wherein the alkyl diglucoside is selected from the group consisting of hexylmaltoside, heptamaltoside, octylmaltoside, nonamaltoside, decylmaltoside, dodecylmaltoside, and tetradecylmaltoside.

24. The pharmaceutical composition of claim 19, wherein the intravenous solubility promoter is polysorbate 80 (Tween 80).

25. The pharmaceutical composition of any one of claims 18-24, further comprising a buffer.

26. The pharmaceutical composition of claim 25, wherein the buffer is histidine, citrate, succinate, lactate, propanoate, arginine, tris(hydroxymethyl)aminomethane (tris), glycine, acetate, or formate.

27. The pharmaceutical composition of any one of claims 18-26, wherein the pharmaceutical composition further comprises between 2% to 10% (w/w) of a saccharide.

28. The pharmaceutical composition of claim 27, wherein the saccharide is mannitol, sucrose, trehalose, maltose, dextrose, or lactose.

29. The pharmaceutical composition of claim 28, wherein the saccharide is mannitol.

30. The pharmaceutical composition of any one of claims 18-29, wherein the CD101 in salt form is CD101 acetate.

31 . A method of treating or preventing a fungal infection in a subject, comprising intravenously administering the pharmaceutical composition of any one of claims 1 -17 to the subject, wherein the method exhibits reduced local irritation upon intravenous administration of the pharmaceutical composition to a subject.

32. A method of treating or preventing a fungal infection in a subject, comprising:

(i) reconstituting the pharmaceutical composition of any one of claims 18-30 to form an aqueous solution; and

(ii) intravenously administering the aqueous solution to the subject,

wherein the concentration of the CD1 01 in the aqueous solution is between 0.4 mg/mL and 10 mg/mL, the CD101 is in its salt or neutral form, and the method exhibits reduced local irritation upon intravenous administration of the aqueous solution to a subject.

33. The method of claim 32, wherein the concentration of CD101 in the pharmaceutical composition is about 0.8 mg/mL.

34. The method of claim 32, wherein the concentration of CD101 in the pharmaceutical composition is about 1 .6 mg/mL.

35. The method of any one of claims 31 -34, comprising intravenously administering to the subject by infusion.

36. The method of any one of claims 31 -35, comprising intravenously administering to the subject by infusion at a constant infusion rate of between 2 mL/minute and 9 mL/minute.

37. The method of any one of claims 31 -36, comprising intravenously administering to the subject by infusion over 30 to 120 minutes.

38. The method of any one of claims 31 -37, comprising intravenously administering one dose every 5 to 15 days.

39. The method of any one of claims 31 -38, wherein the fungal infection to be treated or prevented is selected from candidemia, invasive candidiasis, tinea capitis, tinea corporis, tinea pedis, onychomycosis, perionychomycosis, pityriasis versicolor, oral thrush, vaginal candidiasis, respiratory tract candidiasis, biliary candidiasis, eosophageal candidiasis, urinary tract candidiasis, systemic candidiasis,

mucocutaneous candidiasis, aspergillosis, mucormycosis, paracoccidioidomycosis, North American blastomycosis, histoplasmosis, coccidioidomycosis, sporotrichosis, fungal sinusitis, or chronic sinusitis.

40. The method of claim 39, wherein the infection is candidemia or invasive candidiasis.

41 . The method of any one of claims 31 -40, wherein the fungal infection is an infection of Candida albicans, C. glabrata, C. dubliniensis, C. krusei, C. parapsilosis, C. tropicalis, C. orthopsilosis, C.

guilliermondii, C. rugosa, C. auris, C. lusitaniae, Aspergillus fumigatus, A. flavus, A. terreus, A. niger, A. candidus, A. clavatus, or A. ochraceus.

42. A method of preventing or treating a biofilm in a subject comprising administering to the subject a pharmaceutical composition comprising CD101 salt, or a neutral form thereof, and one or more pharmaceutically acceptable excipients.

43. The method of claim 42, wherein the biofilm is a Candida biofilm.

44. The method of claim 43, wherein the Candida biofilm is a Candida albicans biofilm.

45. The method of any one of claims 42-44, wherein the biofilm is attached to a mucous membrane of the subject.

46. A method of preventing biofilm growth on a catheter or killing a biofilm attached to a catheter comprising submerging the catheter in an aqueous solution comprising CD101 salt, or a neutral form thereof, or running an aqueous solution comprising CD101 salt, or a neutral form thereof, through the lumen of the catheter.

47. The method of claim 46, wherein the biofilm is a Candida biofilm.

48. The method of claim 47, wherein the Candida biofilm is a Candida albicans biofilm.

Description:
COMPOSITIONS AND METHODS FOR THE TREATMENT OF FUNGAL INFECTIONS

Background

The disclosure relates to the field of treatment of fungal infections. Systemic infections caused by Candida are serious and life-threatening infections that represent a significant public health issue, particularly in highly vulnerable patient populations such as the elderly, post-surgical, critically ill, and other hospitalized patients with serious medical conditions. The Centers for Disease Control and Prevention recently warned that fluconazole-resistant Candida have the potential to pose a serious threat to public health. However, since 2007, no new antifungal agents have been approved for treatment of candidemia. There is an urgent need to develop new and more effective treatments of Candida and other fungal infections.

Summary of the Disclosure

The disclosure relates to compositions and methods for the treatment of fungal infections. In particular, we have discovered pharmaceutical compositions for intravenous administration of CD101 (e.g., in salt or neutral form) that exhibit reduced local irritation upon intravenous administration to a subject. CD1 01 is a broad-spectrum antifungal agent with excellent activity against fungal species, e.g., Candida spp. and Aspergillus spp.

In a first aspect is a pharmaceutical composition for intravenous injection including an aqueous solution including at least 85% (w/w) water, between 0.4 mg/mL and 10 mg/mL CD101 (e.g., between 0.4 mg/mL and 9 mg/mL, between 0.4 mg/mL and 8 mg/mL, between 0.4 mg/mL and 7 mg/mL, between 0.4 mg/mL and 6 mg/mL, between 0.4 mg/mL and 5 mg/mL, between 0.4 mg/mL and 4 mg/mL, between 0.4 mg/mL and 3 mg/mL, between 0.4 mg/mL and 2 mg/mL, between 0.4 mg/mL and 1 mg/mL, between 0.6 mg/mL and 10 mg/mL, between 0.7 mg/mL and 10 mg/mL, between 0.8 mg/mL and 10 mg/mL, between 0.9 mg/mL and 10 mg/mL, between 1 mg/mL and 1 0 mg/mL, between 2 mg/mL and 10 mg/mL, between 3 mg/mL and 10 mg/mL, between 4 mg/mL and 1 0 mg/mL, between 5 mg/mL and 10 mg/mL, between 6 mg/mL and 10 mg/mL, between 7 mg/mL and 10 mg/mL, between 8 mg/mL and 10 mg/mL, or between 9 mg/mL and 10 mg/mL CD101 ; about 0.4 mg/mL, about 0.6 mg/mL, about 0.8 mg/mL, about 1 mg/mL, about 1 .2 mg/mL, about 1 .4 mg/mL, about 1 .6 mg/mL, about 1 .8 mg/mL, about 2 mg/mL, about 2.5 mg/mL, about 3 mg/mL, about 3.5 mg/mL, about 4 mg/mL, about 4.5 mg/mL, about 5 mg/mL, about 5.5 mg/mL, about 6 mg/mL, about 6.5 mg/mL, about 7 mg/mL, about 7.5 mg/mL, about 8 mg/mL, about 8.5 mg/mL, about 9 mg/mL, about 9.5 mg/mL, or about 1 0 mg/mL CD101 ), and an intravenous solubility promoter, wherein the weight to weight (w/w) ratio of the intravenous solubility promoter to the CD101 in the pharmaceutical composition is at least 2 (e.g., between 2 and 8, e.g., 2, 2.2, 2.4, 2.6, 2.8, 3, 3.2, 3.4, 3.6, 3.8, 4, 4.2, 4.4, 4.6, 4.8, 5, 5.2, 5.4, 5.6, 5.8, 6, 6.2, 6.4, 6.6, 6.8, 7, 7.2, 7.4, 7.6, 7.8, or 8), the

CD101 is in its salt or neutral form, the pharmaceutical composition has a pH of between 5 and 7 (e.g., 5, 5.1 , 5.2, 5.3, 5.4, 5.5, 5.6, 5.7, 5.8, 5.9, 6, 6.1 , 6.2, 6.3, 6.4, 6.5, 6.6, 6.7, 6.8, 6.9, or 7), and the pharmaceutical composition exhibits reduced local irritation upon intravenous administration to a subject.

In some embodiments of this aspect, the w/w ratio of the intravenous solubility promoter to the CD101 in the pharmaceutical composition is between 2 and 8 (e.g., 2, 2.2, 2.4, 2.6, 2.8, 3, 3.2, 3.4, 3.6, 3.8, 4, 4.2, 4.4, 4.6, 4.8, 5, 5.2, 5.4, 5.6, 5.8, 6, 6.2, 6.4, 6.6, 6.8, 7, 7.2, 7.4, 7.6, 7.8, or 8). In some embodiments of this aspect, the concentration of CD101 in the pharmaceutical composition is between 0.4 mg/mL and 4 mg/mL (e.g., about 0.8 mg/mL (e.g., 0.8±0.2 mg/mL) or about 1 .6 mg/mL (e.g., 1 .6±0.2 mg/mL)).

In a second aspect is a pharmaceutical composition including an effective amount of CD101 and an intravenous solubility promoter in a lyophilized composition, wherein the weight to weight (w/w) ratio of the intravenous solubility promoter to the CD101 in the lyophilized composition is between 2 and 8 (e.g., 2, 2.2, 2.4, 2.6, 2.8, 3, 3.2, 3.4, 3.6, 3.8, 4, 4.2, 4.4, 4.6, 4.8, 5, 5.2, 5.4, 5.6, 5.8, 6, 6.2, 6.4, 6.6, 6.8, 7, 7.2, 7.4, 7.6, 7.8, or 8), the CD101 is in its salt or neutral form, and the lyophilized composition, once reconstituted, provides an aqueous solution having a pH of between 5 and 7 (e.g., 5, 5.1 , 5.2, 5.3, 5.4, 5.5, 5.6, 5.7, 5.8, 5.9, 6, 6.1 , 6.2, 6.3, 6.4, 6.5, 6.6, 6.7, 6.8, 6.9, or 7).

In some embodiments, the intravenous solubility promoter is selected from the group consisting of polysorbate 20 (Tween 20; polyoxyethylene (20) sorbitan monolaurate), polysorbate 40 (Tween40; polyoxyethylene (40) sorbitan monopalmitate), polysorbate 60 (Tween 60; polyoxyethylene (60) sorbitan monostearate), polysorbate 80 (Tween 80; polyoxyethylene (80) sorbitan monooleate), β-cyclodextrin, polyoxyl 35 castor oil (Cremophor EL), polyoxyl 40 hydrogenated castor oil (Cremophor RH 40), polyoxyl 60 hydrogenated castor oil (Cremophor RH 60), D-a-tocopheryl polyethylene glycol 1000 succinate (TPGS), sorbitan monooleate (Span 20), polyoxyl 8 stearate (PEG 400 monosterate), polyoxyl 40 stearate (PEG 1750 monosterate), PEG 400 caprylic/capric glycerides (Labrasol), PEG 300 oleic glycerides (Labrafil M-1944CS), phosphatidylcholine (lecithin), alkylglucoside, sucrose monolaurate, sucrose monooleate, and polyoxyethylene-polyoxypropylene block copolymer (Poloxamer). In some embodiments, the alkylglucoside is an alkyl monoglucoside (e.g., hexylglucoside, heptaglucoside, octylglucoside, nonaglucoside, decylglucoside, dodecylglucoside, or tetradecylglucoside). In some embodiments, the alkylglucoside is an alkyl diglucoside (e.g., hexylmaltoside, heptamaltoside, octylmaltoside, nonamaltoside, decylmaltoside, dodecylmaltoside (e.g., dodecyl-p-D-maltoside (DDM)), or tetradecylmaltoside (e.g., tetradecyl-p-D-maltoside (TDM))).

In some embodiments, the intravenous solubility promoter is polysorbate 80 (Tween 80;

polyoxyethylene (80) sorbitan monooleate).

In some embodiments of the first two aspects of the invention, the pharmaceutical composition further includes a buffer. In some embodiments, the buffer is histidine, citrate, succinate, lactate, propanoate, arginine, tris(hydroxymethyl)aminomethane (tris), glycine, acetate, or formate. In some embodiments of the first two aspects of the invention, the buffer includes monovalent molecules (e.g., acetate, lactate, formate, histidine, tris(hydroxymethyl)aminomethane (tris), arginine, and/or glycine). In some embodiments of the first two aspects of the invention, the buffer includes amino acids (e.g., histidine, arginine, and/or glycine).

In some embodiments of the first aspect of the invention, the pharmaceutical composition further includes between 0.12% and 0.6% (w/w) of a saccharide (e.g., between 0.12% and 0.58% (w/w), between 0.12% and 0.56% (w/w), between 0.12% and 0.54% (w/w), between 0.12% and 0.52% (w/w), between 0.12% and 0.5% (w/w), between 0.12% and 0.48% (w/w), between 0.12% and 0.46% (w/w), between 0.12% and 0.44% (w/w), between 0.12% and 0.42% (w/w), between 0.12% and 0.4% (w/w), between 0.12% and 0.38% (w/w), between 0.12% and 0.36% (w/w), between 0.12% and 0.34% (w/w), between 0.12% and 0.32% (w/w), between 0.12% and 0.3% (w/w), between 0.12% and 0.28% (w/w), between 0.12% and 0.26% (w/w), between 0.12% and 0.24% (w/w), between 0.12% and 0.22% (w/w), between 0.12% and 0.2% (w/w), between 0.12% and 0.1 8% (w/w), between 0.12% and 0.1 6% (w/w), or between 0.12% and 0.14% (w/w) of a saccharide; about 0.12% (w/w), about 0.14% (w/w), about 0.16% (w/w), about 0.18% (w/w), about 0.2% (w/w), about 0.22% (w/w), about 0.24% (w/w), about 0.26% (w/w), about 0.28% (w/w), about 0.3% (w/w), about 0.32% (w/w), about 0.34% (w/w), about 0.36% (w/w), about 0.38% (w/w), about 0.4% (w/w), about 0.42% (w/w), about 0.44% (w/w), about 0.46% (w/w), about 0.48% (w/w), about 0.5% (w/w), about 0.52% (w/w), about 0.54% (w/w), about 0.56% (w/w), about 0.58% (w/w), or about 0.6% (w/w) of a saccharide). In some embodiments, the saccharide is mannitol, sucrose, trehalose, maltose, dextrose, or lactose (e.g., mannitol).

In some embodiments of the second aspect of the invention, the pharmaceutical composition further includes between 2% and 10% (w/w) of a saccharide (e.g., between 2% and 9% (w/w), between 2% and 8% (w/w), between 2% and 7% (w/w), between 2% and 6% (w/w), between 2% and 5% (w/w), between 2% and 4% (w/w), or between 2% and 3% (w/w) of a saccharide; about 2% (w/w), about 3% (w/w), about 4% (w/w), about 5% (w/w), about 6% (w/w), about 7% (w/w), about 8% (w/w), about 9% (w/w), or about 10% (w/w) of a saccharide).

In some embodiments, the CD101 in salt form in pharmaceutical compositions and methods described herein is CD101 acetate.

In another aspect is a method of treating or preventing a fungal infection in a subject by intravenously administering the pharmaceutical composition of the first aspect of the invention to the subject, wherein the method exhibits reduced local irritation upon intravenous administration of the pharmaceutical composition to a subject.

In another aspect is a method of treating or preventing a fungal infection in a subject by: (i) reconstituting the pharmaceutical composition of the second aspect of the invention to form an aqueous solution; and (ii) intravenously administering the aqueous solution to the subject, wherein the concentration of the CD101 in the aqueous solution is between 0.4 mg/mL and 10 mg/mL (e.g., between 0.4 mg/mL and 9 mg/mL, between 0.4 mg/mL and 8 mg/mL, between 0.4 mg/mL and 7 mg/mL, between 0.4 mg/mL and 6 mg/mL, between 0.4 mg/mL and 5 mg/mL, between 0.4 mg/mL and 4 mg/mL, between 0.4 mg/mL and 3 mg/mL, between 0.4 mg/mL and 2 mg/mL, between 0.4 mg/mL and 1 mg/mL, between 0.6 mg/mL and 10 mg/mL, between 0.7 mg/mL and 10 mg/mL, between 0.8 mg/mL and 10 mg/mL, between 0.9 mg/mL and 10 mg/mL, between 1 mg/mL and 10 mg/mL, between 2 mg/mL and 10 mg/mL, between 3 mg/mL and 1 0 mg/mL, between 4 mg/mL and 1 0 mg/mL, between 5 mg/mL and 10 mg/mL, between 6 mg/mL and 1 0 mg/mL, between 7 mg/mL and 1 0 mg/mL, between 8 mg/mL and 10 mg/mL, or between 9 mg/mL and 1 0 mg/mL CD101 ; about 0.4 mg/mL, about 0.6 mg/mL, about 0.8 mg/mL, about 1 mg/mL, about 1 .2 mg/mL, about 1 .4 mg/mL, about 1 .6 mg/mL, about 1 .8 mg/mL, about 2 mg/mL, about 2.5 mg/mL, about 3 mg/mL, about 3.5 mg/mL, about 4 mg/mL, about 4.5 mg/mL, about 5 mg/mL, about 5.5 mg/mL, about 6 mg/mL, about 6.5 mg/mL, about 7 mg/mL, about 7.5 mg/mL, about 8 mg/mL, about 8.5 mg/mL, about 9 mg/mL, about 9.5 mg/mL, or about 10 mg/mL CD101 ), the CD101 is in its salt or neutral form, and the method exhibits reduced local irritation upon intravenous administration of the aqueous solution to a subject.

In some embodiments of this aspect, the concentration of CD101 in the pharmaceutical composition is about 0.4 mg/mL. In some embodiments of this aspect, the concentration of CD101 in the pharmaceutical composition is about 0.8 mg/mL. In some embodiments, the concentration of CD101 in the pharmaceutical composition is about 1 .6 mg/mL.

In some embodiments of this aspect, the concentration of CD101 in the pharmaceutical composition is about 3.0 mg/mL. In some embodiments, the concentration of CD101 in the

pharmaceutical composition is about 4.0 mg/mL.

In some embodiments of the methods of the invention, the method includes intravenously administering to the subject by infusion. In some embodiments, the method includes intravenously administering to the subject by infusion at a constant infusion rate of between 2 mL/minute and 9 mL/minute (e.g., between 2 mL/minute and 8 mL/minute, between 2 mL/minute and 7 mL/minute, between 2 mL/minute and 6 mL/minute, between 2 mL/minute and 5 mL/minute, between 2 mL/minute and 4 mL/minute, between 2 mL/minute and 3 mL/minute, between 3 mL/minute and 9 mL/minute, between 4 mL/minute and 9 mL/minute, between 5 mL/minute and 9 mL/minute, between 6 mL/minute and 9 mL/minute, between 7 mL/minute and 9 mL/minute, or between 8 mL/minute and 9 mL/minute; e.g., 2, 2.5, 3, 3.5, 4, 4.5, 5, 5.5, 6, 6.5, 7, 7.5, 8, 8.5, or 9 mL/minute). In some embodiments, the method includes intravenously administering to the subject by infusion over 30 to 120 minutes (e.g., 30, 40, 50, 60, 70, 80, 90, 1 00, 1 10, or 120 minutes).

In some embodiments of the methods of the invention, the method includes intravenously administering at least one dose every 5 to 15 days (e.g., every 5, 6, 7, 8, 9, 10, 1 1 , 12, 13, 14, or 15 days). In some embodiments, the method includes intravenously administering one dose every 5 to 15 days (e.g., every 5, 6, 7, 8, 9, 10, 1 1 , 12, 13, 14, or 15 days).

In some embodiments of the methods of the invention, the fungal infection to be treated or prevented is selected from candidemia, invasive candidiasis, tinea capitis, tinea corporis, tinea pedis, onychomycosis, perionychomycosis, pityriasis versicolor, oral thrush, vaginal candidiasis, respiratory tract candidiasis, biliary candidiasis, eosophageal candidiasis, urinary tract candidiasis, systemic candidiasis, mucocutaneous candidiasis, aspergillosis, mucormycosis, paracoccidioidomycosis, North American blastomycosis, histoplasmosis, coccidioidomycosis, sporotrichosis, fungal sinusitis, or chronic sinusitis. In some embodiments, the infection is candidemia or invasive candidiasis.

In some embodiments of the methods of the invention, the fungal infection to be treated or prevented is an infection of Candida albicans, C. glabrata, C. dubliniensis, C. krusei, C. parapsilosis, C. tropicalis, C. orthopsilosis, C. guilliermondii, C. rugosa, C. auris, C. lusitaniae, Aspergillus fumigatus, A. flavus, A. terreus, A. niger, A. candidus, A. clavatus, or A. ochraceus.

In another aspect, the invention features a method of preventing or treating a biofilm in a subject. The method includes administering to the subject a pharmaceutical composition comprising CD101 salt, or a neutral form thereof, and one or more pharmaceutically acceptable excipients.

In some embodiments of this aspect, the biofilm in the subject is a Candida biofilm (e.g., Candida albicans biofilm). In some embodiments, the biofilm is attached to a mucous membrane of the subject.

In another aspect, the invention features a method of preventing biofilm growth on a catheter or killing a biofilm attached to a catheter comprising submerging the catheter in an aqueous solution comprising CD101 salt, or a neutral form thereof, or running an aqueous solution comprising CD101 salt, or a neutral form thereof, through the lumen of the catheter. In some embodiments of this aspect, the biofilm on the catheter is a Candida biofilm (e.g., Candida albicans biofilm).

Definitions:

The terms "intravenous administration" or "intravenously administering," as used herein, refer to intravenous infusion of a drug to a subject.

The term "fungal infection," as used herein, refers to the invasion of a subject's cells, tissues, and/or organs by fungi (e.g., Candida spp. or Aspergillus spp.), thus, causing an infection. In some embodiments, the fungi may grow, multiply, and/or produce toxins in the subject's cells, tissues, and/or organs. In some embodiments, a fungal infection can be any situation in which the presence of a fungal population(s) is latent within or damaging to a host body. Thus, a subject is "suffering" from a fungal infection when a latent fungal population is detectable in or on the subject's body, an excessive amount of a fungal population is present in or on the subject's body, or when the presence of a fungal population(s) is damaging the cells, tissues, and/or organs of the subject.

The term "protecting against a fungal infection" or "preventing a fungal infection" as used herein, refers to preventing a subject from developing a fungal infection or decreasing the risk that a subject may develop a fungal infection (e.g., a fungal infection caused by Candida spp. or Aspergillus spp.).

Prophylactic drugs used in methods of protecting against a fungal infection in a subject are often administered to the subject prior to any detection of the fungal infection. In some embodiments of methods of protecting against a fungal infection, a subject (e.g., a subject at risk of developing a fungal infection) may be administered a pharmaceutical composition of the invention to prevent the fungal infection development or decrease the risk of the fungal infection development.

As used herein, the term "biofilm" refers to a three-dimensional structure composed of heterogeneous fungi (e.g., Candida) and hyphae that can attach to various surfaces, e.g., a mucous membrane or the inside of a catheter. Biofilms can form on the surfaces of medical devices and cause biofilm device-associated infections. For example, having a biofilm on an indwelling device, e.g., a vascular catheter, can cause life-threatening infections.

The term "treating" or "to treat," as used herein, refers to a therapeutic treatment of a fungal infection (e.g., a fungal infection caused by Candida spp. or Aspergillus spp.) in a subject. In some embodiments, a therapeutic treatment may slow the progression of the fungal infection, improve the subject's outcome, and/or eliminate the infection. In some embodiments, a therapeutic treatment of a fungal infection (e.g., a fungal infection caused by Candida spp. or Aspergillus spp.) in a subject may alleviate or ameliorate of one or more symptoms or conditions associated with the fungal infection, diminish the extent of the fungal infection, stabilize (i.e., not worsening) the state of the fungal infection, prevent the spread of the fungal infection, and/or delay or slow the progress of the fungal infection, as compare the state and/or the condition of the fungal infection in the absence of therapeutic treatment.

The term "local irritation," as used herein, refers to any adverse reactions the subject might experience at the site of the intravenous injection after a drug is administered. Local irritations at the injection site may include, e.g., pain or tenderness at the injection site, itching, bruising, and/or swelling of the skin, venous discoloration (e.g., darkening of the blood vein leading away from the site of injection), and skin rash. "Reduced local irritation" refers to diminished local irritation or no local irritation observed when intravenously administering a pharmaceutical composition of the invention to a subject, relative the irritation observed when intravenously administering other compositions.

The term "intravenous solubility promoter," as used herein, refers to an agent that promotes the intravenous solubility of CD101 (e.g., CD101 in salt or neutral form) in the bloodstream of a subject once the pharmaceutical composition is intravenously administered. The intravenous precipitation of CD101 can cause local irritations at the site of injection (e.g., pain or tenderness at the injection site, itching, bruising, and/or swelling of the skin, venous discoloration (e.g., darkening of the blood vein leading away from the site of injection), and skin rash). The intravenous solubility promoter in a pharmaceutical composition of the invention functions to decrease the intravenous precipitation of CD101 (e.g., CD101 in salt or neutral form) in the bloodstream of a subject relative the intravenous precipitation of CD101 if it was in a composition without the intravenous solubility promoter. In some embodiments, a

pharmaceutical composition of the invention contains polysorbate 80 (Tween 80) as the intravenous solubility promoter.

The term "alkylglucoside," as used herein, refers to a surfactant having an alkyl chain (e.g., an alkyl chain ranging from six to 18 carbon atoms) conjugated to one or more glucose molecules. An alkylglucoside may be an alkyl monoglucoside (e.g., an alkyl chain conjugated to one glucose molecule) or an alkyl diglucoside (e.g., an alkyl chain conjugated to a disaccharide containing two glucose molecules linked through an interglycosidic bond (e.g., a maltose)).

The term "immunocompromised," as used herein, refers to a subject (e.g., a human) having a weakened immune system. The subject's immune system can be weakened or compromised by a disease (e.g., an HIV infection, an autoimmune disease, cancer), a medical procedure (e.g., an organ transplant (e.g., a solid organ transplant) or a bone marrow transplant), a drug (e.g., an

immunosuppressant), and/or a pathogen (e.g., bacteria, fungus, virus). The immune system of the host may also have a congenital defect that renders the host more susceptible to infection.

The term "immunosuppression therapy," as used herein, refers to a therapy that uses one or more immunosuppressants to reduce the activation and/or efficacy of the immune system of a subject (e.g., a human). In some embodiments, an immunosuppression therapy is used to prevent the body from rejecting a transplant (e.g., an organ transplant (e.g., a solid organ transplant) or a bone marrow transplant), to treat graft-versus-host disease after a bone marrow transplant, and/or to treat autoimmune diseases (e.g., systemic lupus erythematosus, rheumatoid arthritis, Crohn's disease, multiple sclerosis, myasthenia gravis, Sarcoidosis, Behcet's disease). Immunosuppressants include, but are not limited to, calcineurin inhibitors, mTOR inhibitors, and tyrosine kinase inhibitors (e.g., cyclosporine A, cyclosporine G, voclosporin, tacrolimus, pimecrolimus, sirolimus, temsirolimus, deforolimus, everolimus, zotarolimus, biolimus, imatinib, dasatinib, nilotinib, erlotinib, sunitinib, gefitinib, bosutinib, neratinib, axitinib, crizotinib, lapatinib, toceranib and vatalanib).

The term "effective amount," as used herein, is meant the amount of drug required to treat or prevent an infection or a disease associated with an infection. The effective amount of drug used to practice the methods described herein for therapeutic or prophylactic treatment of conditions caused by or contributed to by a microbial infection varies depending upon the manner of administration, the age, body weight, and general health of the subject. Ultimately, the attending physician will decide the appropriate amount and dosage regimen. Such amount is referred to as an "effective amount." The term "CD101 salt," as used herein, refers to a salt of the compound of Formula 1 . CD101 has a structure (below) in which the tertiary ammonium ion positive charge of CD101 is balanced with a negative counterion (e.g., an acetate) in its salt form.

the compound of Formula 1 has no net positive or negative charge. The zwitterion is present in a higher proportion in basic medium (e.g., pH 9) relative to CD101 or a salt of CD101 . In some embodiments, the zwitterion may also be present in its salt form.

The term "concentration of CD101 ," as used herein, is calculated based on the molecular weight of CD101 as shown in Formula 1 , not including the negative counterion (e.g., an acetate) if CD101 is in its salt form. For example, in some embodiments of the invention, the concentration of CD101 in the pharmaceutical composition is between 0.4 mg/mL and 4 mg/mL (e.g., about 0.8 mg/mL (e.g., 0.8±0.2 mg/mL) or about 1 .6 mg/mL (e.g., 1 .6±0.2 mg/mL)). These concentrations are calculated based on the molecular weight of CD101 as shown in Formula 1 .

The term "weight to weight (w/w) ratio," as used herein, is calculated based on the molecular weight of CD101 as shown in Formula 1 , not including the negative counterion (e.g., an acetate) if CD1 01 is in its salt form. For example, in some embodiments of the invention, the weight to weight (w/w) ratio of the blood solubilizing agent to CD101 in the pharmaceutical composition is at least 2 (e.g., between 2 and 8, e.g., 2, 2.2, 2.4, 2.6, 2.8, 3, 3.2, 3.4, 3.6, 3.8, 4, 4.2, 4.4, 4.6, 4.8, 5, 5.2, 5.4, 5.6, 5.8, 6, 6.2, 6.4, 6.6, 6.8, 7, 7.2, 7.4, 7.6, 7.8, or 8). The w/w ratio of the blood solubilizing agent to CD101 is calculated based on the molecular weight of CD101 as shown in Formula 1 .

The term "salt," as used herein, refers to any pharmaceutically acceptable salt, such as a nontoxic acid addition salt, metal salt, or metal complex, commonly used in the pharmaceutical industry. Examples of acid addition salts include organic acids, such as acetic, lactic, palmoic, maleic, citric, cholic acid, capric acid, caprylic acid, lauric acid, glutaric, glucuronic, glyceric, glycocolic, glyoxylic, isocitric, isovaleric, lactic, malic, oxalo acetic, oxalosuccinic, propionic, pyruvic, ascorbic, succinic, benzoic, palmitic, suberic, salicylic, tartaric, methanesulfonic, toluenesulfonic, and trifluoroacetic acids, and inorganic acids, such as hydrochloric acid, hydrobromic acid, sulfuric acid, and phosphoric acid.

Representative alkali or alkaline earth metal salts include sodium, lithium, potassium, calcium, and magnesium, among others. The term "dose," as used herein, is meant the amount of CD101 administered to the subject. As used herein, the amount in each dose refers to the amount of CD101 (Formula 1 shown above) that does not include the negative counterion (e.g., an acetate) if CD101 is in its salt form.

The term "between," as used herein, refers to any quantity within the range indicated and enclosing each of the ends of the range indicated. For example, a pH of between 5 and 7 refers to any quantity within 5 and 7, as well as a pH of 5 and a pH of 7.

The term "subject" or "patient," as used herein, is meant a human.

Other than in the operating examples, or where otherwise indicated, all numbers expressing quantities of ingredients, concentrations, or reaction conditions used herein should be understood as modified in all instances by the term "about." As used herein, the term "about" indicates a deviation of ±10%. For example, about 10% refers to from 9.5% to 10.5%. The term "about," as used herein, indicates a deviation of ±10%. For example, about 10% refers to from 9% to 1 1 %.

Other features and advantages of the disclosure will be apparent from the following detailed description and the claims.

Brief Description of the Drawings

FIG. 1 is a graph showing percent survival over time in mice infected with Candida auris and treated with 20 mg/kg CD101 (IP), 20 mg/kg fluconazole (PO), or 0.3 mg/kg amphotericin B (IP).

FIGS. 2A and 2B are bar graphs showing the effect of CD101 (0.25 or 1 μς/ιηΙ) (FIG. 2A) and fluconazole (1 or 4 μς/ιηΙ) (FIG. 2B) on metabolic activity of adhesion phase C. albicans biofilms compared to untreated control.

FIGS. 3A-3E are confocal scanning laser micrographs showing the effect of CD101 and fluconazole on adhesion phase C. albicans biofilms (prevention): top-down three-dimensional view (top panels) and side-views (bottom panels) of biofilms formed by C. albicans treated with: no drug (control; FIG .3A), 0.25 1 ig/m\ CD101 (FIG. 3B), 1 ig/m\ CD101 (FIG. 3C), 1 ig/m\ fluconazole (FIG. 3D), and 4 μg/ml fluconazole (FIG. 3E).

FIGS. 3F and 3G are bar graphs showing the thickness of C. albicans biofilms exposed to CD1 01 (FIG. 3F) and fluconazole (FIG. 3G).

FIGS. 4A and 4B are bar graphs showing the effect of CD101 (0.25 or 1 μς/ιηΙ) (FIG. 4A) and fluconazole (1 or 4 μς/ιηΙ) (FIG. 4B) on metabolic activity of mature phase C. albicans biofilms compared to untreated control.

FIGS. 5A-5E are confocal scanning laser micrographs showing the effect of CD101 and

Fluconazole on Mature phase C. albicans Biofilms (treatment): Top-down three-dimensional view (top panels) and side-view (bottom panels) of biofilms exposed to: no drug (FIG. 5A), 0.25 μg/ml CD101 (FIG. 5B), 1 ig/m\ CD101 (FIG. 5C), 1 ig/m\ fluconazole (FIG. 5D), and 4 ig/m\ fluconazole (FIG. 5E). Arrows show bulged/broken cells.

FIGS. 5F and 5G are bar graphs showing thickness of C. albicans biofilms exposed to: CD101 (FIG. 5F) and fluconazole (FIG. 5G).

FIGS. 6A-6F are images showing the temporal effect of CD101 (0.25 μς/ιηΙ) on formation of C. albicans biofilms. Images were captured immediately from Oh and followed up to 16h for biofilms treated with: no drug (FIGS. 6A and 6B), CD101 at low magnification, x20 (FIGS. 6C and 6D), and CD101 at high magnification, x63 (FIGS. 6E and 6F). Arrows show bulging, deformed, and broken cells.

FIGS. 7A and 7B are images showing the temporal effect of CD101 (0.25 μς/ιηΙ) on 3h formed C. albicans Biofilms. CD101 was added after 3h biofilm formation and images were captured immediately after adding CD101 (FIG. 7A) and followed up to 16h (FIG. 7B), magnification, x63. Arrows show bulging, deformed, and broken cells.

Detailed Description

Provided are methods of treating a fungal infection in a subject in need thereof by intravenously administering to the subject CD101 (e.g., CD101 in salt or neutral form) formulated as an aqueous composition.

Pharmaceutical compositions

CD101 is a semi-synthetic echinocandin that inhibits the synthesis of 1 ,3-p-D-glucan, an essential component of the fungal cell wall of yeast forms of Candida species and regions of active cell growth of Aspergillus hyphae. The synthesis of 1 ,3-p-D-glucan is dependent upon the activity of 1 ,3-p-D-glucan synthase, an enzyme complex in which the catalytic subunit is encoded by FKS1, FKS2, and FKS3 genes. Inhibition of this enzyme results in rapid, concentration-dependent, fungicidal activity for Candida spp. The structure of CD101 is depicted above.

The disclosure features pharmaceutical compositions for intravenous administration of CD101

(e.g., CD101 in salt or neutral form) to a subject. In some embodiments, the pharmaceutical composition for intravenous administration includes between 0.4 mg/mL and 10 mg/mL CD101 (e.g., between 0.4 mg/mL and 9 mg/mL, between 0.4 mg/mL and 8 mg/mL, between 0.4 mg/mL and 7 mg/mL, between 0.4 mg/mL and 6 mg/mL, between 0.4 mg/mL and 5 mg/mL, between 0.4 mg/mL and 4 mg/mL, between 0.4 mg/mL and 3 mg/mL, between 0.4 mg/mL and 2 mg/mL, between 0.4 mg/mL and 1 mg/mL, between 0.6 mg/mL and 10 mg/mL, between 0.7 mg/mL and 10 mg/mL, between 0.8 mg/mL and 10 mg/mL, between 0.9 mg/mL and 10 mg/mL, between 1 mg/mL and 1 0 mg/mL, between 2 mg/mL and 10 mg/mL, between 3 mg/mL and 10 mg/mL, between 4 mg/mL and 1 0 mg/mL, between 5 mg/mL and 10 mg/mL, between 6 mg/mL and 10 mg/mL, between 7 mg/mL and 10 mg/mL, between 8 mg/mL and 10 mg/mL, or between 9 mg/mL and 10 mg/mL CD101 ) and an intravenous solubility promoter.

The intravenous solubility promoter functions to promote the intravenous solubility of CD101 (e.g., CD101 in salt or neutral form) in the bloodstream of a subject once the pharmaceutical composition is intravenously administered. In some embodiments, the intravenous precipitation of CD101 causes local irritations at the site of injection (e.g., pain or tenderness at the injection site, itching, bruising, and/or swelling of the skin, venous discoloration (e.g., darkening of the blood vein leading away from the site of injection), and skin rash). The intravenous solubility promoter in a pharmaceutical composition of the invention decreases the intravenous precipitation of CD101 in the bloodstream of a subject relative the intravenous precipitation of CD101 if it was in a composition without the intravenous solubility promoter. In some embodiments, a pharmaceutical composition that includes between 0.4 mg/mL and 10 mg/mL CD101 and an intravenous solubility promoter exhibits reduced local irritation upon intravenous administration to the subject. The amount of an intravenous solubility promoter in a pharmaceutical composition may depend on, e.g., the concentration of CD101 in the pharmaceutical composition, and/or the pH of the

pharmaceutical composition. In some embodiments, the weight to weight (w/w) ratio of the intravenous solubility promoter to the CD101 in the pharmaceutical composition is at least 2 (e.g., between 2 and 8, e.g., 2, 2.2, 2.4, 2.6, 2.8, 3, 3.2, 3.4, 3.6, 3.8, 4, 4.2, 4.4, 4.6, 4.8, 5, 5.2, 5.4, 5.6, 5.8, 6, 6.2, 6.4, 6.6, 6.8, 7, 7.2, 7.4, 7.6, 7.8, or 8). Examples of intravenous solubility promoters that may be included in a pharmaceutical composition described herein include, but are not limited to, e.g., polysorbate 20 (Tween 20; polyoxyethylene (20) sorbitan monolaurate), polysorbate 40 (Tween40; polyoxyethylene (40) sorbitan monopalmitate), polysorbate 60 (Tween 60; polyoxyethylene (60) sorbitan monostearate), polysorbate 80 (Tween 80; polyoxyethylene (80) sorbitan monooleate), β-cyclodextrin, polyoxyl 35 castor oil (Cremophor EL), polyoxyl 40 hydrogenated castor oil (Cremophor RH 40), polyoxyl 60 hydrogenated castor oil (Cremophor RH 60), D-a-tocopheryl polyethylene glycol 1 000 succinate (TPGS), sorbitan monooleate (Span 20), polyoxyl 8 stearate (PEG 400 monosterate), polyoxyl 40 stearate (PEG 1750 monosterate), PEG 400 caprylic/capric glycerides (Labrasol), PEG 300 oleic glycerides (Labrafil M-1944CS), phosphatidylcholine (lecithin), alkylglucoside, sucrose monolaurate, sucrose monooleate, and polyoxyethylene-polyoxypropylene block copolymer (Poloxamer (e.g., poloxamers 101 , 1 05, 108, 122, 123, 124, 181 , 1 82, 183, 184, 185, 188, 212, 21 5, 217, 231 , 234, 235, 237, 238, 282, 284, 288, 331 , 333, 334, 335, 338, 401 , 402, 403, and 407, poloxamer 105 benzoate, and poloxamer 182 dibenzoate)). In some embodiments, the alkylglucoside is an alkyl monoglucoside (e.g., hexylglucoside, heptaglucoside, octylglucoside, nonaglucoside, decylglucoside, dodecylglucoside, or tetradecylglucoside). In some embodiments, the alkylglucoside is an alkyl diglucoside (e.g., hexylmaltoside, heptamaltoside, octylmaltoside, nonamaltoside, decylmaltoside, dodecylmaltoside (e.g., dodecyl-p-D-maltoside (DDM)), or tetradecylmaltoside (e.g., tetradecyl-p-D-maltoside (TDM))). In some embodiments, the intravenous solubility promoter in the pharmaceutical composition is polysorbate 80.

The concentration of CD101 in a pharmaceutical composition for intravenous administration described herein may be between 0.4 mg/mL and 4 mg/mL (e.g., 0.4, 0.5, 0.6, 0.7, 0.8, 0.9, 1 , 1 .1 , 1 .2, 1 .3, 1 .4, 1 .5, 1 .6, 1 .7, 1 .8, 1 .9, or 2, 2.1 , 2.2, 2.3, 2.4, 2.5, 2.6, 2.7, 2.8, 2.9, 3, 3.1 , 3.2, 3.3, 3.4, 3.5, 3.6, 3.7, 3.8, 3.9, or 4 mg/mL). In some embodiments, the concentration of CD101 in a pharmaceutical composition for intravenous administration described herein is 0.8±0.2 mg/mL. In some embodiments, the concentration of CD101 in a pharmaceutical composition for intravenous administration described herein is 1 .6±0.2 mg/mL.

A pharmaceutical composition for intravenous administration of the invention may also include a buffer. A buffer functions to maintain the pH of the composition. In some embodiments, a

pharmaceutical composition for intravenous administration of the invention has a pH of between 5 and 7 (e.g., 5, 5.1 , 5.2, 5.3, 5.4, 5.5, 5.6, 5.7, 5.8, 5.9, 6, 6.1 , 6.2, 6.3, 6.4, 6.5, 6.6, 6.7, 6.8, 6.9, or 7). A buffer included in a pharmaceutical composition for intravenous administration described herein may be, e.g., histidine, citrate, succinate, lactate, propanoate, arginine, tris(hydroxymethyl)aminomethane (tris), glycine, acetate, or formate.

Furthermore, a saccharide (e.g., mannitol, sucrose, trehalose, maltose, dextrose, or lactose) may also be included in a pharmaceutical composition of the invention. A pharmaceutical composition for intravenous injection including an aqueous solution including between 0.4 mg/mL and 10 mg/mL CD101 may include between 0.12% and 0.6% (w/w) of a saccharide (e.g., between 0.12% and 0.58% (w/w), between 0.12% and 0.56% (w/w), between 0.12% and 0.54% (w/w), between 0.12% and 0.52% (w/w), between 0.12% and 0.5% (w/w), between 0.12% and 0.48% (w/w), between 0.12% and 0.46% (w/w), between 0.12% and 0.44% (w/w), between 0.12% and 0.42% (w/w), between 0.12% and 0.4% (w/w), between 0.12% and 0.38% (w/w), between 0.12% and 0.36% (w/w), between 0.12% and 0.34% (w/w), between 0.12% and 0.32% (w/w), between 0.12% and 0.3% (w/w), between 0.12% and 0.28% (w/w), between 0.12% and 0.26% (w/w), between 0.12% and 0.24% (w/w), between 0.12% and 0.22% (w/w), between 0.12% and 0.2% (w/w), between 0.12% and 0.1 8% (w/w), between 0.12% and 0.1 6% (w/w), or between 0.12% and 0.14% (w/w) of a saccharide; about 0.12% (w/w), about 0.14% (w/w), about 0.16% (w/w), about 0.18% (w/w), about 0.2% (w/w), about 0.22% (w/w), about 0.24% (w/w), about 0.26% (w/w), about 0.28% (w/w), about 0.3% (w/w), about 0.32% (w/w), about 0.34% (w/w), about 0.36% (w/w), about 0.38% (w/w), about 0.4% (w/w), about 0.42% (w/w), about 0.44% (w/w), about 0.46% (w/w), about 0.48% (w/w), about 0.5% (w/w), about 0.52% (w/w), about 0.54% (w/w), about 0.56% (w/w), about 0.58% (w/w), or about 0.6% (w/w) of a saccharide). In some embodiments, the saccharide in the pharmaceutical composition is mannitol.

A pharmaceutical composition including an effective amount of CD101 (e.g., CD101 in salt or neutral form) and an intravenous solubility promoter in a lyophilized composition may include between 2% and 10% (w/w) of a saccharide (e.g., between 2% and 9% (w/w), between 2% and 8% (w/w), between 2% and 7% (w/w), between 2% and 6% (w/w), between 2% and 5% (w/w), between 2% and 4% (w/w), or between 2% and 3% (w/w) of a saccharide; about 2% (w/w), about 3% (w/w), about 4% (w/w), about 5% (w/w), about 6% (w/w), about 7% (w/w), about 8% (w/w), about 9% (w/w), or about 10% (w/w) of a saccharide).

A pharmaceutical composition of the invention may also include an effective amount of CD101 and an intravenous solubility promoter in a lyophilized composition. The lyophilized composition may include a weight to weight (w/w) ratio of the intravenous solubility promoter to the CD101 that is between 2 and 8(e.g., 2, 2.2, 2.4, 2.6, 2.8, 3, 3.2, 3.4, 3.6, 3.8, 4, 4.2, 4.4, 4.6, 4.8, 5, 5.2, 5.4, 5.6, 5.8, 6, 6.2, 6.4, 6.6, 6.8, 7, 7.2, 7.4, 7.6, 7.8, or 8) . The CD101 in the lyophilized composition may be in its salt or neutral form. The lyophilized composition may be reconstituted to provide an aqueous solution having a pH of between 5 and 7 (e.g., 5, 5.1 , 5.2, 5.3, 5.4, 5.5, 5.6, 5.7, 5.8, 5.9, 6, 6.1 , 6.2, 6.3, 6.4, 6.5, 6.6, 6.7, 6.8, 6.9, or 7).

Therapy

Methods of the invention include, e.g., methods of treating or preventing a fungal infection (e.g., a fungal infection caused by Candida spp. or Aspergillus spp.) in a subject by intravenously administering to the subject a pharmaceutical composition described herein. When using a pharmaceutical composition including CD101 (e.g., CD1 01 in salt or neutral form) and an intravenous solubility promoter in a lyophilized composition, the pharmaceutical composition may first be reconstituted to form an aqueous solution (e.g., an aqueous solution having a concentration of the CD101 that is between 0.4 mg/mL and 10 mg/mL), which can then be intravenously administered to the subject.

Methods of treating or preventing a fungal infection described herein exhibit reduced local irritation upon intravenous administration of a pharmaceutical composition of the invention to a subject. The subject may experience reduced local irritation or no local irritation, e.g., pain or tenderness at the injection site, itching, bruising, and/or swelling of the skin, venous discoloration (e.g., darkening of the blood vein leading away from the site of injection), and skin rash, upon intravenous administration of a pharmaceutical composition of the invention, relative to the irritation experienced when intravenously administered other compositions.

In some embodiments of the methods of the invention, a pharmaceutical composition described herein may be intravenously administered to the subject by infusion. The intravenous infusion may be performed at a constant infusion rate of between 2 mL/minute and 9 mL/minute (e.g., 2, 2.5, 3, 3.5, 4, 4.5, 5, 5.5, 6, 6.5, 7, 7.5, 8, 8.5, or 9 mL/minute). In some embodiments, the pharmaceutical composition may be administered to the subject over 30 to 120 minutes (e.g., 30, 40, 50, 60, 70, 80, 90, 100, 1 1 0, or 120 minutes). In certain embodiments of the methods of the invention, 100 mL to 500 mL (e.g., 100, 105, 1 10, 1 15, 120, 125, 135, 145, 155, 165, 175, 185, 1 95, 205, 215, 225, 235, 245, 255, 265, 275, 285, 295, 305, 315, 325, 335, 345, 355, 365, 375, 385, 395, 405, 415, 425, 435, 445, 455, 465, 475, 485, 495, or 500 mL; 100 mL to 480 mL, 100 mL to 460 mL, 100 mL to 440 mL, 100 mL to 420 mL, 100 mL to 400 mL, 100 mL to 380 mL, 100 mL to 360 mL, 100 mL to 340 mL, 100 mL to 320 mL, 100 mL to 300 mL, 100 mL to 280 mL, 100 mL to 260 mL, 100 mL to 240 mL, 100 mL to 220 mL, 100 mL to 200 mL, 100 mL to 190 mL, 100 mL to 180 mL, 100 mL to 170 mL, 100 mL to 1 60 mL, 100 mL to 150 mL, 100 mL to 140 mL, or 100 mL to 130 mL; 120 mL to 500 mL, 140 mL to 500 mL, 160 mL to 500 mL, 180 mL to 500 mL, 200 mL to 500 mL, 220 mL to 500 mL, 240 mL to 500 mL, 260 mL to 500 mL, 280 mL to 500 mL, 300 mL to 500 mL, 320 mL to 500 mL, 340 mL to 500 mL, 360 mL to 500 mL, 380 mL to 500 mL, 400 mL to 500 mL, 420 mL to 500 mL, 440 mL to 500 mL, 460 mL to 500 mL, or 480 mL to 500 mL) of a pharmaceutical composition (e.g., a pharmaceutical composition having a concentration of CD101 at 0.4 mg/mL, 0.8 mg/mL, or 1 .6 mg/mL) is administered to the subject in 20 minutes to 60 minutes (e.g., 20, 25, 30, 35, 40, 45, 50, 55, or 60 minutes). In some embodiments, 1 00 mL to 250 mL (e.g., 105, 1 15, 125, 135, 145, 1 55, 165, 175, 185, 1 95, 205, 215, 225, 235, 245, or 250 mL) of a pharmaceutical composition (e.g., a pharmaceutical composition having a concentration of CD101 at 0.4 mg/mL, 0.8 mg/mL, or 1 .6 mg/mL) is administered to the subject in 30 minutes. In some embodiments, 125 mL to 250 mL (e.g., 125, 135, 145, 155, 165, 175, 1 85, 195, 205, 21 5, 225, 235, 245, or 250 mL) of a pharmaceutical composition (e.g., a pharmaceutical composition having a concentration of CD101 at 0.4 mg/mL, 0.8 mg/mL, or 1 .6 mg/mL) is administered to the subject in 60 minutes. In some embodiments, 250 mL of a pharmaceutical composition (e.g., a pharmaceutical composition having a concentration of CD101 at 0.4 mg/mL, or 0.8 mg/mL, or 1 .6 mg/mL) is administered to the subject in 30 minutes. In some embodiments, 250 mL of a pharmaceutical composition (e.g., a pharmaceutical composition having a concentration of CD101 at 0.4 mg/mL, 0.8 mg/mL, or 1 .6 mg/mL) is administered to the subject in 60 minutes.

Furthermore, in some embodiments, at least one dose (e.g., a dose between 50 mg and 800 mg of CD101 ; about 50, 60, 70, 80, 90, 100, 1 10, 120, 130, 140, 150, 160, 170, 180, 190, 200, 210, 220, 230, 240, 250, 260, 270, 280, 290, 300, 310, 320, 330, 340, 350, 360, 370, 380, 390, 400, 41 0, 420, 430, 440, 450, 460, 470, 480, 490, 500, 51 0, 520, 530, 540, 550, 560, 570, 580, 590, 600, 610, 620, 630, 640, 650, 660, 670, 680, 690, 700, 710, 720, 730, 740, 750, 760, 770, 780, 790, or 800 mg of CD101 ) may be intravenously administered to the subject every 5 to 1 5 days (e.g., every 5, 6, 7, 8, 9, 10, 1 1 , 12, 13, 14, or 15 days). In some embodiments, one dose (e.g., a dose between 50 mg and 800 mg of CD101 ; about 50, 60, 70, 80, 90, 100, 1 10, 120, 130, 140, 150, 160, 1 70, 180, 190, 200, 210, 220, 230, 240, 250, 260, 270, 280, 290, 300, 310, 320, 330, 340, 350, 360, 370, 380, 390, 400, 410, 420, 430, 440, 450, 460, 470, 480, 490, 500, 510, 520, 530, 540, 550, 560, 570, 580, 590, 600, 610, 620, 630, 640, 650, 660, 670, 680, 690, 700, 710, 720, 730, 740, 750, 760, 770, 780, 790, or 800 mg of CD101 ) may be intravenously administered to the subject every 5 to 1 5 days (e.g., every 5, 6, 7, 8, 9, 10, 1 1 , 12, 13, 14, or 15 days).

In some embodiments of the methods of the invention, the subject may be immunocompromised, and thus, is at a higher risk for developing a fungal infection. In some embodiments, the subject is being prepared for an invasive medical procedure or undergoing long term antibiotic therapy. In some embodiments, the subject has been diagnosed with humoral immune deficiency, T cell deficiency, neutropenia, asplenia, or complement deficiency. In some embodiments, the subject is being treated or is about to be treated with immunosuppresive drugs. In some embodiments, the subject has been diagnosed with a disease which causes immunosuppression (e.g., cancer or acquired immunodeficiency syndrome). In some embodiments, the subject has cancer (e.g., leukemia, lymphoma, or multiple myeloma). In some embodiments, the subject has undergone or is about to undergo immunosuppression therapy. In some embodiments, the subject has undergone or is about to undergo hematopoietic stem cell transplantation. In some embodiments, the subject has undergone or is about to undergo an organ transplant. In some embodiments, subjects may receive prophylaxis treatment while being prepared for an invasive medical procedure (e.g., preparing for surgery, such as receiving a transplant, stem cell therapy, a graft, a prosthesis, receiving long-term or frequent intravenous catheterization, or receiving treatment in an intensive care unit).

In some embodiments of the methods described herein, the fungal infection to be treated or prevented is selected from candidemia, invasive candidiasis, tinea capitis, tinea corporis, tinea pedis, onychomycosis, perionychomycosis, pityriasis versicolor, oral thrush, vaginal candidiasis, respiratory tract candidiasis, biliary candidiasis, eosophageal candidiasis, urinary tract candidiasis, systemic candidiasis, mucocutaneous candidiasis, aspergillosis, mucormycosis, paracoccidioidomycosis, North American blastomycosis, histoplasmosis, coccidioidomycosis, sporotrichosis, fungal sinusitis, or chronic sinusitis.

In some embodiments of the methods described herein, the fungal infection is candidemia or invasive candidiasis.

In some embodiments of the methods described herein, the fungal infection to be treated or prevented is an infection of Candida albicans, C. glabrata, C. dubliniensis, C. krusei, C. parapsilosis, C. tropicalis, C. orthopsilosis, C. guilliermondii, C. rugosa, C. auris, C. lusitaniae, Aspergillus fumigatus, A. flavus, A. terreus, A. niger, A. candidus, A. clavatus, or A. ochraceus. The following examples are put forth so as to provide those of ordinary skill in the art with a complete disclosure and description of how the methods and compositions claimed herein are performed, made, and evaluated, and are intended to be purely exemplary of the disclosure and are not intended to be limiting. Examples

Example 1 : Solubility of CD101 in various media

Table 1 below provides the solubility of CD101 acetate in different solutions.

Table 1

Medium Solubility (mg/mL)

0.9% Saline Solution (USP), 4% dodecylmaltoside (DDM),

>10 *

1 % Tween 20

0.9% Saline Solution (USP), 7% tetradecylmaltoside (TDM) >1 1 *

* Maximum solubility was not reached

1 - IV injection solution

CD101 acetate is soluble (>1 mg/mL) in 5% dextrose, 5% mannitol and 0.22% - 0.45% Sterile Saline Solution, but demonstrated a significant decrease in solubility at higher salt concentrations such as normal saline (0.9% Sterile Saline Solution) or Lactated Ringer's Solution and is nearly insoluble in PBS buffer. The addition of a non-ionic surfactant such as polysorbate 20 (Tween 20) and polysorbate 80 (Tween 80) improves the solubility of CD101 acetate in high salt solutions such as 0.9% Sterile Saline, neutral pH phosphate buffer media, and also in Lactated Ringer's solution.

Example 2: Investigation of Use of Tween 80 for Use in CD101 Acetate Formulations

Methods

CD101 acetate 20 mg/kg dose was prepared in w/w in 5% dextrose solution (CD1 01 acetate at 4 mg/mL solution) for the following solutions using the salt to free base correction factor of 0.8784 for CD101 acetate:

CD101 acetate control:

No Tween 80 used. The pH of the solution is 5.14 (22.7 mg CD101 acetate into 5 mL 5% Dextrose) CD101 acetate 0.5% Tween 80:

0.5% w/w Tween 80 used. The pH of the solution is 5.21 (22.8 mg CD101 acetate into 5 mL 5% Dextrose with 0.5% w/w Tween 80 (0.1 g Tween 80 + 1 9.9 g 5% Dextrose))

CD101 acetate 1.0% Tween 80:

1 .0% w/w Tween 80 used. The pH of the solutions is 5.27 (22 .9 mg CD101 acetate into 5 mL

5% Dextrose with 1 .0% w/w Tween 80 (0.2 g Tween 80 + 19.8 g 5% Dextrose))

CD101 acetate 2.5% Tween 80:

2.5% w/w Tween 80 used. The pH of the solutions is 5.29 (22.7 mg CD101 acetate into 5 mL 5% Dextrose with 2.5% w/w Tween 80 (0.5 g Tween 80 + 1 9.5 mL 5% Dextrose))

CD101 acetate 1.0% Tween 80, pH adjusted to 6.64:

1 .0% w/w Tween 80 used. The pH of the solution was adjusted to 6.64 using 0.1 N NaOH. (22.9 mg CD101 acetate into 5 mL 5% Dextrose with 1 .0% w/w Tween 80 (0 .2 g Tween 80 + 1 9.8 g 5% Dextrose)) 1 .5 ml_ each of CD101 acetate 20mg/kg dose (CD101 acetate at 4 mg/mL solution) prepared above was added via syringe drop wise to the following solutions and were observed for drug insolubility in the solution (A) 5 mL 0.9% saline solution, or (B) 5 mL PBS buffer. Results

CD101 acetate prepared in 1 .0% Tween 80 w/w in 5% dextrose solution (CD101 acetate at 4 mg/mL), pH 5.4 solution and added to (A) 5 mL 0.9% saline solution and (B) 5 mL PBS buffer: both (A) and (B) were clear after CD101 acetate addition. CD101 acetate prepared in 1 .0% Tween 80 w/w in 5% dextrose solution (CD101 acetate at 4 mg/mL), adjusted to pH 6.64 using 0.1 N NaOH solution and added to (A) 5 mL 0.9% saline solution and (B) 5 mL PBS buffer: both (A) and (B) were clear after CD101 acetate addition. The results are summarized in Table 2 below.

Table 2

Example 3: PBS Buffer Drop Test

Different concentrations of CD101 acetate were prepared in the various vehicles listed in Tables 2-5. These solutions were tested for solubility in the PBS drop test, which involved adding the solution dropwise into PBS solution (1 :4.3 dilution) and visually observing the resulting solution for any precipitation. PBS buffer serves as a surrogate for plasma, and the PBS drop test is used to evaluate the potential for insolubility as the drug enters the bloodstream at the injection site.

Table 3 below provides the solubility observations of CD101 acetate in different IV infusion solutions in the presence and absence of Tween 80 in a PBS drop test. Table 3

* First CD101 acetate and Tween 80 were mixed together and then 0.9% saline solution was added.

Table 4 below provides the solubility observations of CD101 acetate in different 5% Dextrose IV infusion solutions in a PBS drop test.

Table 4

Table 5 below provides the solubility observations of CD101 acetate in different 5% Mannitol IV infusion solutions in a PBS drop test. Table 5

Conclusion

The solubility studies of CD101 intravenous (IV) formulations when introduced into PBS buffer solutions demonstrated that higher Tween 80 concentrations facilitated better solubility. A 6 mg/mL CD101 solution formulated in 1 .25% Tween 80, 5% mannitol, 0.3% glacial acetic acid, pH -4.5 vehicle remained soluble in PBS for at least 1 hour at room temperature. The data indicate that a Tween 80 concentration greater than 1 % w/w or w/v is required to avoid insolubility of CD101 in PBS buffer, which is a surrogate for plasma. Precipitation of CD101 at the site of injection may result in irritation in the tissue at the injection site and the vein leading away from the injection site. Example 4: Solubility of CD101 in intravenous (IV) formulations

Solubility of CD101 -containing formulations with different percentages of Tween 80 (also referred to as polysorbate 80) was evaluated in a "PBS drop test." This involved preparing a known concentration of CD101 in a formulation and injecting the formulation into PBS to visually examine for precipitation. This test was used to assess if a CD101 -containing formulation would remain soluble in blood during IV infusions.

Vehicle formulations tested

(1 ) 1 .25% Tween 80, 5% mannitol, 0.3% glacial acetic acid, pH -4.5

(2) 0.50% Tween 80, 0.76% mannitol, 0.125% sodium lactate in 3.5% dextrose solution, pH -5.5

Different concentrations of CD101 acetate were prepared in vehicles (1 ) and (2) and tested for solubility in the PBS drop test.

Dilution into PBS buffer reservoir (PBS drop test)

Add dropwise the formulated solution into PBS solution (1 :4 dilution). Visually observe the resulting solution upon addition for any precipitation.

Results

1.25% Tween 80, 5% mannitol, 0.3% glacial acetic acid, pH -4.5

One mL of a 6 mg/mL CD101 acetate solution was prepared in the formulation (1 .25% Tween 80, 5% mannitol, 0.3% glacial acetic acid, pH -4.5). Following formulation, 0.25 mL of the formulated solution was introduced into 1 mL of the PBS solution (PBS drop test). The PBS drop test demonstrated that the formulated 6 mg/mL solution remained soluble in PBS for at least 1 hour. This test was repeated to confirm the observation.

0.50% Tween 80, 0.76% mannitol, 0. 125%, sodium lactate in 3.5% dextrose solution, pH -5.5 One mL of a 2 mg/mL CD101 acetate solution was prepared in the formulation (0.50% Tween 80, 0.76% mannitol, 0.125% sodium lactate in 3.5% dextrose solution, pH -5.5. Following formulation, 0.25 mL of the formulated solution was introduced into 1 mL of the PBS solution (PBS drop test). The PBS drop test demonstrated that the formulated 2 mg/mL solution remained soluble in PBS for at least 1 hour. This test was repeated to confirm the observation.

One mL of a 3 mg/mL CD101 solution was prepared in the same formulation (0.50% Tween 80, 0.76% mannitol, 0.125% sodium lactate in 3.5% dextrose solution, pH -5.5). Following formulation, 0.25 mL of the formulated solution was introduced into 1 mL of the PBS solution (PBS drop test). The PBS drop test indicated that the formulated 3 mg/mL solution remained soluble in PBS for 15 to 30 minutes. This test was repeated to confirm the observation.

Conclusion

The solubility of CD101 IV formulations when introduced into phosphate-buffered saline (PBS) indicated that higher Tween 80 concentrations facilitated better solubility. A 6 mg/mL CD101 acetate solution formulated in 1 .25% Tween 80, 5% mannitol, 0.3% glacial acetic acid, pH -4.5 vehicle remained soluble in PBS for at least 1 hour. A lower concentration (2 mg/mL) CD101 solution formulated in 0.50% Tween 80, 0.76% mannitol, 0.125% sodium lactate in 3.5% dextrose solution, pH -5.5 remained soluble in PBS for at least 1 hour. In contrast, a 3 mg/mL CD101 solution prepared in the 0.50% Tween 80, 0.76% mannitol, 0.125% sodium lactate in 3.5% dextrose solution, pH -5.5 was soluble in PBS for only 15 to 30 minutes.

Example 5: 14-Day Repeat-Dose Intravenous Infusion Toxicity Study with CD101 acetate in Sprague Dawley Rats

The primary purpose of this study was to evaluate the toxicity of CD101 when administered intravenously as a 20-minute infusion into the lateral tail vein of Sprague Dawley rats for fourteen consecutive days. A positive control group (Eraxis ® ) with a similar structure and pharmacodynamic activity was included in the study design.

Methods

The test article, CD101 acetate, was a white to off-white powder. The test article was prepared into dosing formulations for intravenous dose administration. One hundred experimentally na ' ive Sprague Dawley rats (50 males and 50 females), a minimum of 8 weeks old and weighing 255-318 grams (males) and 192-215 (females) at the outset of the study were assigned to treatment groups as shown in Table 7 below:

Table 7

@ The total volume administered over a 20-minute continuous infusion was less than the maximum allowable volume of 1 mL/min in the rat species

* These dose levels represent the salt free (base) form of the test article

# Each Eraxis ® vial contained 1 00 mg of Anidulafungin in lyophilized form

All animals were dosed on Days 1 through 3 inclusive. As a result of tail vein irritation observed among all study groups, dose administration was suspended on Day 4 to allow the tails to recover. Dosing resumed on Day 5 and continued for a maximum of ten additional days (Days 5 through 14 inclusive). Animals that could no longer be dosed during this time remained on study.

Results and Conclusion

Male and female Sprague Dawley rats were to receive CD101 acetate at doses of 0, 2, 6 and 20 mg/kg/day (Groups 1 , 3, 4 and 5, respectively) or Eraxis ® (the positive control article, Group 2) at a dose of 40 mg/kg/day, intravenously as a 20 minute infusion into the left or right lateral tail vein for fourteen consecutive days. During the first three treatment days, all animals were dosed with the first vehicle used on this study (1 .75% Tween 80/5% Dextrose: Groups 1 and 3-5; 5% Dextrose: Group 2). As a result of the localized tail swelling observed during the first three days of treatment, study Day 4 served as a drug- free day to alleviate tail vein irritation. Dose administration resumed on Day 5 using a new vehicle (5% Mannitol/2.5% Tween 80/0.3% acetic acid: Groups 1 and 3-5; 5% Mannitol/0.3% acetic acid: Group 2) for the remainder of the treatment period (Days 5-14). Those animals that could no longer be dosed because of severe vein irritation were retained on study, untreated, until the end of the study (Day 1 5).

All animals were dosed on Days 1 -3. Beginning on Day 5, some of the animals could no longer be dosed as a result of tail swelling/lesions. Ten percent (10%) of vehicle control males, thirty percent (30%) of Positive Control (Group 2) males and 30% of high dose males (Group 5) were dosed for less than the intended ten-day duration with the second vehicle control used on Days 5-14. Ten percent (10%) of females treated with the low dose (Group 3), 10% with the mid-dose (Group 4), and 90% with the high dose (Group 5) did not receive ten consecutive days of treatment with the second vehicle from Days 5-14.

Clinical signs indicative of tail vein irritation were recorded for all animals on this study (Groups 1 - 5) and included a combination of the following: a red or purple discolored tail, the presence of white and/or yellow patches on the tail, a swollen tail, and abrasions or scabs. The incidence of tail vein irritation when using the first vehicle during Days 1 -3 (1 .75% Tween 80 and 5% Dextrose: Groups 1 and 3-5; 5% Dextrose: Group 2) was highest for males and females treated with Eraxis® and CD1 01 acetate at doses of 6 and 20 mg/kg/day. Distinctions between dose groups when using the second vehicle during Days 5-14 (5% Mannitol, 2.5% Tween 80, and 0.3% Acetic Acid: Groups 1 and 3-5); 5% Mannitol and 0.3% Acetic Acid: Group 2) were not as evident as when using the first vehicle. The vein irritation observed during Days 5-14 was only marginally higher for Group 2 (Eraxis ® ) and Group 5 (CD101 acetate at 20 mg/kg/day) compared to the vein irritation for Group 1 and test article-treated Groups 3 and 4 during Days 5-14.

Example 6: A Single-Dose Intravenous Infusion Tolerability Study of CD101 in Cynomolgus Monkeys

The objective of this study was to evaluate and compare the tolerability of CD101 when administered once by either a 20-minute or 60-minute intravenous infusion to cynomolgus monkeys. In addition, a vehicle similar to the intended initial clinical formulation was assessed when administered once by 20-minute intravenous infusion up to the highest feasible dose (1 0 mg/kg). Methods

This study was divided into 2 phases (Table 8). In phase 1 , CD101 acetate was administered in vehicle-1 (1 .25% Tween 80, 5% mannitol, 0.3% glacial acetic acid adjusted to pH 4.5 ± 0.1 ) as a single 20-minute (± 2 minutes) intravenous infusion to 1 animal/sex/group at 0 and 30 mg/kg (Groups 1 and 2, respectively). In addition, CD101 acetate was administered in vehicle-1 as a single 60-minute (± 5 minutes) intravenous infusion to 1 animal/sex/group at 0 and 30 mg/kg (Groups 3 and 4, respectively). In phase 2, CD1 01 acetate was administered in vehicle-2 (0.50% Tween 80, 0.76% mannitol, 0.125% sodium lactate in 3.5% dextrose solution, pH 5.5 ± 0.1 ) as a single 20-minute (± 2 minutes) intravenous infusion to 1 animal/sex at 10 mg/kg (Group 5). Dosing was staggered so that Group 5 was dosed 2 days after Groups 1 -4.

Table 8

a = The test article formulations were corrected for purity/potency using a correction factor of 1 .183. b = Dose volume delivered by infusion over a 20-minute (± 2 minutes) period for Groups 1 , 2, and 5 and over a 60-minute (± 5 minutes) period for Groups 3 and 4.

Results

No other remarkable clinical observations were observed and there were no differences that could be attributed to the duration of infusion (i.e., 20 minutes versus 60 minutes). Conclusion

The results of this study indicate no differences between the tolerability of a 20-minute and 60-minute infusion. In addition, use of a vehicle can be administered safely to cynomolgus monkeys up to the highest feasible dose (1 0 mg/kg) in the vehicle. Example 7. Efficacy CD101 in the treatment of Candida auris infection in a murine model of disseminated candidiasis

Methods

Female 6-8 week old CD-1 mice were immunosuppressed with cyclophosphamide (200 mg/kg) 3 days prior to infection and 150 mg/kg 1 day post-infection. On the day of infection, mice were inoculated with 3 x 10 7 C. auris blastospores via the lateral tail vein. Mice were randomized into 5 groups (n=5 for colony forming units (CFU) and n=10 for survival): CD101 20 mg/kg administered by intraperitoneal (IP) injection, fluconazole 20 mg/kg administered per os (PO), amphotericin B 0.3 mg/kg IP, and a vehicle control. Treatments were administered 2 hours post-infection (day 1 ) and again on day 4 of the study for a total of 2 doses. Mice were monitored daily and a survival curve was generated. CFU groups were sacrificed on day 8 of the study. One kidney was removed from each mouse, homogenized, plated on potato dextrose agar (PDA), and incubated at 35 °C for 2 days to determine CFU. The remaining survival mice were monitored until the end of the study (day 14). Results

CD101 showed an average 3 log reduction in kidney CFU compared to fluconazole, amphotericin B, and vehicle treated groups, which was statistically significant (P = 0.03, 0.03, and 0.04, respectively). At the end of the study, percent survival of mice in CD101 , fluconazole, amphotericin B, vehicle, and untreated groups was 80, 0, 30, 20, and 0%, respectively (FIG. 1 ).

Conclusion

Taken together, our findings show that CD101 possesses potent antifungal activity against C. auris infection in a disseminated model of candidiasis. Additionally, treatment with CD101 resulted in a significantly higher overall percent survival.

Example 8. Evaluate the ability of CD101 to prevent and treat Candida albicans biofilms and explore its temporal effect by time lapse photography

In this study, we determined the effect of CD101 on prevention and treatment of biofilms formed by Candida albicans in vitro, and evaluated the effect of CD101 (at effective concentration) on formation of biofilm in real time using Time Lapse Microscopy (TLM).

Materials and Methods

Test Compounds

CD101 powder stocks were reconstituted in water or Yeast Nitrogen Base (YNB) medium, and diluted in YNB to a final working concentration of 0.25 μg/ml and 1 μg/ml. YNB with no CD1 01 was prepared in parallel and used as controls. Fluconazole was used as a comparator.

Test Media

YNB and Sabouraud dextrose agar (SDA) media

CD101 (powder and reconstituted solution) stored at -80 °C when not in use.

Strains

C. albicans SC-5314 was used for the current study.

Activity of CD 101 Against Candida biofilms

In this study, biofilms were grown in vitro using a biofilm model (Chandra et al., Nature Protocols 3:1909, 2008) and the effect of CD101 on adhesion phase biofilms (representing prevention of biofilms) or mature phase biofilms (representing treatment of biofilms) was determined.

Activity Against Adhesion Phase (Prevention) or Mature Phase (Treatment) Biofilms

Biofilms were formed on silicone elastomer (SE) discs using a catheter-associated-biofilm model (Chandra et al., Nature Protocols 3:1909, 2008; Chandra et al., J. Bacteriol. 183: 5385, 2001 ; Chandra et al., J. Dental Research 80: 903, 2001 ). For evaluation of activity against adhesion phase biofilms (prevention), Candida cells were adhered to catheter discs for 90 min. Next, discs were incubated for 24 h with CD101 (0.25 or 1 μg/ml concentrations) to allow biofilm formation. For evaluation of activity against mature phase biofilms (treatment), Candida cells were adhered to catheter discs for 90 min, then transferred to fresh media and incubated for a further 24 h to allow formation of biofilms. Mature biofilms were then exposed to CD101 (0.25 or 1 μg/ml concentrations) for another 24 h. Discs incubated with fluconazole or media alone were used as controls in all experiments.

At the end of drug exposure in both adhesion and mature phase biofilms, biofilms were quantified by measuring their metabolic activity using XTT assay (Chandra et al., Nature Protocols 3:1909, 2008; Chandra et al., J. Bacteriol. 183: 5385, 2001 ; Chandra et al., J. Dental Research 80: 903, 2001 ).

Following incubation with drugs, discs were transferred to fresh plates containing phosphate buffered saline with XTT and menadione, incubated for 3 h at 37 S C and optical density was read at 492 nm. Separate batches of biofilms were stained with fluorescent dyes (FUN1™, CONA) and observed under Confocal Scanning Laser Microscope (CSLM) to evaluate biofilm architecture and thickness (Chandra et al., Nature Protocols 3:1909, 2008; Chandra et al., J. Bacteriol. 183: 5385, 2001 ).

Time Lapse Microscopy

The effective CD101 concentration obtained from the above experiments was used to monitor its effect on biofilm formation in real time using TLM, which involves capturing real-time images of a single frame at specific time intervals, allowing temporal monitoring of the interactions occurring between the drug and Candida biofilms. Captured images were combined in a time sequence, resulting in an animation depicting the sequence of events that occurred with the passage of time. Briefly, the discs with C. albicans (adhered for 90 min as above) were placed in a 35-mm-diameter glass-bottom Petri dish (MatTek Corp., Ashland, MA). Next, CD101 (dissolved in the growth medium) was added to the Petri dish, and incubated at 37°C to allow formation of biofilm. Phase contrast images for this interaction were captured immediately from 0 h and followed up to 16-1 7 h on a Leica DMI 6000 B inverted microscope connected to a Retiga EXi Aqua camera (Q-imaging Vancouver British Columbia). To determine the structural changes in the maturing biofilm, both acquisition and analysis of a series of horizontal (xy) optical sections of the biofilm was done using Metamorph Imaging software (Molecular Devices, Downington, PA). Disc incubated with media alone was used as control.

Statistical Analyses

Statistical analyses for all data were performed using GraphPad Prism 6 software. Drug treated groups were compared to control untreated groups using unpaired t-tests. P-value of <0.05 was considered significant.

Results

Activity Against Adhesion Phase Biofilms (Prevention)

Our metabolic activity and CSLM results showed that CD101 prevented formation of robust biofilms at both concentrations tested (0.25 and 1 μg/ml). Assessment of metabolic activity revealed that

C. albicans treated with CD101 formed significantly less biofilms compared to untreated C. albicans (FIG.

2A, P < 0.05), In contrast, fluconazole did not inhibit biofilm formation at the two concentrations tested (1 and 4 μg/ml, FIG. 2B, P > 0.05). CSLM images showed highly heterogeneous architecture of biofilms with cells/hyphae embedded within extracellular matrix for untreated control (FIG. 3A) while exposure to both concentrations of CD1 01 showed only remnants of adhered cells, and no biofilm maturation (FIGS. 3B and 3C). In contrast, fluconazole did not inhibit biofilm formation (FIGS. 3D and 3E). Additionally, exposure to CD101 significantly reduced the thickness of biofilms compared to untreated control (36 μιη vs. 4 μιτι, P < 0.05, FIG. 3F), while fluconazole had no effect on biofilm thickness (FIG. 3G).

Activity Against Mature Phase Biofilms (Treatment)

Metabolic activity and CSLM results showed that CD1 01 was active against mature biofilms at both tested concentrations (0.25 and 1 μς/ιηΙ). Mature C. albicans biofilms exposed to CD1 01 exhibited significantly less metabolic activity compared to those formed by untreated biofilms (FIG. 4A, P < 0.05). In contrast, neither concentrations of fluconazole (1 and 4 μς/ιηΙ) affected these biofilms (FIG. 4B, P > 0.05 compared to untreated controls). CSLM analyses showed highly heterogeneous architecture of biofilms for untreated control (FIG. 5A), while biofilm treated with CD101 were eradicated and showed bulged, deformed/broken cells (FIGS. 5B and 5C). In contrast, fluconazole did not affect Candida biofilms at both concentrations used (FIGS. 5D and 5E). Additionally, CD1 01 significantly reduced thickness of biofilms compared to untreated control (43 μιη vs. 24 μιτι, P < 0.05, FIG. 5F) while fluconazole had no effect (FIG. 5G).

Time lapse

Time lapse movies showed that untreated biofilms formed highly heterogeneous architecture of biofilms with cells/hyphae embedded within extracellular matrix (screen frames in FIGS. 6A and 6B). In contrast, biofilms exposed to 0.25 μg/ml CD1 01 showed only adhered cells with stunted growth, which failed to grow into mature biofilms (FIGS. 6C-6F). Under high magnification, bulging, deformed, and broken cells were clearly visible (arrows, FIGS. 6D and 6F). The effect of CD101 (0.25 ig/m\) was also studied on 3h formed biofilms and images were captured immediately after adding the drug and followed up to 16h. Screen frames in FIG. 7A showed 3h biofilm hyphal growth which after adding drug remained stunted and failed to grow into mature biofilms (FIG. 7B). Bulged, deformed, broken cells/hypha were clearly visible after 16h (arrows, FIG. 7B).

Conclusion

Our results demonstrate that CD1 01 possesses anti-biofilm activity against both adhesion phase and mature phase biofilms formed by C. albicans. This antifungal agent may have utility in both prevention and treatment of fungal biofilm infections.

Other Embodiments

All publications, patents, and patent applications mentioned in this specification are herein incorporated by reference to the same extent as if each independent publication or patent application was specifically and individually indicated to be incorporated by reference.

While the disclosure has been described in connection with specific embodiments thereof, it will be understood that it is capable of further modifications and this application is intended to cover any variations, uses, or adaptations of the disclosure following, in general, the principles of the disclosure and including such departures from the present disclosure that come within known or customary practice within the art to which the disclosure pertains and may be applied to the essential features hereinbefore set forth, and follows in the scope of the claims.

Other embodiments are within the claims.

What is claimed is: