[go: up one dir, main page]

WO2025101647A1 - Combination therapy of itraconazole and acylhydrazone-based antifungalagents for the treatment of sporotrichosis in cats - Google Patents

Combination therapy of itraconazole and acylhydrazone-based antifungalagents for the treatment of sporotrichosis in cats Download PDF

Info

Publication number
WO2025101647A1
WO2025101647A1 PCT/US2024/054777 US2024054777W WO2025101647A1 WO 2025101647 A1 WO2025101647 A1 WO 2025101647A1 US 2024054777 W US2024054777 W US 2024054777W WO 2025101647 A1 WO2025101647 A1 WO 2025101647A1
Authority
WO
WIPO (PCT)
Prior art keywords
compound
halogen
amount
itc
pharmaceutically acceptable
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Pending
Application number
PCT/US2024/054777
Other languages
French (fr)
Other versions
WO2025101647A9 (en
Inventor
Maurizio Del Poeta
Iwao Ojima
Christina LAZZARINI
Krupanandan HARANAHALLI
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Research Foundation of the State University of New York
Original Assignee
Research Foundation of the State University of New York
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Research Foundation of the State University of New York filed Critical Research Foundation of the State University of New York
Publication of WO2025101647A1 publication Critical patent/WO2025101647A1/en
Publication of WO2025101647A9 publication Critical patent/WO2025101647A9/en
Pending legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/10Antimycotics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/13Amines
    • A61K31/15Oximes (>C=N—O—); Hydrazines (>N—N<); Hydrazones (>N—N=) ; Imines (C—N=C)
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/496Non-condensed piperazines containing further heterocyclic rings, e.g. rifampin, thiothixene or sparfloxacin

Definitions

  • the present invention demonstrated the antifungal efficacy of the AH derivative D13 [4-bromo-N′-(3,5- dibromo-2-hydroxybenzylidene)-benzohydrazide] against both planktonic cells and biofilms formed by Sporothrix brasiliensis. Br 3).
  • the eff ect of D13 was then tested in combination with itraconazole (ITC), with or without potassium iodide (KI), in 10 cats with sporotrichosis refractory to the treatment of standard of care with ITC. Improvement or total clinical cure was achieved in five cases after 12 weeks of treatment.
  • ITC itraconazole
  • KI potassium iodide
  • This invention provides a first veterinary clinical study of an acylhydrazone antifungal (D13) combined with itraconazole against a dimorphic fungal infection, sporotrichosis, which is highly endemic in South America in animals and humans. Overall, the results show that the combination treatment was efficacious in ⁇ 50% of the infected animals. In addition, D13 was well tolerated during the course of the study. Thus, these yesults warrant the continuation of the research and development of this new class of antifungals.
  • D13 acylhydrazone antifungal
  • the present invention provides a method of treating fungal infection in a subject, wherein the method comprises administering to the subject a combination comprising an amount of itraconazole (ITC) and an amount of a compound, or a pharmaceutically acceptable salt thereof, wherein the compound has the following structure: , R 1 is -H, alkyl, alkenyl, or alkynyl; R 2 is -H, alkyl, alkenyl, or alkynyl; R 9 , R 10 , R 11 , and R 12 are each independently, H, halogen, -CN, -CF 3 , -OCF 3 , -NO 2 , alkyl, alkenyl, alkynyl, aryl, heteroaryl, cycloalkyl, heterocycloalkyl, -OH, -OAc, -OR 13 , -COR 13 , -SH, -SR 13 , - SO 2
  • the present invention provides a combination comprising an amount of itraconazole (ITC) and an amount of a compound, or a pharmaceutically acceptable salt thereof, wherein the compound has the following structure: , R 1 is -H, alkyl, alkenyl, or alkynyl; R 2 is -H, alkyl, alkenyl, or alkynyl; R 9 , R 10 , R 11 , and R 12 are each independently, H, halogen, -CN, -CF 3 , -OCF 3 , -NO 2 , alkyl, alkenyl, alkynyl, aryl, heteroaryl, cycloalkyl, heterocycloalkyl, -OH, -OAc, -OR 13 , -COR 13 , -SH, -SR 13 , - SO 2 R 13 , -SO 2 NR 14 R 15 , -NH 2 , -NHR 13 , -NR 14 R 15 , -
  • the present invention provides a pharmaceutical composition in unit dosage form, useful in treating a subject afflicted with fungal infection, which comprises: (a) an amount of ITC; (b) an amount of the compound described herein, or a pharmaceutically acceptable salt thereof; and (c) optionally an amount of KI; wherein the respective amounts of said compound, or a pharmaceutically acceptable salt thereof, said ITC and said KI, if present, in said composition are effective, upon concomitant administration to said subject of one or more of said unit dosage forms of said composition, to treat the subject.
  • the present invention provides a therapeutic package for dispensing to, or for use in dispensing to, a subject afflicted with fungal infection, which comprises: (a) one or more unit doses, each such unit dose comprising: i) an amount of ITC; ii) an amount of the compound described herein, or a pharmaceutically acceptable salt thereof; and iii) optionally an amount of KI; wherein the respective amounts of said compound, or a pharmaceutically acceptable salt thereof, said ITC and said KI, if present, in said unit doses are effective, upon concomitant administration to said subject, to treat the subject, and (b) a finished pharmaceutical container therefor, said container containing said unit dose or unit doses, said container further containing or comprising labeling directing the use of said package in the treatment of said subject.
  • FIG. 1 Effect of D13 on Sporothrix Biofilm.
  • Sporothrix yeasts were cultured in RPMI 1640 medium together with cat's claw fragments and incubated at 37°C for 3 days. Following this, the claws were washed 3x and subsequently treated with 10 ⁇ g/mL of D13 or ITC for another 3 days before being prepared for SEM analysis.
  • both the ATCC strain of S. brasiliensis and the clinically isolated (C.I 17522) strain displayed abundant yeasts and hyphae with a consistently uniform and intact surface.
  • cells from both strains treated with either D13 or ITC exhibited slight growth and irregularities on their surfaces.
  • Figure 2 Fungal load of Sporothrix biofilm on cat claw fragments. Mature Sporothrix biofilms grown for 3 days were treated with D13 or ITC at 10 ⁇ g/ml. After 3 days of incubation, the claws were washed and suspended in RMPI/PenStrep 1%. Claws were sonicated and vortexed to drop the cells from the biofilm. Aliquots were placed on BHI plates and incubated at 37°C for up to 3 days. Floating bars represent the mean, min to max value of three independent experiments. Statistical analysis was performed by One-way ANOVA and Sidak’s multiple comparison test, **, p ⁇ 0,0028; **** p ⁇ 0,0001. [0012] Figure 3.
  • Case 1 The cat presented no improvement of the cutaneous lesions on the head and on the thoracic limbs upon treatment with ITC and KI. After adding D13, the cat significantly improved and clinical cure was achieved after 8 months of treatment with ITC, KI and D13 combination.
  • Figure 4. Case 2. The cat presented no improvement of the cutaneous and nasal mucosal lesions upon treatment with ITC, TRB and KI. After adding D13, the cat significantly improved and clinical cure was achieved after 12 weeks of treatment with ITC, TRB, KI and D13 combination.
  • Figure 5. Case 3. The cat presented no improvement of the cutaneous and nasal mucosal lesions upon treatment with ITC.
  • Sporotrichosis is a mycotic disease that affects humans and animals and it is caused by pathogenic species of the genus Sporothrix, such as Sporothrix schenckii and Sporothrix brasiliensis. It is the most common subcutaneous mycosis(Poester VR, 2018). S. brasiliensis and S. schenckii has been described as the main worldwide causal agents of the mycosis in cats (Della Terra PP, 2017). However, in South America, and particularly in Brazil, S. brasiliensis is the most prevalent species (de Carvalho JA, 2021 and Boechat JS 2018), and in certain Asian countries, S.
  • schenckii is the most common species ( Han HS, 2021).
  • the cat is considered the most susceptible animal to Sporothrix infection, presenting various clinical forms, such as single skin lesion, multiple skin lesions, ulcers, or/and disseminated systemic infection. These clinical manifestations are often not responsive to treatment (Gremiao ID, 2015, Miranda LHM, 2016, de Souza EW, 2018, and Schubach TM, 2001).
  • the treatment of sporotrichosis in cats is an important measure of disease control, not only to reduce the animal burden but also to diminish or/and prevent the transmission from cats to humans (de Miranda LHM, 2018). However, the treatment is often protracted for long time (weeks or months), and because of this long treatment the cat owner compliance is low.
  • the fungus can also become refractory to the treatment.
  • cats do not respond well to the standard of care, resulting in therapeutic failure (da Rocha , 2018, Nakasu CCT, 2021, Gremiao IDF, 2022 and Silva FS, 2022 ).
  • the standard of care (SOC) includes itraconazole (ITC) alone or associated with potassium iodide (KI), and this regime is the most common therapeutic regimen in Brazil (Gremiao IDF, 2021 and Xavier MO, 2023 ).
  • Ketoconazole, sodium iodide, fluconazole, amphotericin B (AMB), terbinafine, posaconazole, local heat therapy, cryosurgery and surgical removal of the lesions have also been described for treating sporotrichosis in cats, but their efficacy varies greatly (16).
  • ITC is a triazole with a broad spectrum of antifungal activity that blocks the synthesis of ergosterol (major sterol component of fungal plasma membranes) through the inhibition of the fungal cytochrome P450-dependent enzyme lanosterol 14-a-demethylase (Como JA, 1994).
  • the decrease of ergosterol synthesis leads to a fungal growth arrest and not to cell death.
  • ITC is fungistatic and therapeutic failure with ITC is common (Gremiao IDF, 2022).
  • the testing of new and fungicidal antifungal agents is warranted (Johnson MD, 2010).
  • acylhydrazones are a new class of antifungal agent molecules, which target the vesicular transport and cell cycle progression of fungi and indirectly impact glucosylceramide (GlcCer) synthesis (Mor V, Rella A, 2015).
  • GlcCer glucosylceramide
  • toxic sphingolipids accumulate, such as sphingosine, leading to mitochondria and vesicular damage (Mor V, Rella A, 2015).
  • the acylhydrazone compounds are highly fungicidal and able to kill fungi within few hours (Lazzarini C, 2018).
  • acylhydrazone known as D13 [4-bromo-N′-(3,5-dibromo-2-hydroxybenzylidene)-benzohydrazide] showed strong antifungal activity with very low toxicity in mammalian cells (Lazzarini C, 2018).
  • D13 improved the efficacy of commercially available drugs against various fungi.
  • S. brasiliensis D13 promoted yeast disruption and ultrastructural changes (Artunduaga Bonilla JJ, Honorato L, 2021).
  • Pharmacokinetic studies of D13 were performed in a murine model and were very promising with high oral availability (Lazzarini C, 2018).
  • D13 exhibited strong activity against different strains of Sporothrix in vitro when used alone or in combination with ITC (Artunduaga Bonilla JJ, Honorato L, 2021). The D13 antifungal effectiveness was further displayed in a murine model of sporotrichosis (Artunduaga Bonilla JJ, Honorato L, 2021).
  • This invention performed a preliminary clinical study using D13 in cats affected by sporotrichosis. The study was only approved as a “compassionate drug use”, meaning D13 could only be administered after the SOC failed. As such, it was not possible to have a control group in which D13 was not administered. Out of ten cats that were enrolled, four cats were clinically cured and one significantly improved.
  • the present invention provides a method of treating fungal infection in a subject, wherein the method comprises administering to the subject a combination comprising an amount of itraconazole (ITC) and an amount of a compound, or a pharmaceutically acceptable salt thereof, wherein the compound has the following structure: , R 1 is -H, alkyl, alkenyl, or alkynyl; R 2 is -H, alkyl, alkenyl, or alkynyl; R 9 , R 10 , R 11 , and R 12 are each independently, H, halogen, -CN, -CF 3 , -OCF 3 , -NO 2 , alkyl, alkenyl, alkynyl, aryl, heteroaryl, cycloalkyl, heterocycloalkyl, -OH, -OAc, -OR 13 ,
  • R 9 and R 11 are each –H and R 10 and R 12 are each –Br, then A is other than para-bromophenyl, meta-bromophenyl, ortho-tolyl or 3-quinolinyl.
  • R 9 , R 11 and R 12 are each –H and R 10 is –Br, then A is other than 3,5-dibromo-ortho-hydroxyphenyl, para-bromophenyl, meta-bromophenyl or ortho-tolyl.
  • the aryl or heteroaryl is substituted with halogen, C 1 -C 6 alkyl, -OH, -O- (C 1 -C 6 alkyl), -CHF 2 ,-CF 3 , -OCHF 2 or -OCF 3 .
  • the fused aryl or fused heteroaryl is substituted with halogen, C 1 -C 6 alkyl, -OH, -O-(C 1 -C 6 alkyl), -CHF 2 , -CF 3 , -OCHF 2 or -OCF 3 .
  • the compound having the following structure wherein ndently, -H, halogen, C 1 -C 6 alkyl, -OH, -O-(C 1 -C 6 alkyl), - CHF 2 , -CF 3 , -OCHF 2, -OCF 3, -CN, -CH 2 OCH 3 , -N(CH 3 ) 2 , -CH 2 F, -N 3 or -CCH.
  • R 1 and R 2 are each H.
  • R 3 , R 4 , R 5 , R 6 and R 7 are each independently -H, halogen, C 1 -C 6 alkyl, -OH, -O-(C 1 -C 6 alkyl), -CHF 2 , -CF 3 , -OCHF 2 or -OCF 3 .
  • R 3 , R 4 , R 5 , and R 6 are each independently halogen, -O-(C 1 -C 6 alkyl), - OCF 3 or -CF 3 .
  • R 3 , R 4 , R 5 , and R 6 are each independently halogen or -O-(C 1 -C 6 alkyl).
  • R 3 , R 4 , R 5 , and R 6 are each independently –Cl, -Br, -F, -O-(C 1 -C 6 alkyl), -OCF 3 or -CF 3 .
  • R 3 , R 4 , R 5 , and R 6 are each independently –Cl, -Br, or -O-(C 1 -C 6 alkyl).
  • R 9 , R 10 , R 11 , and R 12 are each independently H, halogen, C 1 -C 6 alkyl, - OH, -O-(C 1 -C 6 alkyl), -CHF 2 , -CF 3 , -OCHF 2 or -OCF 3 .
  • R 9 , R 10 , R 11 , and R 12 are each independently halogen, -O-(C 1 -C 6 alkyl), -OCF 3 or -CF 3 .
  • R 9 , R 10 , R 11 , and R 12 are each independently halogen or -O-(C 1 -C 6 alkyl). [0038] In some embodiments, R 9 , R 10 , R 11 , and R 12 are each independently –Cl, -Br, -F, -O-(C 1 -C 6 alkyl), -OCF 3 or -CF 3 . [0039] In some embodiments, R 9 , R 10 , R 11 , and R 12 are each independently –Cl, -Br, or -O-(C 1 -C 6 alkyl).
  • At least one of R 3 , R 4 , R 5 , R 6 and R 7 is not H. [0041] In some embodiments, at least two of R 3 , R 4 , R 5 , R 6 and R 7 are not H. [0042] In some embodiments, at least one of R 9 , R 10 , R 11 , and R 12 is not H. [0043] In some embodiments, at least two of R 9 , R 10 , R 11 , and R 12 are not H. [0044] In some embodiments, R 3 , R 4 , R 6 , R 7 , R 10 and R 12 are H.
  • R 3 , R 4 , R 6 , R 9, R 10 and R 12 are H.
  • R 5 , R 9, and R 10 are halogen; preferably R 5 , R 9, and R 10 are Br.
  • R 5 , R 7, R 11 are halogen; preferably R 5 , R 9, and R 10 are Br.
  • R 1 is -H; R 2 is -H; R 3 , R 4 , R 5 , R 6 and R 7 are each, independently, H, halogen, C 1 -C 6 alkyl, -OH; and R 9 , R 10 , R 11 , and R 12 are each independently H, halogen, C 1 -C 6 alkyl, -OH.
  • R 1 is -H; R 2 is -H; R 3 , R 4 , R 5 , R 6 and R 7 are each, independently, H or halogen; and R 9 , R 10 , R 11 , and R 12 are each independently H or halogen.
  • R 1 is -H
  • R 2 is -H
  • R 3 , R 4 , R 5 , R 6 and R 7 are each, independently H or Br
  • R 9 , R 10 , R 11 , and R 12 are each independently H or Br.
  • the compound has the following structure: Br Br .
  • the compound has the following structure: Br r .
  • d has the following structure: Br O Br .
  • e compound has the following structure: R 11 is -H, -Br, or -NR 14 R 15 , R 10 , R 12 , and R 12 are each independently -H, -Br, -OH, -OR 13 , or -NR 14 R 15 ; preferably, R 10 , R 12 , and R 12 are each independently -H, -Br, -OH, -OCH 3 , or -N(CH 3 ) 2 . [0062] In some embodiments, the compound has the following structure: R 5 R 5 R 5 R 5 R R R R R R R R R R R 6 R 7 , R 5 R 5 R 4 R 6 R 4 R 6 7 .
  • pound has the following structure: Br Br CH 3 , [ ] n some em o ments, t e compoun avng t e o owng structure: R 10 R R 11 12 , wherein R 3 , R 4 , R 5 , R 6 and R 7 are each independently, -H, halogen, C 1 -C 6 alkyl, -OH, -O-(C 1 -C 6 alkyl), - CHF 2 , -CF 3 , -OCHF 2, -OCF 3, -CN, -CH 2 OCH 3 , -N(CH 3 ) 2 , -CH 2 F, -N 3 or -CCH.
  • R 3 , R 4 , R 5 , R 6 , and R 7 are each independently H, halogen, -CN, alkyl, alkenyl, alkynyl, aryl, heteroaryl, heterocycle, -OCF 3 , -OH, -OAc, -OR 13 , -COR 13 , -CH 2 OR 13 .
  • R 3 , R 4 , R 5 , R 6 , and R 7 are each independently, H, halogen, -OCF 3 , heterocycle , or -CH 2 OR 13 .
  • R 3 , R 4 , R 5 , R 6 , and R 7 are each independently, H or halogen.
  • the heterocycle is six-membered heterocycle. [0070] In some embodiments, the heterocycle is five-membered heterocycle. [0071] In some embodiments, the heterocycle is four-membered heterocycle. [0072] In some embodiments, the heterocycle is three-membered heterocycle. [0073] In some embodiments, R 3 is OH, R 4 , R 5 , R 6 and R 7 are each independently H or halogen.
  • R 3 is OH, R 4 , R 5 , and R 6 are H, R 7 is halogen; b) R 3 is OH, R 5 , R 6 , and R 7 are H, R 4 is halogen; c) R 3 is OH, R 4 , R 6 , and R 7 are H, R 5 is halogen; d) R 3 is OH, R 4 , R 5 , and R 7 are H, R 6 is halogen; e) R 3 is OH, R 4 and R 5 are halogen, R 6 and R 7 are H; f) R 3 is OH, R 4 and R 6 are halogen, R 5 and R 7 are H; g) R 3 is OH, R 4 and R 7 are halogen, R 5 and R 6 are H; h) R 3 is OH, R 5 and R 6 are halogen, R 4 and R 7 are H; i) R 3 is OH, R 5 and R 7 are halogen;
  • the halogen is F, Cl, Br, and I. [0076] In some embodiments, the halogen is Br. [0077] In some embodiments, R 7 is OH, R 3 , R 4 , R 5 and R 6 are each independently H or halogen.
  • R 7 is OH, R 4 and R 5 are halogen, R 3 and R 6 are H; b) R 7 is OH, R 4 and R 6 are halogen, R 3 and R 5 are H; c) R 7 is OH, R 3 and R 4 are halogen, R 5 and R 6 are H; d) R 7 is OH, R 3 and R 5 are halogen, R 4 and R 6 are H; e) R 7 is OH, R 3 and R 6 are halogen, R 4 and R 5 are H; f) R 7 is OH, R 5 and R 6 are halogen, R 3 and R 4 are H; g) R 7 is OH, R 3 , R 4 , and R 5 are H, R 6 is halogen; h) R 7 is OH, R 3 , R 4 , and R 6 are H, R 5 is halogen; i) R 7 is OH, R 4 , R 5 , and R 6 are H, R 5 is halogen;
  • the halogen is Br.
  • R 3 , R 4 , R 5 , R 6 , R 7 are each independently, H or CH 2 OR 13 .
  • a) R 3 is CH 2 OR 13 , R 4 , R 5 , R 6 , and R 7 are H;
  • R 4 is CH 2 OR 13 , R 3 , R 5 , R 6 , and R 7 are H;
  • c) R 5 is CH 2 OR 13 , R 3 , R 4 , R 6 , and R 7 are H;
  • R 6 is CH 2 OR 13 , R 3 , R 4 , R 5 , and R 7 are H;
  • e) R 7 is CH 2 OR 13 , R 3 , R 4 , R 5 , and R 6 are H;
  • f) R 3 and R 4 are CH 2 OR 13 , R 5 , R 6 , R 7 are H;
  • g) R 3 and R 5 are CH 2 OR 13 .
  • R 5 is CH 2 OR 13 and R 13 is branched or unbranched alkyl.
  • alkyl is C 1 -C 6 alkyl, branched or unbranched.
  • alkyl is methyl, ethyl, propyl, or isopropyl;
  • R 3 , R 4 , R 5 , R 6 , and R 7 are each independently, H, halogen or -OCF 3 .
  • R 3 is halogen, R 4 , R 5 , and R 6 are H, R 7 is -OCF 3 ;
  • R 3 is halogen, R 5 , R 6 , and R 7 are H, R 4 is -OCF 3 ;
  • R 3 is halogen, R 4 , R 6 , and R 7 are H, R 5 is -OCF 3 ;
  • R 3 is halogen, R 4 , R 5 , and R 7 are H, R 6 is -OCF 3 ;
  • e) R 4 is halogen, R 3 , R 5 , and R 6 are H, R 7 is -OCF 3 ;
  • R 4 is halogen, R 3 , R 5 , and R 7 are H, R 6 is -OCF 3 ;
  • R 4 is halogen, R 3 , R 5 , and R 7 are H, R 6 is -OCF 3 ;
  • g) R 4 is halogen, R 3 , R 6 , and R
  • the halogen is F, Cl, Br, and I. [0089] In some embodiments, the halogen is F. [0090] In some embodiments, R 3 , R 4 , R 5 , R 6 , and R 7 are each H or heterocycle.
  • R 3 is heterocycle, R 4 , R 5 , R 6 , and R 7 are H; b) R 4 is heterocycle, R 3 , R 5 , R 6 , and R 7 are H; c) R 5 is heterocycle, R 3 , R 4 , R 6 , and R 7 are H; d) R 6 is heterocycle, R 3 , R 4 , R 5 , and R 7 are H; or e) R 7 is heterocycle, R 3 , R 4 , R 5 , and R 6 are H.
  • heterocycle is six-membered heterocycle.
  • heterocycle is five-membered heterocycle.
  • heterocycle is four-membered heterocycle.
  • heterocycle is three-membered heterocycle.
  • the three-membered heterocycle is aziridine, 2H-azirine, oxirane, thiirane or azirine.
  • the azirine is 3-methyl-3-(trifluoromethyl)-3H-diazirine.
  • R 4 , R 5 , and R 6 are each H or heterocycle, R 7 is H.
  • a) R 4 is heterocycle, R 3 , R 5 , R 6 , and R 7 are H; b) R 5 is heterocycle, R 3 , R 4 , R 6 , and R 7 are H; or c) R 6 is heterocycle, R 3 , R 4 , R 5 , and R 7 are H.
  • heterocycle is six-membered heterocycle.
  • heterocycle is five-membered heterocycle.
  • heterocycle is four-membered heterocycle.
  • heterocycle is three-membered heterocycle.
  • three-membered heterocycle is aziridine, 2H-azirine, oxirane, thiirane or azirine.
  • the azirine is 3-methyl-3-(trifluoromethyl)-3H-diazirine.
  • the compound has the following structure: , g structure: , , . [0107] , , g structure: , . [0108] lowing structure: , g structure: , or . [0109]
  • the compound has the following structure: , wherein ring A has the following structure: , .
  • ompound has the following structure: , or , , , , , , , , , HO O o r
  • the compound has the following structure: , [0114] , , , , und, is administered to the subject in a capsule.
  • the amount of ITC and the amount of the compound, or pharmaceutically acceptable salt of the compound, when taken together are effective to achieve an additive or a greater than additive therapeutic result in treating the subject.
  • the amount of ITC and the amount of the compound, or pharmaceutically acceptable salt of the compound, when taken together are effective to achieve a synergism in treating the subject.
  • the combination is more effective in treating the subject against fungal infection than when ITC and the compound, or pharmaceutically acceptable salt of the compound, at the same amount is administered alone.
  • the amount of the ITC and the amount of the compound, or pharmaceutically acceptable salt of the compound, when administered together are more effective to treat the subject than when each at the same amount is administered alone.
  • the amount of the ITC administered in the combination is less than the fungicidally effective amount of the ITC when the ITC is administered alone.
  • the amount of the compound, or pharmaceutically acceptable salt thereof, administered in the combination is less than the fungicidally effective amount of the compound, or pharmaceutically acceptable salt thereof, when the compound, or pharmaceutically acceptable salt thereof, is administered alone.
  • the amount of ITC is administered in the range of 1mg/kg to 100 mg/kg subject body weight per day.
  • the amount of ITC is administered in the range of 1mg/kg to 70 mg/kg subject body weight per day.
  • the amount of ITC is administered in the range of 5mg/kg to 60 mg/kg subject body weight per day.
  • the amount of ITC is administered in the range of 10 mg/kg to 20 mg/kg subject body weight per day.
  • the amount of the compound, or pharmaceutically acceptable salt thereof is administered in the range of 1mg/kg to 100 mg/kg subject body weight per day.
  • the amount of the compound, or pharmaceutically acceptable salt thereof is administered in the range of 1mg/kg to 70 mg/kg subject body weight per day.
  • the amount of the compound, or pharmaceutically acceptable salt thereof is administered 5mg/kg to 60 mg/kg subject body weight per day.
  • the amount of the compound, or pharmaceutically acceptable salt thereof is administered10 mg/kg to 20 mg/kg subject body weight per day.
  • the ratio between the amount of ITC and the amount of the compound, or pharmaceutically acceptable salt thereof is from 100:1 to 1:100, 50:1 to 1:50, 20:1 to 1:20, 10:1 to 1:10, 5:1 to 1:5 or 2:1 to 1:2.
  • the ratio between the amount of ITC and the amount of the compound, or pharmaceutically acceptable salt thereof is 2:1.
  • the ratio between the amount of ITC and the amount of the compound, or pharmaceutically acceptable salt thereof is 5:1.
  • the ratio between the amount of ITC and the amount of the compound, or pharmaceutically acceptable salt thereof is from 100:1 to 1:2, 50:1 to 1:2, 20:1 to 1:2, 10:1 to 1:2, 5:1 to 1:2 or 2.5:1 to 1:2.5.
  • the ratio between the amount of ITC and the amount of the compound, or pharmaceutically acceptable salt thereof is from 1:100 to 1:2, 1:50 to 1:2, 1:20 to 1:2, 1:10 to 1:2, 1:5 to 1:2 or 1:2.5 to 1:2.
  • the amount of ITC and the amount of the compound, or pharmaceutically acceptable salt thereof is administered once, twice, or third times per day for a period of time.
  • the period is 4-72 weeks. [0137] In some embodiments, the period is 4-32 weeks. [0138] In some embodiments, the subject is a mammal. [0139] In some embodiments, the mammal is cat or human. [0140] In some embodiments, the mammal is cat. [0141] In some embodiments, the mammal is human. [0142] In some embodiments, the ITC and the compound, or pharmaceutically acceptable salt thereof, are administered together. [0143] In some embodiments, the ITC and the compound, or pharmaceutically acceptable salt thereof, are administered in a single dose. [0144] In some embodiments, the ITC and the compound, or pharmaceutically acceptable salt thereof, are administered separately.
  • the ITC and the compound, or pharmaceutically acceptable salt thereof are administered simultaneously. [0146] In some embodiments, the ITC and the compound, or pharmaceutically acceptable salt thereof, are applied contemporaneously. [0147] In some embodiments, the ITC and the compound, or pharmaceutically acceptable salt thereof, are applied successively. [0148] In some embodiments, the method further comprises administering to the subject potassium iodine (KI). [0149] In some embodiments, the method further comprises administering to the subject sylimarin. [0150] In some embodiments, the method further comprises administering to the subject terbinafine.
  • KI potassium iodine
  • the method further comprises administering to the subject sylimarin.
  • the amounts of ITC, KI and the compound, or pharmaceutically acceptable salt thereof, when taken together are effective to achieve an additive or a greater than additive therapeutic result in treating the subject.
  • the amounts of ITC, KI and the compound, or pharmaceutically acceptable salt thereof, when taken together are effective to achieve a synergism in treating the subject.
  • the combination is more effective in treating the subject against fungal infection than when ITC, KI, and the compound, or pharmaceutically acceptable salt thereof, at the same amount is administered alone.
  • the amount of the ITC, KI, and the amount of the compound, or pharmaceutically acceptable salt thereof, when administered together, are more effective to treat the subject than when each at the same amount is administered alone.
  • the amount of the ITC and KI administered in the combination is less than the fungicidally effective amount of the ITC and KI when the ITC and KI is administered alone.
  • the amount of the compound, or pharmaceutically acceptable salt thereof, administered in the combination is less than the fungicidally effective amount of the compound, or pharmaceutically acceptable salt thereof, when the compound, or pharmaceutically acceptable salt thereof, is administered alone.
  • the amount of ITC is administered in the range of 1mg/kg to 200 mg/kg subject body weight per day. [0158] In some embodiments, the amount of ITC is administered in the range of 10mg/kg to 150 mg/kg subject body weight per day. [0159] In some embodiments, the amount of ITC is administered in the range of 50mg/kg to 120mg/kg subject body weight per day. [0160] In some embodiments, the amount of ITC is administered in the range of 80mg/kg to 120mg/kg subject body weight per day. [0161] In some embodiments, the amount of ITC is administered in the range of 90mg/kg to 110mg/kg subject body weight per day.
  • the amount of ITC is administered in the range of 10mg/kg to 100 mg/kg subject body weight per day. [0163] In some embodiments, the amount of ITC is administered in the range of 20mg/kg to 100 mg/kg subject body weight per day. [0164] In some embodiments, the amount of ITC is administered in the range of 30mg/kg to 100 mg/kg subject body weight per day. [0165] In some embodiments, the amount of ITC is administered in the range of 40mg/kg to 100 mg/kg subject body weight per day. [0166] In some embodiments, the amount of ITC is administered in the range of 50mg/kg to 100 mg/kg subject body weight per day.
  • the amount of ITC is administered in the range of 60mg/kg to 100 mg/kg subject body weight per day. [0168] In some embodiments, the amount of ITC is administered in the range of 70mg/kg to 100 mg/kg subject body weight per day. [0169] In some embodiments, the amount of ITC is administered in the range of 80mg/kg to 100 mg/kg subject body weight per day. [0170] In some embodiments, the amount of ITC is administered in the range of 90mg/kg to 100 mg/kg subject body weight per day. [0171] In some embodiments, the amount of ITC is administered in the range of 1mg/kg to 70 mg/kg subject body weight per day.
  • the amount of ITC is administered in the range of 5mg/kg to 60 mg/kg subject body weight per day. [0173] In some embodiments, the amount of ITC is administered in the range of 10 mg/kg to 20 mg/kg subject body weight per day. [0174] In some embodiments, the amount of ITC is administered 50 mg/kg subject body weight per day. [0175] In some embodiments, the amount of ITC is administered 100 mg/kg subject body weight per day. [0176] In some embodiments, the amount of KI is administered in the range of 1mg/kg to 100 mg/kg subject body weight per day.
  • the amount of KI is administered in the range of 1mg/kg to 20 mg/kg subject body weight per day. [0178] In some embodiments, the amount of KI is administered in the range of 2 mg/kg to 16 mg/kg subject body weight per day. [0179] In some embodiments, the amount of KI is administered in the range of 2 mg/kg to 10 mg/kg subject body weight per day. [0180] In some embodiments, the amount of KI is administered in the range of 1 mg/kg to 8 mg/kg subject body weight per day. [0181] In some embodiments, the amount of KI is administered in the range of 1 mg/kg to 6 mg/kg subject body weight per day.
  • the amount of KI is administered in the range of 2 mg/kg to 5mg/kg subject body weight per day. [0183] In some embodiments, the amount of KI is administered in the range of 5 mg/kg to 10 mg/kg subject body weight per day. [0184] In some embodiments, the amount of KI is 2.5 mg/kg subject body weight per day. [0185] In some embodiments, the amount of the compound, or pharmaceutically acceptable salt thereof, is administered in the range of 1mg/kg to 100 mg/kg subject body weight per day. [0186] In some embodiments, the amount of the compound, or pharmaceutically acceptable salt thereof, is administered in the range of 1mg/kg to 70 mg/kg subject body weight per day.
  • the amount of the compound, or pharmaceutically acceptable salt thereof is administered in the range of 1 mg/kg to 70 mg/kg subject body weight per day. [0188] In some embodiments, the amount of the compound, or pharmaceutically acceptable salt thereof, is administered in the range of 5 mg/kg to 60 mg/kg subject body weight per day. [0189] In some embodiments, the amount of the compound, or pharmaceutically acceptable salt thereof, is administered in the range of 5 mg/kg to 50 mg/kg subject body weight per day. [0190] In some embodiments, the amount of the compound, or pharmaceutically acceptable salt thereof, is administered in the range of 5 mg/kg to 40 mg/kg subject body weight per day.
  • the amount of the compound, or pharmaceutically acceptable salt thereof is administered in the range of 5 mg/kg to 40 mg/kg subject body weight per day.
  • the amount of the compound, or pharmaceutically acceptable salt thereof is administered in the range of 10 mg/kg to 30 mg/kg subject body weight per day.
  • the amount of the compound, or pharmaceutically acceptable salt thereof is administered in the range of 10 mg/kg to 20 mg/kg subject body weight per day.
  • the amount of the compound, or pharmaceutically acceptable salt thereof is administered 20 mg/kg subject body weight per day.
  • the amount of sylimarin is administered in the range of 1mg/kg to 100 mg/kg subject body weight per day. [0196] In some embodiments, the amount of sylimarin is administered in the range of 1mg/kg to 70 mg/kg subject body weight per day. [0197] In some embodiments, the amount of sylimarin is administered in the range of 5 mg/kg to 60 mg/kg subject body weight per day. [0198] In some embodiments, the amount of sylimarin is administered in the range of 10 mg/kg to 50 mg/kg subject body weight per day.
  • the amount of sylimarin is administered in the range of 10 mg/kg to 40 mg/kg subject body weight per day. [0200] In some embodiments, the amount of sylimarin is administered in the range of 10 mg/kg to 30 mg/kg subject body weight per day. [0201] In some embodiments, the amount of sylimarin is administered in the range of 20 mg/kg to 40 mg/kg subject body weight per day. [0202] In some embodiments, the amount of sylimarin is administered in the range of 25 mg/kg to 35 mg/kg subject body weight per day. [0203] In some embodiments, the amount of sylimarin is administered 30 mg/kg subject body weight per day.
  • the amount of terbinafine is administered in the range of 1mg/kg to 100 mg/kg subject body weight per day. [0205] In some embodiments, the amount of terbinafine is administered in the range of 1mg/kg to 70 mg/kg subject body weight per day. [0206] In some embodiments, the amount of terbinafine is administered in the range of 5 mg/kg to 60 mg/kg subject body weight per day. [0207] In some embodiments, the amount of terbinafine is administered in the range of 10 mg/kg to 50 mg/kg subject body weight per day. [0208] In some embodiments, the amount of terbinafine is administered in the range of 10 mg/kg to 40 mg/kg subject body weight per day.
  • the amount of terbinafine is administered in the range of 10 mg/kg to 30 mg/kg subject body weight per day. [0210] In some embodiments, the amount of terbinafine is administered in the range of 20 mg/kg to 40 mg/kg subject body weight per day. [0211] In some embodiments, the amount of terbinafine is administered in the range of 25 mg/kg to 35 mg/kg subject body weight per day. [0212] In some embodiments, the amount of terbinafine is administered 30 mg/kg subject body weight per day.
  • the ratio between the amount of ITC and the amount of the compound, or pharmaceutically acceptable salt thereof is from 100:1 to 1:100, 50:1 to 1:50, 20:1 to 1:20, 10:1 to 1:10, 5:1 to 1:5 or 2:1 to 1:2. [0214] In some embodiments, the ratio between the amount of ITC and the amount of the compound, or pharmaceutically acceptable salt thereof, is from 100:1 to 1:2, 50:1 to 1:2, 20:1 to 1:2, 10:1 to 1:2, 5:1 to 1:2 or 2:1 to 1:2.
  • the ratio between the amount of ITC and the amount of the compound, or pharmaceutically acceptable salt thereof is from 2:1 to 1:100, 2:1 to 1:50, 2:1 to 1:20, 2:1 to 1:10, 2:1 to 1:5 or 2:1 to 1:2. [0216] In some embodiments, the ratio between the amount of ITC and the amount of the compound, or pharmaceutically acceptable salt thereof, is 2:1. [0217] In some embodiments, the ratio between the amount of ITC and the amount of the compound, or pharmaceutically acceptable salt thereof, is 5:1.
  • the ratio between the amount of ITC and KI is from 100:1 to 1:100, 50:1 to 1:50, 20:1 to 1:20, 10:1 to 1:10, 5:1 to 1:5 or 2:1 to 1:2. [0219] In some embodiments, the ratio between the amount of ITC and KI is from 100:1 to 1:2, 50:1 to 1:2, 20:1 to 1:2, 10:1 to 1:2, 5:1 to 1:2 or 2:1 to 1:2. [0220] In some embodiments, the ratio between the amount of ITC and KI is from 2:1 to 1:100, 2:1 to 1:50, 2:1 to 1:20, 2:1 to 1:10, 2:1 to 1:5 or 2:1 to 1:2.
  • the ratio between the amount of ITC and KI is 20:1.
  • the ratio between the amount of KI and the compound, or pharmaceutically acceptable salt thereof is from 100:1 to 1:100, 50:1 to 1:50, 20:1 to 1:20, 10:1 to 1:10, 5:1 to 1:5 or 2:1 to 1:2.
  • the ratio between the amount of KI and the compound, or pharmaceutically acceptable salt thereof is from 100:1 to 1:2, 50:1 to 1:2, 20:1 to 1:2, 10:1 to 1:2, 5:1 to 1:2 or 2:1 to 1:2.
  • the ratio between the amount of KI and the compound, or pharmaceutically acceptable salt thereof is from 2:1 to 1:100, 2:1 to 1:50, 2:1 to 1:20, 2:1 to 1:10, 2:1 to 1:5 or 2:1 to 1:2.
  • the ITC, KI and the compound, or pharmaceutically acceptable salt thereof are administered together.
  • the ITC, KI and the compound, or pharmaceutically acceptable salt thereof are administered in a single dose.
  • the ITC, KI and the compound, or pharmaceutically acceptable salt thereof are administered separately.
  • the ITC, KI and the compound, or pharmaceutically acceptable salt thereof are administered simultaneously.
  • the ITC, KI and the compound, or pharmaceutically acceptable salt thereof are applied contemporaneously.
  • the ITC, KI and the compound, or pharmaceutically acceptable salt thereof are applied successively.
  • the amount of ITC, KI and the compound, or pharmaceutically acceptable salt thereof is administered once twice, or third times per day for a period of time.
  • the period is 4-72 weeks.
  • the period is 4-32 weeks.
  • the period is 4 weeks.
  • the period is 3 months.
  • the period is 6 months.
  • the period is 8 months.
  • the subject is a mammal.
  • the mammal is cat or human.
  • the fungal infection is caused by Cryptococcus Neoformans, Cryptococcus gattii, Candida albicans, Candida krusei, Candida glabrata, Candida parapsilosis, Candida guilliermondii, Aspergillus fumigatus, Rhizopus oryzae, Rhizopus spp., Blastomyces dermatitis, Histoplasma capsulatum, Coccidioides spp., Paecilomyces variotii, Pneumocystis murina, Pneumocystis jiroveci, Histoplasma capsulatum, Aspergillus spp., S.
  • the fungal infection is caused by is Cryptococcus Neoformans or Sporothrix brasiliensis.
  • the fungal infection is caused by Sporothrix brasiliensis.
  • the present invention provides a combination comprising an amount of itraconazole (ITC) and an amount of a compound, or a pharmaceutically acceptable salt thereof, wherein the compound has the following structure: , R 1 is -H, alkyl, alkenyl, or alkynyl; R 2 is -H, alkyl, alkenyl, or alkynyl; R 9 , R 10 , R 11 , and R 12 are each independently, H, halogen, -CN, -CF 3 , -OCF 3 , -NO 2 , alkyl, alkenyl, alkynyl, aryl, heteroaryl, cycloalkyl, heterocycloalkyl, -OH, -OAc, -OR 13 , -COR 13 , -SH, -SR 13 , - SO 2 R 13 , -SO 2 NR 14 R 15 , -NH 2 , -NHR 13 , -NR 14 R 15 , -
  • the amount of the compound, or pharmaceutically acceptable salt thereof improves the fungicidal efficacy of ITC compared to when the same amount of the ITC is administered not in combination with the amount of the compound, or pharmaceutically acceptable salt thereof.
  • the amount of ITC improves the fungicidal efficacy of the compound, or pharmaceutically acceptable salt thereof, compared to when the same amount of the compound, or pharmaceutically acceptable salt thereof, is applied not in combination with the amount of ITC.
  • a substantially similar level of fungicidal efficacy is achieved by using a lesser amount of the ITC and/or the compound, or pharmaceutically acceptable salt thereof.
  • the amount of the compound, or pharmaceutically acceptable salt thereof is effective to increase sensitivity of the fungus to the amount of ITC compared to the sensitivity of the fungus to the amount of the ITC when it is administered not in combination with the amount of the compound, or pharmaceutically acceptable salt thereof.
  • the amount of the ITC is effective to increase sensitivity of the fungus to the amount of the compound, or pharmaceutically acceptable salt thereof, compared to the sensitivity of the fungus to the amount of the compound, or pharmaceutically acceptable salt thereof, when it is administered not in combination with the amount of ITC.
  • the amount of the compound, or pharmaceutically acceptable salt thereof is effective to increase bioavailability of the amount of ITC compared to the bioavailability of the amount of the ITC when it is administered not in combination with the amount of the compound, or pharmaceutically acceptable salt thereof.
  • the amount of ITC is effective to increase bioavailability of the amount of the compound, or pharmaceutically acceptable salt thereof, compared to the bioavailability of the amount of the compound, or pharmaceutically acceptable salt thereof, when it is administered not in combination with the amount of ITC.
  • the combination is more effective in treating the subject against fungal infection than when ITC and the compound, or pharmaceutically acceptable salt thereof, at the same amount is administered alone.
  • the combination prolongs the period of protection against fungal infection and/or control of fungal infection than when the amount of ITC and the amount of the compound, or pharmaceutically acceptable salt thereof, are administered alone. [0253] In some embodiments, the combination reduces the amount of time needed to achieve a level of fungal control than when the amount of ITC and the amount of the compound, or pharmaceutically acceptable salt thereof, are administered alone.
  • the present invention provides a pharmaceutical composition in unit dosage form, useful in treating a subject afflicted with fungal infection, which comprises: (a) an amount of ITC; (b) an amount of the compound described herein, or a pharmaceutically acceptable salt thereof; (c) optionally an amount of KI; wherein the respective amounts of said compound, or a pharmaceutically acceptable salt thereof, said ITC and said KI, if present, in said composition are effective, upon concomitant administration to said subject of one or more of said unit dosage forms of said composition, to treat the subject.
  • the present invention provides a therapeutic package for dispensing to, or for use in dispensing to, a subject afflicted with fungal infection, which comprises: (c) one or more unit doses, each such unit dose comprising: iv) an amount of ITC; v) an amount of the compound described herein, or a pharmaceutically acceptable salt thereof; vi) optionally an amount of KI; wherein the respective amounts of said compound, or a pharmaceutically acceptable salt thereof, said ITC and said KI, if present, in said unit doses are effective, upon concomitant administration to said subject, to treat the subject, and (d) a finished pharmaceutical container therefor, said container containing said unit dose or unit doses, said container further containing or comprising labeling directing the use of said package in the treatment of said subject.
  • the pharmaceutical composition, unit dose, or unit dosage form comprises: (a) 25-75 mg of the compound, or a pharmaceutically acceptable salt thereof, 10-100 mg of ITC, 1-20 mg of KI, if present. (b) 25-75 mg of the compound, or a pharmaceutically acceptable salt thereof, 10-100 mg of ITC, 1-20 mg of KI, if present and 11-50 mg of sylimarin, if present.
  • the pharmaceutical composition, unit dose, or unit dosage form comprises: (a) 25 mg of the compound, or a pharmaceutically acceptable salt thereof, 25 mg of ITC, 2.5 mg of KI and 30 mg sylimarin; (b) 20 mg of the compound, or a pharmaceutically acceptable salt thereof, 50 mg of ITC, and 2.5 mg of KI; and (c) 20 mg of the compound or a pharmaceutically acceptable salt thereof, and 100 mg of ITC.
  • the unit dosage form is administered to the subject in a capsule.
  • the synthetic schemes of compounds described herein can be found in International Publication No. WO 2016/094307, International Publication No. WO 2018/232298, and International Publication No.
  • adjectives such as “substantially” and “about” modifying a condition or relationship characteristic of a feature or features of an embodiment of the invention are understood to mean that the condition or characteristic is defined to within tolerances that are acceptable for operation of the embodiment for an application for which it is intended.
  • about means within a standard deviation using measurements generally acceptable in the art.
  • about means a range extending to +/- 10% of the specified value.
  • about includes the specified value.
  • the word “or” in the specification and claims is considered to be the inclusive “or” rather than the exclusive or, and indicates at least one of and any combination of items it conjoins.
  • each of the verbs, “comprise,” “include” and “have” and conjugates thereof, are used to indicate that the object or objects of the verb are not necessarily a complete listing of components, elements or parts of the subject or subjects of the verb.
  • Other terms as used herein are meant to be defined by their well-known meanings in the art.
  • a “symptom” associated with a fungal infection includes any clinical or laboratory manifestation associated with the fungal infection and is not limited to what the subject can feel or observe.
  • the compounds of the present invention include all hydrates, solvates, and complexes of the compounds used by this invention. If a chiral center or another form of an isomeric center is present in a compound of the present invention, all forms of such isomer or isomers, including enantiomers and diastereomers, are intended to be covered herein. Compounds containing a chiral center may be used as a racemic mixture, an enantiomerically enriched mixture, or the racemic mixture may be separated using well-known techniques and an individual enantiomer may be used alone.
  • the compounds described in the present invention are in racemic form or as individual enantiomers.
  • the enantiomers can be separated using known techniques, such as those described in Pure and Applied Chemistry 69, 1469–1474, (1997) IUPAC. In cases in which compounds have unsaturated carbon-carbon double bonds, both the cis (Z) and trans (E) isomers are within the scope of this invention.
  • the compounds of the subject invention may have spontaneous tautomeric forms. In cases wherein compounds may exist in tautomeric forms, such as keto-enol tautomers, each tautomeric form is contemplated as being included within this invention whether existing in equilibrium or predominantly in one form.
  • isomers arising from such asymmetry are included within the scope of this invention, unless indicated otherwise.
  • Such isomers can be obtained in substantially pure form by classical separation techniques and by stereochemically controlled synthesis, such as those described in "Enantiomers, Racemates and Resolutions" by J. Jacques, A. Collet and S. Wilen, Pub. John Wiley & Sons, NY, 1981.
  • the resolution may be carried out by preparative chromatography on a chiral column.
  • the subject invention is also intended to include all isotopes of atoms occurring on the compounds disclosed herein.
  • Isotopes include those atoms having the same atomic number but different mass numbers.
  • isotopes of hydrogen include tritium and deuterium.
  • isotopes of carbon include C-13 and C-14.
  • any notation of a carbon in structures throughout this application when used without further notation, are intended to represent all isotopes of carbon, such as 12 C, 13 C, or 14 C.
  • any compounds containing 13 C or 14 C may specifically have the structure of any of the compounds disclosed herein.
  • any notation of a hydrogen in structures throughout this application when used without further notation, are intended to represent all isotopes of hydrogen, such as 1 H, 2 H, or 3 H. Furthermore, any compounds containing 2 H or 3 H may specifically have the structure of any of the compounds disclosed herein.
  • Isotopically-labeled compounds can generally be prepared by conventional techniques known to those skilled in the art using appropriate isotopically-labeled reagents in place of the non-labeled reagents employed.
  • the substituents may be substituted or unsubstituted, unless specifically defined otherwise.
  • alkyl, heteroalkyl, monocycle, bicycle, aryl, heteroaryl and heterocycle groups can be further substituted by replacing one or more hydrogen atoms with alternative non-hydrogen groups.
  • substituents and substitution patterns on the compounds used in the method of the present invention can be selected by one of ordinary skill in the art to provide compounds that are chemically stable and that can be readily synthesized by techniques known in the art from readily available starting materials.
  • substituents are itself substituted with more than one group, it is understood that these multiple groups may be on the same carbon or on different carbons, so long as a stable structure results.
  • substituents i.e. R 1 , R 2 , etc. are to be chosen in conformity with well-known principles of chemical structure connectivity.
  • alkyl is intended to include both branched and straight-chain saturated aliphatic hydrocarbon groups having the specified number of carbon atoms.
  • C 1 -C n as in “C 1 –C n alkyl” is defined to include groups having 1, 2ising, n-1 or n carbons in a linear or branched arrangement, and specifically includes methyl, ethyl, propyl, butyl, pentyl, hexyl, heptyl, isopropyl, isobutyl, sec-butyl and so on.
  • An embodiment can be C 1 -C 12 alkyl, C 2 -C 12 alkyl, C 3 -C 12 alkyl, C 4 -C 12 alkyl and so on.
  • Alkoxy represents an alkyl group as described above attached through an oxygen bridge.
  • alkenyl refers to a non-aromatic hydrocarbon radical, straight or branched, containing at least 1 carbon to carbon double bond, and up to the maximum possible number of non- aromatic carbon-carbon double bonds may be present.
  • C 2 -C n alkenyl is defined to include groups having 1, 2...., n-1 or n carbons.
  • C 2 -C 6 alkenyl means an alkenyl radical having 2, 3, 4, 5, or 6 carbon atoms, and at least 1 carbon-carbon double bond, and up to, for example, 3 carbon-carbon double bonds in the case of a C 6 alkenyl, respectively.
  • Alkenyl groups include ethenyl, propenyl, butenyl and cyclohexenyl. As described above with respect to alkyl, the straight, branched or cyclic portion of the alkenyl group may contain double bonds and may be substituted if a substituted alkenyl group is indicated. An embodiment can be C 2 -C 12 alkenyl, C 3 -C 12 alkenyl, C 4 -C 12 alkenyl and so on. [0284]
  • alkynyl refers to a hydrocarbon radical straight or branched, containing at least 1 carbon to carbon triple bond, and up to the maximum possible number of non-aromatic carbon-carbon triple bonds may be present.
  • C 2 -C n alkynyl is defined to include groups having 1, 2...., n-1 or n carbons.
  • C 2 -C 6 alkynyl means an alkynyl radical having 2 or 3 carbon atoms, and 1 carbon-carbon triple bond, or having 4 or 5 carbon atoms, and up to 2 carbon-carbon triple bonds, or having 6 carbon atoms, and up to 3 carbon-carbon triple bonds.
  • Alkynyl groups include ethynyl, propynyl and butynyl. As described above with respect to alkyl, the straight or branched portion of the alkynyl group may contain triple bonds and may be substituted if a substituted alkynyl group is indicated.
  • An embodiment can be a C 2 - C n alkynyl.
  • An embodiment can be C 2 -C 12 alkynyl, C 3 -C 12 alkynyl, C 4 -C 12 alkynyl and so on [0285]
  • Alkylene”, “alkenylene” and “alkynylene” shall mean, respectively, a divalent alkane, alkene and alkyne radical, respectively. It is understood that an alkylene, alkenylene, and alkynylene may be straight or branched. An alkylene, alkenylene, and alkynylene may be unsubstituted or substituted.
  • heteroalkyl includes both branched and straight-chain saturated aliphatic hydrocarbon groups having the specified number of carbon atoms and at least 1 heteroatom within the chain or branch.
  • heterocycle or “heterocyclyl” as used herein is intended to mean a 5- to 10- membered nonaromatic ring containing from 1 to 4 heteroatoms selected from the group consisting of O, N and S, and includes bicyclic groups.
  • Heterocyclyl therefore includes, but is not limited to the following: imidazolyl, piperazinyl, piperidinyl, pyrrolidinyl, morpholinyl, thiomorpholinyl, tetrahydropyranyl, dihydropiperidinyl, tetrahydrothiophenyl and the like. If the heterocycle contains a nitrogen, it is understood that the corresponding N-oxides thereof are also encompassed by this definition.
  • cycloalkyl shall mean cyclic rings of alkanes of three to eight total carbon atoms, or any number within this range (i.e., cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl or cyclooctyl).
  • monocycle includes any stable polyatomic carbon ring of up to 10 atoms and may be unsubstituted or substituted. Examples of such non-aromatic monocycle elements include but are not limited to: cyclobutyl, cyclopentyl, cyclohexyl, and cycloheptyl.
  • aromatic monocycle elements examples include but are not limited to: phenyl.
  • “bicycle” includes any stable polyatomic carbon ring of up to 10 atoms that is fused to a polyatomic carbon ring of up to 10 atoms with each ring being independently unsubstituted or substituted.
  • non-aromatic bicycle elements examples include but are not limited to: decahydronaphthalene.
  • aromatic bicycle elements examples include but are not limited to: naphthalene.
  • aryl is intended to mean any stable monocyclic, bicyclic or polycyclic carbon ring of up to 10 atoms in each ring, wherein at least one ring is aromatic, and may be unsubstituted or substituted.
  • aryl elements include phenyl, p-toluenyl (4-methylphenyl), naphthyl, tetrahydro-naphthyl, indanyl, biphenyl, phenanthryl, anthryl or acenaphthyl.
  • the aryl substituent is bicyclic and one ring is non-aromatic, it is understood that attachment is via the aromatic ring.
  • polycyclic refers to unsaturated or partially unsaturated multiple fused ring structures, which may be unsubstituted or substituted.
  • arylalkyl refers to alkyl groups as described above wherein one or more bonds to hydrogen contained therein are replaced by a bond to an aryl group as described above. It is understood that an “arylalkyl” group is connected to a core molecule through a bond from the alkyl group and that the aryl group acts as a substituent on the alkyl group.
  • arylalkyl moieties include, but are not limited to, benzyl (phenylmethyl), p-trifluoromethylbenzyl (4-trifluoromethylphenylmethyl), 1-phenylethyl, 2- phenylethyl, 3-phenylpropyl, 2-phenylpropyl and the like.
  • heteroaryl represents a stable monocyclic, bicyclic or polycyclic ring of up to 10 atoms in each ring, wherein at least one ring is aromatic and contains from 1 to 4 heteroatoms selected from the group consisting of O, N and S.
  • Bicyclic aromatic heteroaryl groups include phenyl, pyridine, pyrimidine or pyridizine rings that are (a) fused to a 6-membered aromatic (unsaturated) heterocyclic ring having one nitrogen atom; (b) fused to a 5- or 6-membered aromatic (unsaturated) heterocyclic ring having two nitrogen atoms; (c) fused to a 5-membered aromatic (unsaturated) heterocyclic ring having one nitrogen atom together with either one oxygen or one sulfur atom; or (d) fused to a 5- membered aromatic (unsaturated) heterocyclic ring having one heteroatom selected from O, N or S.
  • Heteroaryl groups within the scope of this definition include but are not limited to: benzoimidazolyl, benzofuranyl, benzofurazanyl, benzopyrazolyl, benzotriazolyl, benzothiophenyl, benzoxazolyl, carbazolyl, carbolinyl, cinnolinyl, furanyl, indolinyl, indolyl, indolazinyl, indazolyl, isobenzofuranyl, isoindolyl, isoquinolyl, isothiazolyl, isoxazolyl, naphthpyridinyl, oxadiazolyl, oxazolyl, oxazoline, isoxazoline, oxetanyl, pyranyl, pyrazinyl, pyrazolyl, pyridazinyl, pyridopyridinyl, pyridazinyl, pyridyl, pyr
  • heteroaryl substituent is bicyclic and one ring is non-aromatic or contains no heteroatoms, it is understood that attachment is via the aromatic ring or via the heteroatom containing ring, respectively. If the heteroaryl contains nitrogen atoms, it is understood that the corresponding N-oxides thereof are also encompassed by this definition.
  • alkylheteroaryl refers to alkyl groups as described above wherein one or more bonds to hydrogen contained therein are replaced by a bond to an heteroaryl group as described above.
  • alkylheteroaryl is connected to a core molecule through a bond from the alkyl group and that the heteroaryl group acts as a substituent on the alkyl group.
  • alkylheteroaryl moieties include, but are not limited to, -CH 2 -(C 5 H 4 N), -CH 2 -CH 2 -(C 5 H 4 N) and the like.
  • heterocycle or “heterocyclyl” refers to a mono- or poly-cyclic ring system which can be saturated or contains one or more degrees of unsaturation and contains one or more heteroatoms.
  • Preferred heteroatoms include N, O, and/or S, including N-oxides, sulfur oxides, and dioxides.
  • the ring is three to ten-membered and is either saturated or has one or more degrees of unsaturation.
  • the heterocycle may be unsubstituted or substituted, with multiple degrees of substitution being allowed.
  • Such rings may be optionally fused to one or more of another "heterocyclic" ring(s), heteroaryl ring(s), aryl ring(s), or cycloalkyl ring(s).
  • heterocycles include, but are not limited to, tetrahydrofuran, pyran, 1,4-dioxane, 1,3-dioxane, piperidine, piperazine, pyrrolidine, morpholine, thiomorpholine, tetrahydrothiopyran, tetrahydrothiophene, 1,3-oxathiolane, and the like.
  • the alkyl, alkenyl, alkynyl, aryl, heteroaryl and heterocyclyl substituents may be substituted or unsubstituted, unless specifically defined otherwise.
  • alkyl, alkenyl, alkynyl, aryl, heterocyclyl and heteroaryl groups can be further substituted by replacing one or more hydrogen atoms with alternative non-hydrogen groups.
  • non-hydrogen groups include, but are not limited to, halo, hydroxy, mercapto, amino, carboxy, cyano and carbamoyl.
  • halogen refers to F, Cl, Br, and I.
  • substitution refers to a functional group as described above in which one or more bonds to a hydrogen atom contained therein are replaced by a bond to non-hydrogen or non-carbon atoms, provided that normal valencies are maintained and that the substitution results in a stable compound.
  • Substituted groups also include groups in which one or more bonds to a carbon(s) or hydrogen(s) atom are replaced by one or more bonds, including double or triple bonds, to a heteroatom.
  • substituent groups include the functional groups described above, and halogens (i.e., F, Cl, Br, and I); alkyl groups, such as methyl, ethyl, n-propyl, isopropryl, n-butyl, tert-butyl, and trifluoromethyl; hydroxyl; alkoxy groups, such as methoxy, ethoxy, n-propoxy, and isopropoxy; aryloxy groups, such as phenoxy; arylalkyloxy, such as benzyloxy (phenylmethoxy) and p- trifluoromethylbenzyloxy (4-trifluoromethylphenylmethoxy); heteroaryloxy groups; sulfonyl groups, such as trifluoromethanesulfonyl, methanesulfonyl, and p-toluenesulfonyl; nitro, nitrosyl; mercapto; sulfanyl groups, such as
  • the substituted compound can be independently substituted by one or more of the disclosed or claimed substituent moieties, singly or pluraly.
  • independently substituted it is meant that the (two or more) substituents can be the same or different.
  • substituents and substitution patterns on the compounds of the instant invention can be selected by one of ordinary skill in the art to provide compounds that are chemically stable and that can be readily synthesized by techniques known in the art, as well as those methods set forth below, from readily available starting materials. If a substituent is itself substituted with more than one group, it is understood that these multiple groups may be on the same carbon or on different carbons, so long as a stable structure results.
  • the compounds used in the method of the present invention may be prepared by techniques described in Vogel’s Textbook of Practical Organic Chemistry, A.I. Vogel, A.R. Tatchell, B.S. Furnis, A.J. Hannaford, P.W.G. Smith, (Prentice Hall) 5 th Edition (1996), March's Advanced Organic Chemistry: Reactions, Mechanisms, and Structure, Michael B. Smith, Jerry March, (Wiley-Interscience) 5 th Edition (2007), and references therein, which are incorporated by reference herein. However, these may not be the only means by which to synthesize or obtain the desired compounds.
  • Another aspect of the invention comprises a compound used in the method of the present invention as a pharmaceutical composition.
  • a pharmaceutical composition comprising the compound of the present invention and a pharmaceutically acceptable carrier.
  • pharmaceutically active agents include, but are not limited to, substances and compounds described in the Physicians’ Desk Reference (PDR Network, LLC; 64th edition; November 15, 2009) and “Approved Drug Products with Therapeutic Equivalence Evaluations” (U.S.
  • compositions which have pendant carboxylic acid groups may be modified in accordance with the present invention using standard esterification reactions and methods readily available and known to those having ordinary skill in the art of chemical synthesis. Where a pharmaceutically active agent does not possess a carboxylic acid group, the ordinarily skilled artisan will be able to design and incorporate a carboxylic acid group into the pharmaceutically active agent where esterification may subsequently be carried out so long as the modification does not interfere with the pharmaceutically active agent’s biological activity or effect.
  • the compounds used in the method of the present invention may be in a salt form.
  • a “salt” is a salt of the instant compounds which has been modified by making acid or base salts of the compounds.
  • the salt is pharmaceutically acceptable.
  • pharmaceutically acceptable salts include, but are not limited to, mineral or organic acid salts of basic residues such as amines; alkali or organic salts of acidic residues such as phenols.
  • the salts can be made using an organic or inorganic acid.
  • Such acid salts are chlorides, bromides, sulfates, nitrates, phosphates, sulfonates, formates, tartrates, maleates, malates, citrates, benzoates, salicylates, ascorbates, and the like.
  • Phenolate salts are the alkaline earth metal salts, sodium, potassium or lithium.
  • pharmaceutically acceptable salt in this respect, refers to the relatively non-toxic, inorganic and organic acid or base addition salts of compounds of the present invention.
  • salts can be prepared in situ during the final isolation and purification of the compounds of the invention, or by separately reacting a purified compound of the invention in its free base or free acid form with a suitable organic or inorganic acid or base, and isolating the salt thus formed.
  • Representative salts include the hydrobromide, hydrochloride, sulfate, bisulfate, phosphate, nitrate, acetate, valerate, oleate, palmitate, stearate, laurate, benzoate, lactate, phosphate, tosylate, citrate, maleate, fumarate, succinate, tartrate, napthylate, mesylate, glucoheptonate, lactobionate, and laurylsulphonate salts and the like. (See, e.g., Berge et al. (1977) "Pharmaceutical Salts", J. Pharm. Sci.66:1-19).
  • the compounds of the present invention may also form salts with basic amino acids such a lysine, arginine, etc. and with basic sugars such as N-methylglucamine, 2-amino-2-deoxyglucose, etc. and any other physiologically non-toxic basic substance.
  • “administering” an agent may be performed using any of the various methods or delivery systems well known to those skilled in the art.
  • the administering can be performed, for example, orally, parenterally, intraperitoneally, intravenously, intraarterially, transdermally, sublingually, intramuscularly, rectally, transbuccally, intranasally, liposomally, via inhalation, vaginally, intraoccularly, via local delivery, subcutaneously, intraadiposally, intraarticularly, intrathecally, into a cerebral ventricle, intraventicularly, intratumorally, into cerebral parenchyma or intraparenchchymally.
  • the compounds used in the method of the present invention may be administered in various forms, including those detailed herein.
  • the treatment with the compound may be a component of a combination therapy or an adjunct therapy, i.e.
  • a "pharmaceutically acceptable carrier” is a pharmaceutically acceptable solvent, suspending agent or vehicle, for delivering the instant compounds to the animal or human.
  • the carrier may be liquid or solid and is selected with the planned manner of administration in mind.
  • Liposomes are also a pharmaceutically acceptable carrier as are slow-release vehicles.
  • a dosage unit of the compounds used in the method of the present invention may comprise a single compound or mixtures thereof with additional antitumor agents.
  • the compounds can be administered in oral dosage forms as tablets, capsules, pills, powders, granules, elixirs, tinctures, suspensions, syrups, and emulsions.
  • the compounds may also be administered in intravenous (bolus or infusion), intraperitoneal, subcutaneous, or intramuscular form, or introduced directly, e.g. by injection, topical application, or other methods, into or topically onto a site of disease or lesion, all using dosage forms well known to those of ordinary skill in the pharmaceutical arts.
  • the compounds used in the method of the present invention can be administered in admixture with suitable pharmaceutical diluents, extenders, excipients, or in carriers such as the novel programmable sustained-release multi-compartmental nanospheres (collectively referred to herein as a pharmaceutically acceptable carrier) suitably selected with respect to the intended form of administration and as consistent with conventional pharmaceutical practices.
  • the unit will be in a form suitable for oral, nasal, rectal, topical, intravenous or direct injection or parenteral administration.
  • the compounds can be administered alone or mixed with a pharmaceutically acceptable carrier.
  • This carrier can be a solid or liquid, and the type of carrier is generally chosen based on the type of administration being used.
  • the active agent can be co-administered in the form of a tablet or capsule, liposome, as an agglomerated powder or in a liquid form.
  • suitable solid carriers include lactose, sucrose, gelatin and agar. Capsule or tablets can be easily formulated and can be made easy to swallow or chew; other solid forms include granules, and bulk powders.
  • Tablets may contain suitable binders, lubricants, diluents, disintegrating agents, coloring agents, flavoring agents, flow-inducing agents, and melting agents.
  • suitable liquid dosage forms include solutions or suspensions in water, pharmaceutically acceptable fats and oils, alcohols or other organic solvents, including esters, emulsions, syrups or elixirs, suspensions, solutions and/or suspensions reconstituted from non-effervescent granules and effervescent preparations reconstituted from effervescent granules.
  • Such liquid dosage forms may contain, for example, suitable solvents, preservatives, emulsifying agents, suspending agents, diluents, sweeteners, thickeners, and melting agents.
  • Oral dosage forms optionally contain flavorants and coloring agents.
  • Parenteral and intravenous forms may also include minerals and other materials to make them compatible with the type of injection or delivery system chosen.
  • Techniques and compositions for making dosage forms useful in the present invention are described in the following references: 7 Modern Pharmaceutics, Chapters 9 and 10 (Banker & Rhodes, Editors, 1979); Pharmaceutical Dosage Forms: Tablets (Lieberman et al., 1981); Ansel, Introduction to Pharmaceutical Dosage Forms 2nd Edition (1976); Remington's Pharmaceutical Sciences, 17th ed. (Mack Publishing Company, Easton, Pa., 1985); Advances in Pharmaceutical Sciences (David Ganderton, Trevor Jones, Eds., 1992); Advances in Pharmaceutical Sciences Vol. 7.
  • Tablets may contain suitable binders, lubricants, disintegrating agents, coloring agents, flavoring agents, flow-inducing agents, and melting agents.
  • the active drug component can be combined with an oral, non-toxic, pharmaceutically acceptable, inert carrier such as lactose, gelatin, agar, starch, sucrose, glucose, methyl cellulose, magnesium stearate, dicalcium phosphate, calcium sulfate, mannitol, sorbitol and the like.
  • Suitable binders include starch, gelatin, natural sugars such as glucose or beta-lactose, corn sweeteners, natural and synthetic gums such as acacia, tragacanth, or sodium alginate, carboxymethylcellulose, polyethylene glycol, waxes, and the like.
  • Lubricants used in these dosage forms include sodium oleate, sodium stearate, magnesium stearate, sodium benzoate, sodium acetate, sodium chloride, and the like.
  • Disintegrators include, without limitation, starch, methyl cellulose, agar, bentonite, xanthan gum, and the like.
  • the compounds used in the method of the present invention may also be administered in the form of liposome delivery systems, such as small unilamellar vesicles, large unilamellar vesicles, and multilamellar vesicles.
  • Liposomes can be formed from a variety of phospholipids such as lecithin, sphingomyelin, proteolipids, protein-encapsulated vesicles or from cholesterol, stearylamine, or phosphatidylcholines.
  • the compounds may be administered as components of tissue-targeted emulsions.
  • the compounds used in the method of the present invention may also be coupled to soluble polymers as targetable drug carriers or as a prodrug.
  • Such polymers include polyvinylpyrrolidone, pyran copolymer, polyhydroxylpropylmethacrylamide-phenol, polyhydroxyethylasparta-midephenol, or polyethyleneoxide-polylysine substituted with palmitoyl residues.
  • the compounds may be coupled to a class of biodegradable polymers useful in achieving controlled release of a drug, for example, polylactic acid, polyglycolic acid, copolymers of polylactic and polyglycolic acid, polyepsilon caprolactone, polyhydroxy butyric acid, polyorthoesters, polyacetals, polydihydropyrans, polycyanoacylates, and crosslinked or amphipathic block copolymers of hydrogels.
  • Gelatin capsules may contain the active ingredient compounds and powdered carriers, such as lactose, starch, cellulose derivatives, magnesium stearate, stearic acid, and the like. Similar diluents can be used to make compressed tablets. Both tablets and capsules can be manufactured as immediate release products or as sustained release products to provide for continuous release of medication over a period of hours. Compressed tablets can be sugar-coated or film-coated to mask any unpleasant taste and protect the tablet from the atmosphere, or enteric coated for selective disintegration in the gastrointestinal tract. [0321] For oral administration in liquid dosage form, the oral drug components are combined with any oral, non-toxic, pharmaceutically acceptable inert carrier such as ethanol, glycerol, water, and the like.
  • any oral, non-toxic, pharmaceutically acceptable inert carrier such as ethanol, glycerol, water, and the like.
  • liquid dosage forms examples include solutions or suspensions in water, pharmaceutically acceptable fats and oils, alcohols or other organic solvents, including esters, emulsions, syrups or elixirs, suspensions, solutions and/or suspensions reconstituted from non-effervescent granules and effervescent preparations reconstituted from effervescent granules.
  • Such liquid dosage forms may contain, for example, suitable solvents, preservatives, emulsifying agents, suspending agents, diluents, sweeteners, thickeners, and melting agents.
  • Liquid dosage forms for oral administration can contain coloring and flavoring to increase patient acceptance.
  • parenteral solutions In general, water, asuitable oil, saline, aqueous dextrose (glucose), and related sugar solutions and glycols such as propylene glycol or polyethylene glycols are suitable carriers for parenteral solutions.
  • Solutions for parenteral administration preferably contain a water soluble salt of the active ingredient, suitable stabilizing agents, and if necessary, buffer substances.
  • Antioxidizing agents such as sodium bisulfite, sodium sulfite, or ascorbic acid, either alone or combined, are suitable stabilizing agents.
  • citric acid and its salts and sodium EDTA are also used.
  • parenteral solutions can contain preservatives, such as benzalkonium chloride, methyl- or propyl-paraben, and chlorobutanol.
  • Suitable pharmaceutical carriers are described in Remington's Pharmaceutical Sciences, Mack Publishing Company, a standard reference text in this field.
  • the compounds used in the method of the present invention may also be administered in intranasal form via use of suitable intranasal vehicles, or via transdermal routes, using those forms of transdermal skin patches well known to those of ordinary skill in that art.
  • the dosage administration will generally be continuous rather than intermittent throughout the dosage regimen.
  • Parenteral and intravenous forms may also include minerals and other materials such as solutol and/or ethanol to make them compatible with the type of injection or delivery system chosen.
  • the compounds and compositions of the present invention can be administered in oral dosage forms as tablets, capsules, pills, powders, granules, elixirs, tinctures, suspensions, syrups, and emulsions.
  • the compounds may also be administered in intravenous (bolus or infusion), intraperitoneal, subcutaneous, or intramuscular form, or introduced directly, e.g. by topical administration, injection or other methods, to the afflicted area, such as a wound, including ulcers of the skin, all using dosage forms well known to those of ordinary skill in the pharmaceutical arts.
  • Specific examples of pharmaceutically acceptable carriers and excipients that may be used to formulate oral dosage forms of the present invention are described in U.S. Pat. No.
  • the active ingredient can be administered orally in solid dosage forms, such as capsules, tablets, powders, and chewing gum; or in liquid dosage forms, such as elixirs, syrups, and suspensions, including, but not limited to, mouthwash and toothpaste. It can also be administered parentally, in sterile liquid dosage forms.
  • Solid dosage forms, such as capsules and tablets, may be enteric-coated to prevent release of the active ingredient compounds before they reach the small intestine.
  • Materials that may be used as enteric coatings include, but are not limited to, sugars, fatty acids, proteinaceous substances such as gelatin, waxes, shellac, cellulose acetate phthalate (CAP), methyl acrylate-methacrylic acid copolymers, cellulose acetate succinate, hydroxy propyl methyl cellulose phthalate, hydroxy propyl methyl cellulose acetate succinate (hypromellose acetate succinate), polyvinyl acetate phthalate (PVAP), and methyl methacrylate- methacrylic acid copolymers.
  • CAP cellulose acetate phthalate
  • CAP methyl acrylate-methacrylic acid copolymers
  • cellulose acetate succinate methyl acrylate-methacrylic acid copolymers
  • cellulose acetate succinate hydroxy propyl methyl cellulose phthalate
  • hydroxy propyl methyl cellulose acetate succinate hyperromellose acetate succinate
  • PVAP poly
  • Itraconazole ITC, ⁇ 98% Thin-Layer Chromatography
  • RPMI 1640 supplemented with L-glutamine, 2% (w/v) of glucose and without sodium bicarbonate were purchased from Sigma-Aldrich, USA. Brain Heart Infusion (BHI) was acquired from Oxoid, Brazil.
  • BHI Brain Heart Infusion
  • Large scale synthesis of D13 Large scale synthesis (1,000 gr) of D13 (NED-59254-02) was commissioned and performed by New England Discovery Partner (NEDP) and a Material Safety Data Sheet (MSDS) was provided (see supplementary material). The compound was subjected to HPLC, LC-MS and 1H-NMR analysis (data not shown). When shipped, it was placed in dry ice.
  • Sporothrix brasiliensis ATCC 5110/MYA4823 was obtained from the American Type Culture Collection (Manassas, VA 20108 USA). All strains were cultured in BHI broth at 37°C with orbital agitation (150 rpm) for 3 days and then cryopreserved at -80°C in BHI broth with 20% glycerol. [0343] Clinical isolates were obtained as follow. Exudate from a cutaneous or mucosal lesion were collected by impression smear on a glass slide for cytological examination.
  • Yeasts were cultured in BHI broth at 37 °C/150 rpm for 3 days. Subsequently, the cells were washed in phosphate-buffered saline (PBS) and adjusted to a concentration of 5 x 10 5 yeasts/mL in RPMI 1640. D13 and ITC were diluted in RPMI ranging from 16 to 0.06 ⁇ g/mL, and 100 ⁇ L volumes were added to 96-well plates. Then, 100 ⁇ L of the yeast suspension was added to each well. Cells were incubated at 37 °C for 3 days. RPMI 1640 was used as control.
  • PBS phosphate-buffered saline
  • Yeasts (5 x 10 5 cells/mL) was added into the 96-well microplates in 100 ⁇ L volumes, after which the plates were incubated at 37 °C for 3 days.
  • the optical density (OD) was measured at 530 nm using a Biotek ELx808 microplate reader.
  • the fractional inhibitory concentration index (FICi) was calculated as (MIC combined/MIC drug A alone) + (MIC combined/MIC drug B alone).
  • the Bliss synergistic scores was calculated using the SynergyFinder tool (synergyfinder.fimm.fi/synergy) (Ianevski A, 2017).
  • a score below -10 was interpreted as an antagonistic interaction, while scores ranging from -10 to 10 indicated an additive interaction. Scores greater than 10 denoted a synergistic interaction between the two drugs.
  • SEM scanning electron microscopy
  • Claws collected from healthy cats were previously sterilized by immersion in 90% ethanol / 3h, washed 3x with PBS and transferred to tubes with 200 ⁇ L of RPMI / 1% PenStrep containing 1 x10 6 cells/mL of S. brasiliensis ATCC 5110. Cells were incubated at 37 °C for 3 days. Afterwards, claws were washed 3x with PBS in order to remove the cells not adhered to the biofilm. The biofilm formation was checked using an optical microscope (Olympus CX-31) and then 200 ⁇ L of D13 (10 ⁇ g/mL) or ITC (10 ⁇ g/mL) previously diluted in RPMI 1640 was added.
  • the cat was being already treated with ITC (100 mg/cat/day) for eight months before referral.
  • Clinical examination revealed multiple ulcerated cutaneous lesions and nodules distributed over the head, nasal region, hind and forelimbs, swelling of both nostrils, dyspnea and loss of the normal nasal planum architecture.
  • the cat was in serious overall condition and presented hyporexia. Due to poor clinical response, KI (2.5 mg/kg/day) was initiated, in combination with ITC in a lower dose (25 mg/cat/day). Oral sylimarin (30 mg/kg/day) was also prescribed. After one month, the dose of ITC was increased up to 50 mg/cat/day as the animal gained weight, while KI and sylimarin were maintained at the same dose.
  • the dose by factor method is an empirical approach and use the no observed adverse effect levels (NOAEL) of drug from preclinical toxicological studies to estimate human equivalent dose (HED).
  • NOAEL no observed adverse effect levels
  • HED human equivalent dose
  • the dose selection is based on minimum risk of toxicity, instead of choosing one with minimum pharmacologic activity in humans.
  • the existing pharmacokinetics data for another drug of the same pharmacological category may be used.
  • pharmacokinetically guided approach utilizes the drug activity instead of scaling of dose among species.
  • different methods are utilized to determine initial dose, and the data are compared and optimize to get an initial dose.
  • HED human equivalent dose
  • the K m factor for human is calculated by dividing 60 by 1.62, which is 37 (Table 1 in Nair AB, Jacob S, 2016).
  • HED (mg / kg) Animal does (mg / kg) ⁇ (Animal K m / Human K m ) Eq. (2).
  • the K m ratio values provided in Table 1 is easily obtained by dividing human K m factor by animal K m factor or vice versa.
  • the K m ratio values for rat is 6.2 and 0.162, obtained by dividing 37 (human K m factor) by 6 (animal K m factor) and vice versa, respectively.
  • HED values mg/kg
  • the NOAEL in rats is 50 mg/kg.
  • HED is calculated either by multiplying or dividing the animal dose with the K m ratio values given in Table 1 (Nair AB, Jacob S, 2016). Accordingly, divide the rat dose (50 mg/kg) by 6.2 or multiply by 0.162, the HED is –8.1 mg/kg. (Nair AB, Jacob S, 2016) [0364]
  • the k m factor varies across animal species and increases proportional to W 2/3 within a species as body weight increases. For example, the k m value in rats varies from 5.2 (100 g rat), 6 (150 g rat), and 7 (250 g rat).
  • HED conversion (mg/kg) is also based on body surface area normalization. The conversion can be made by dividing the NOAEL in appropriate species by the conversion factor. Guidelines for maximum injection volumes, by species, site location, and gauge size are summarized in Table 3 (Nair AB, Jacob S, 2016). Injection volume of parenteral formulation is calculated by following equation.
  • the concentration of the formulation is 10 mg/mL
  • its AED is 62 mg/kg
  • the rat weight is 250 g and is administered through intraperitoneal route
  • the injection volume observed here (1.55 mL) is well below the maximum injection volume (5–10 mL) for rat through intraperitoneal route and the injection site is lower left quadrant.
  • Results [0373] Enrollment.
  • the present inventions provide a pilot clinical study. The inventors did not inflict sporotrichosis to cats.
  • Cats affected with sporotrichosis were taken by their owner to the Laboratory of Clinical Research on Dermatozoonoses in Domestic Animals at Oswaldo Cruz Foundation (Fiocruz), in Rio de Janeiro, Brazil, to seek diagnosis and treatment.
  • cats were treated using the standard of care (SOC). They mainly received itraconazole (ITC) alone or in combination with potassium iodide (KI) (Table 1).
  • ITC itraconazole
  • KI potassium iodide
  • One cat (Case 2) also received terbinafine (TRB). If the cat did not respond to the SOC (see below), a possibility to add D13 was proposed to the owner of the cat as “compassionate drug use”. If the owner agreed, D13 was added to the therapeutic protocol.
  • Table 1 Part A. Clinical information and summary table of the 10 cases of sporotrichosis treated with the experimental drug acylhydrazone D13 in various combinations with the standard of care. C ase Sex Age W (Kg) Con Lesions Resp signs is
  • swab samples were collected and placed onto Sabouraud-dextrose agar. Plates were incubated for up to 5 days at room temperature ( ⁇ 25C), fungal growth was monitored and fungal identification was performed by standard methods. Fungal isolates were tested for in vitro susceptibility against ITC and D13. From the 10 cats, only 8 clinical isolates were obtained. Two isolates (from Case 4 and from Case 10) were highly contaminated by bacteria and were lost. [0376] D13 displayed high efficacy in inhibiting the growth of all tested isolates (Table 2), with MIC 50 ranging from 0.12 ⁇ g/ml to 4 ⁇ g/ml.
  • D13 is effective against S. brasiliensis biofilm. Recognizing the crucial role of biofilm as an important reservoir of fungal cells, which become more resistant to antifungals, the effect of D13 on biofilm formation was examined by Sporothrix on cat's claw. This is particularly important because this fungus is frequently transmitted from cats to humans by scratches. If Sporothrix forms biofilm in the cat’s claw its potential for transmission is significantly increased.
  • Feline sporotrichosis a case series of itraconazole-resistant Sporothrix brasiliensis infection. Braz J Microbiol 52:163-171. 13. Gremiao IDF, Miranda LHM, Pereira-Oliveira GR, Menezes RC, Machado ACS, Rodrigues AM, Pereira SA.2022. Advances and challenges in the management of feline sporotrichosis. Rev Iberoam Micol 39:61-67. 14. Silva FS, Cunha SCS, Moraes VA, Leite JS, Ferreira AMR.2022. Refractory feline sporotrichosis: a comparative analysis on the clinical, histopathological, and cytopathological aspects.
  • Mor V Rella A, Farnoud AM, Singh A, Munshi M, Bryan A, Naseem S, Konopka JB, Ojima I, Bullesbach E, Ashbaugh A, Linke MJ, Cushion M, Collins M, Ananthula HK, Sallans L, Desai PB, Wiederhold NP, Fothergill AW, Kirkpatrick WR, Patterson T, Wong LH, Sinha S, Giaever G, Nislow C, Flaherty P, Pan X, Cesar GV, de Melo Tavares P, Frases S, Miranda K, Rodrigues ML, Luberto C, Nimrichter L, Del Poeta M.2015.

Landscapes

  • Health & Medical Sciences (AREA)
  • Veterinary Medicine (AREA)
  • Chemical & Material Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Epidemiology (AREA)
  • Communicable Diseases (AREA)
  • Oncology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Organic Chemistry (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

The present invention provides a combination comprising an amount of itraconazole (ITC) and an amount of a compound, or a pharmaceutically acceptable salt thereof, wherein the compound has the following structure:

Description

COMBINATION THERAPY OF ITRACONAZOLE AND ACYLHYDRAZONE-BASED ANTIFUNGAL AGENTS FOR THE TREATMENT OF SPOROTRICHOSIS IN CATS [0001] Throughout this application, various publications are referenced, including referenced in parenthesis. The disclosures of all publications mentioned in this application in their entireties are hereby incorporated by reference into this application in order to provide additional description of the art to which this invention pertains and of the features in the art which can be employed with this invention. CROSS-REFERENCE TO RELATED APPLICATIONS [0002] This application claims the benefit of U.S. Provisional Application No. 63/570,884, filed March 28, 2024, and U.S. Provisional Application No. 63/596,461, filed November 6, 2023, the contents of which are hereby incorporated by reference. STATEMENT REGARDING FEDERALLY SPONSORED RESEARCH OR DEVELOPMENT [0003] This invention was made with government support under AI116420 awarded by National Institutes of Health. The government has certain rights in the invention. BACKGROUND OF THE INVENTION [0004] Acylhydrazone (AH) derivatives represent a novel category of antifungal medications that exhibit potent activity against Sporothrix species, both in vitro and in a murine model of sporotrichosis. The present invention demonstrated the antifungal efficacy of the AH derivative D13 [4-bromo-N′-(3,5- dibromo-2-hydroxybenzylidene)-benzohydrazide] against both planktonic cells and biofilms formed by Sporothrix brasiliensis. Br 3). In a clinical study, the eff
Figure imgf000003_0001
ect of D13 was then tested in combination with itraconazole (ITC), with or without potassium iodide (KI), in 10 cats with sporotrichosis refractory to the treatment of standard of care with ITC. Improvement or total clinical cure was achieved in five cases after 12 weeks of treatment. Minimal abnormal laboratory findings, e.g. elevation of alanine aminotransferase (ALT), were observed in four cats during the combination treatment and returned to normal level within a week after the treatment was ended. Although highly encouraging, a larger and randomized controlled study is required to evaluate the effectiveness and the safety of this new and exciting drug combination using ITC and D13 for the treatment of feline sporotrichosis. [0005] This invention provides a first veterinary clinical study of an acylhydrazone antifungal (D13) combined with itraconazole against a dimorphic fungal infection, sporotrichosis, which is highly endemic in South America in animals and humans. Overall, the results show that the combination treatment was efficacious in ~50% of the infected animals. In addition, D13 was well tolerated during the course of the study. Thus, these yesults warrant the continuation of the research and development of this new class of antifungals.
BRIEF SUMMARY OF THE INVENTION [0006] The present invention provides a method of treating fungal infection in a subject, wherein the method comprises administering to the subject a combination comprising an amount of itraconazole (ITC) and an amount of a compound, or a pharmaceutically acceptable salt thereof, wherein the compound has the following structure: ,
Figure imgf000005_0001
R1 is -H, alkyl, alkenyl, or alkynyl; R2 is -H, alkyl, alkenyl, or alkynyl; R9, R10, R11, and R12 are each independently, H, halogen, -CN, -CF3, -OCF3, -NO2, alkyl, alkenyl, alkynyl, aryl, heteroaryl, cycloalkyl, heterocycloalkyl, -OH, -OAc, -OR13, -COR13, -SH, -SR13, - SO2R13, -SO2NR14R15, -NH2, -NHR13, -NR14R15, -NHCOR12, or -CONR14R15; or R9 and R10 are each independently -H, halogen, C1-C6 alkyl, -OH, -O-(C1-C6 alkyl), -CHF2, -CF3, -OCHF2 or -OCF3, and R11 and R12 combine to form a fused aryl or fused heteroaryl, which are each unsubstituted or substituted, or R9 and R12 are each independently -H, halogen, C1-C6 alkyl, -OH, -O-(C1-C6 alkyl), -CHF2, -CF3, -OCHF2 or -OCF3, and R10 and R11 combine to form a fused aryl or fused heteroaryl, which are each unsubstituted or substitute, or R11 and R12 are each independently -H, halogen, C1-C6 alkyl, -OH, -O-(C1-C6 alkyl), -CHF2, -CF3, -OCHF2 or -OCF3, and R3 and R4 combine to form a fused aryl or fused heteroaryl, which are each unsubstituted or substituted; wherein each occurrence of R13 is independently alkyl, alkenyl, alkynyl, aryl, or heteroaryl; wherein each occurrence of R14 and R15 is independently –H, alkyl, alkenyl, alkynyl, aryl, or heteroaryl; wherein when R14 is methyl, R15 is not methyl; and wherein at least one of R9, R10, R11, and R12 is not H; so as to thereby treat the subject against fungal infection. [0007] The present invention provides a combination comprising an amount of itraconazole (ITC) and an amount of a compound, or a pharmaceutically acceptable salt thereof, wherein the compound has the following structure: ,
Figure imgf000006_0001
R1 is -H, alkyl, alkenyl, or alkynyl; R2 is -H, alkyl, alkenyl, or alkynyl; R9, R10, R11, and R12 are each independently, H, halogen, -CN, -CF3, -OCF3, -NO2, alkyl, alkenyl, alkynyl, aryl, heteroaryl, cycloalkyl, heterocycloalkyl, -OH, -OAc, -OR13, -COR13, -SH, -SR13, - SO2R13, -SO2NR14R15, -NH2, -NHR13, -NR14R15, -NHCOR12, or -CONR14R15; or R9 and R10 are each independently -H, halogen, C1-C6 alkyl, -OH, -O-(C1-C6 alkyl), -CHF2, -CF3, -OCHF2 or -OCF3, and R11 and R12 combine to form a fused aryl or fused heteroaryl, which are each unsubstituted or substituted, or R9 and R12 are each independently -H, halogen, C1-C6 alkyl, -OH, -O-(C1-C6 alkyl), -CHF2, -CF3, -OCHF2 or -OCF3, and R10 and R11 combine to form a fused aryl or fused heteroaryl, which are each unsubstituted or substitute, or R11 and R12 are each independently -H, halogen, C1-C6 alkyl, -OH, -O-(C1-C6 alkyl), -CHF2, -CF3, -OCHF2 or -OCF3, and R3 and R4 combine to form a fused aryl or fused heteroaryl, which are each unsubstituted or substituted; wherein each occurrence of R13 is independently alkyl, alkenyl, alkynyl, aryl, or heteroaryl; wherein each occurrence of R14 and R15 is independently –H, alkyl, alkenyl, alkynyl, aryl, or heteroaryl; wherein when R14 is methyl, R15 is not methyl; and wherein at least one of R9, R10, R11, and R12 is not H; for use in treating a subject afflicted with a fungal infection by administering the combination to the subject. [0008] The present invention provides a pharmaceutical composition in unit dosage form, useful in treating a subject afflicted with fungal infection, which comprises: (a) an amount of ITC; (b) an amount of the compound described herein, or a pharmaceutically acceptable salt thereof; and (c) optionally an amount of KI; wherein the respective amounts of said compound, or a pharmaceutically acceptable salt thereof, said ITC and said KI, if present, in said composition are effective, upon concomitant administration to said subject of one or more of said unit dosage forms of said composition, to treat the subject. [0009] The present invention provides a therapeutic package for dispensing to, or for use in dispensing to, a subject afflicted with fungal infection, which comprises: (a) one or more unit doses, each such unit dose comprising: i) an amount of ITC; ii) an amount of the compound described herein, or a pharmaceutically acceptable salt thereof; and iii) optionally an amount of KI; wherein the respective amounts of said compound, or a pharmaceutically acceptable salt thereof, said ITC and said KI, if present, in said unit doses are effective, upon concomitant administration to said subject, to treat the subject, and (b) a finished pharmaceutical container therefor, said container containing said unit dose or unit doses, said container further containing or comprising labeling directing the use of said package in the treatment of said subject.
BRIEF DESCRIPTION OF THE DRAWINGS [0010] Figure 1. Effect of D13 on Sporothrix Biofilm. Sporothrix yeasts were cultured in RPMI 1640 medium together with cat's claw fragments and incubated at 37°C for 3 days. Following this, the claws were washed 3x and subsequently treated with 10 µg/mL of D13 or ITC for another 3 days before being prepared for SEM analysis. In the biofilm control groups, both the ATCC strain of S. brasiliensis and the clinically isolated (C.I 17522) strain displayed abundant yeasts and hyphae with a consistently uniform and intact surface. In contrast, cells from both strains treated with either D13 or ITC exhibited slight growth and irregularities on their surfaces. [0011] Figure 2. Fungal load of Sporothrix biofilm on cat claw fragments. Mature Sporothrix biofilms grown for 3 days were treated with D13 or ITC at 10 µg/ml. After 3 days of incubation, the claws were washed and suspended in RMPI/PenStrep 1%. Claws were sonicated and vortexed to drop the cells from the biofilm. Aliquots were placed on BHI plates and incubated at 37°C for up to 3 days. Floating bars represent the mean, min to max value of three independent experiments. Statistical analysis was performed by One-way ANOVA and Sidak’s multiple comparison test, **, p < 0,0028; **** p < 0,0001. [0012] Figure 3. Case 1. The cat presented no improvement of the cutaneous lesions on the head and on the thoracic limbs upon treatment with ITC and KI. After adding D13, the cat significantly improved and clinical cure was achieved after 8 months of treatment with ITC, KI and D13 combination. [0013] Figure 4. Case 2. The cat presented no improvement of the cutaneous and nasal mucosal lesions upon treatment with ITC, TRB and KI. After adding D13, the cat significantly improved and clinical cure was achieved after 12 weeks of treatment with ITC, TRB, KI and D13 combination. [0014] Figure 5. Case 3. The cat presented no improvement of the cutaneous and nasal mucosal lesions upon treatment with ITC. After adding D13, the cat significantly improved and clinical cure was achieved after 12 weeks of treatment with ITC and D13 combination. [0015] Figure 6. Case 4. The cat presented no improvement of the cutaneous lesions in the cephalic region and left cervical region upon treatment with ITC. After adding D13, the cat significantly improved and clinical cure was achieved after 12 weeks of treatment with ITC and D13 combination. [0016] Figure 7. Case 5. The cat presented no improvement of the tumor-like lesion and of an ulcerated lesion on the nasal region upon treatment with ITC. After adding D13, the cat significantly improved even though clinical cure was not achieved after 4 weeks of treatment with ITC and D13 combination. DETAILED DESCRIPTION OF THE INVENTION [0017] Sporotrichosis is a mycotic disease that affects humans and animals and it is caused by pathogenic species of the genus Sporothrix, such as Sporothrix schenckii and Sporothrix brasiliensis. It is the most common subcutaneous mycosis(Poester VR, 2018). S. brasiliensis and S. schenckii has been described as the main worldwide causal agents of the mycosis in cats (Della Terra PP, 2017). However, in South America, and particularly in Brazil, S. brasiliensis is the most prevalent species (de Carvalho JA, 2021 and Boechat JS 2018), and in certain Asian countries, S. schenckii is the most common species ( Han HS, 2021). [0018] The cat is considered the most susceptible animal to Sporothrix infection, presenting various clinical forms, such as single skin lesion, multiple skin lesions, ulcers, or/and disseminated systemic infection. These clinical manifestations are often not responsive to treatment (Gremiao ID, 2015, Miranda LHM, 2016, de Souza EW, 2018, and Schubach TM, 2001). The treatment of sporotrichosis in cats is an important measure of disease control, not only to reduce the animal burden but also to diminish or/and prevent the transmission from cats to humans (de Miranda LHM, 2018). However, the treatment is often protracted for long time (weeks or months), and because of this long treatment the cat owner compliance is low. In addition, the fungus can also become refractory to the treatment. As a result, cats do not respond well to the standard of care, resulting in therapeutic failure (da Rocha , 2018, Nakasu CCT, 2021, Gremiao IDF, 2022 and Silva FS, 2022 ). [0019] Few effective antifungal agents are available for treating feline sporotrichosis, and most of them are often used as monotherapy regimens (Poester VR, 2022). The standard of care (SOC) includes itraconazole (ITC) alone or associated with potassium iodide (KI), and this regime is the most common therapeutic regimen in Brazil (Gremiao IDF, 2021 and Xavier MO, 2023 ). Ketoconazole, sodium iodide, fluconazole, amphotericin B (AMB), terbinafine, posaconazole, local heat therapy, cryosurgery and surgical removal of the lesions have also been described for treating sporotrichosis in cats, but their efficacy varies greatly (16). A recent study shows for the first time that Nikkomicin Z could be an option for the treatment of sporotrichosis caused by S. brasiliensis (Poester VR, 2023), although this treatment regime will be quite expensive. [0020] ITC is a triazole with a broad spectrum of antifungal activity that blocks the synthesis of ergosterol (major sterol component of fungal plasma membranes) through the inhibition of the fungal cytochrome P450-dependent enzyme lanosterol 14-a-demethylase (Como JA, 1994). However, the decrease of ergosterol synthesis leads to a fungal growth arrest and not to cell death. As such, ITC is fungistatic and therapeutic failure with ITC is common (Gremiao IDF, 2022). Thus, the testing of new and fungicidal antifungal agents (alone or in combination with ITC) is warranted (Johnson MD, 2010). [0021] The family of acylhydrazones (AH) are a new class of antifungal agent molecules, which target the vesicular transport and cell cycle progression of fungi and indirectly impact glucosylceramide (GlcCer) synthesis (Mor V, Rella A, 2015). As a result, toxic sphingolipids accumulate, such as sphingosine, leading to mitochondria and vesicular damage (Mor V, Rella A, 2015). Thus, the acylhydrazone compounds are highly fungicidal and able to kill fungi within few hours (Lazzarini C, 2018). An acylhydrazone known as D13 [4-bromo-N′-(3,5-dibromo-2-hydroxybenzylidene)-benzohydrazide] (ChemBridge ID# 5475098) showed strong antifungal activity with very low toxicity in mammalian cells (Lazzarini C, 2018). Using a checkerboard assay, D13 improved the efficacy of commercially available drugs against various fungi. Against S. brasiliensis, D13 promoted yeast disruption and ultrastructural changes (Artunduaga Bonilla JJ, Honorato L, 2021). Pharmacokinetic studies of D13 were performed in a murine model and were very promising with high oral availability (Lazzarini C, 2018). Importantly, D13 exhibited strong activity against different strains of Sporothrix in vitro when used alone or in combination with ITC (Artunduaga Bonilla JJ, Honorato L, 2021). The D13 antifungal effectiveness was further displayed in a murine model of sporotrichosis (Artunduaga Bonilla JJ, Honorato L, 2021). [0022] This invention performed a preliminary clinical study using D13 in cats affected by sporotrichosis. The study was only approved as a “compassionate drug use”, meaning D13 could only be administered after the SOC failed. As such, it was not possible to have a control group in which D13 was not administered. Out of ten cats that were enrolled, four cats were clinically cured and one significantly improved. Thus, the results are promising and encouraging the design of a larger clinical study using D13 as a primary therapy with the inclusion of a control group. [0023] The present invention provides a method of treating fungal infection in a subject, wherein the method comprises administering to the subject a combination comprising an amount of itraconazole (ITC) and an amount of a compound, or a pharmaceutically acceptable salt thereof, wherein the compound has the following structure: ,
Figure imgf000010_0001
R1 is -H, alkyl, alkenyl, or alkynyl; R2 is -H, alkyl, alkenyl, or alkynyl; R9, R10, R11, and R12 are each independently, H, halogen, -CN, -CF3, -OCF3, -NO2, alkyl, alkenyl, alkynyl, aryl, heteroaryl, cycloalkyl, heterocycloalkyl, -OH, -OAc, -OR13, -COR13, -SH, -SR13, - SO2R13, -SO2NR14R15, -NH2, -NHR13, -NR14R15, -NHCOR12, or -CONR14R15; or R9 and R10 are each independently -H, halogen, C1-C6 alkyl, -OH, -O-(C1-C6 alkyl), -CHF2, -CF3, -OCHF2 or -OCF3, and R11 and R12 combine to form a fused aryl or fused heteroaryl, which are each unsubstituted or substituted, or R9 and R12 are each independently -H, halogen, C1-C6 alkyl, -OH, -O-(C1-C6 alkyl), -CHF2, -CF3, -OCHF2 or -OCF3, and R10 and R11 combine to form a fused aryl or fused heteroaryl, which are each unsubstituted or substitute, or R11 and R12 are each independently -H, halogen, C1-C6 alkyl, -OH, -O-(C1-C6 alkyl), -CHF2, -CF3, -OCHF2 or -OCF3, and R3 and R4 combine to form a fused aryl or fused heteroaryl, which are each unsubstituted or substituted; wherein each occurrence of R13 is independently alkyl, alkenyl, alkynyl, aryl, or heteroaryl; wherein each occurrence of R14 and R15 is independently –H, alkyl, alkenyl, alkynyl, aryl, or heteroaryl; wherein when R14 is methyl, R15 is not methyl; and wherein at least one of R9, R10, R11, and R12 is not H; so as to thereby treat the subject against fungal infection. [0024] In some embodiments, when R9 and R11 are each –H and R10 and R12 are each –Br, then A is other than para-bromophenyl, meta-bromophenyl, ortho-tolyl or 3-quinolinyl. [0025] In some embodiments, when R9, R11 and R12 are each –H and R10 is –Br, then A is other than 3,5-dibromo-ortho-hydroxyphenyl, para-bromophenyl, meta-bromophenyl or ortho-tolyl. [0026] In some embodiments, the aryl or heteroaryl is substituted with halogen, C1-C6 alkyl, -OH, -O- (C1-C6 alkyl), -CHF2,-CF3, -OCHF2 or -OCF3. [0027] In some embodiments, the fused aryl or fused heteroaryl is substituted with halogen, C1-C6 alkyl, -OH, -O-(C1-C6 alkyl), -CHF2, -CF3, -OCHF2 or -OCF3. [0028] In some embodiments, the compound having the following structure: , wherein
Figure imgf000012_0001
ndently, -H, halogen, C1-C6 alkyl, -OH, -O-(C1-C6 alkyl), - CHF2, -CF3, -OCHF2, -OCF3, -CN, -CH2OCH3, -N(CH3)2, -CH2F, -N3 or -CCH. [0029] In some embodiments, R1 and R2 are each H. [0030] In some embodiments, R3, R4, R5, R6 and R7 are each independently -H, halogen, C1-C6 alkyl, -OH, -O-(C1-C6 alkyl), -CHF2, -CF3, -OCHF2 or -OCF3. [0031] In some embodiments, R3, R4, R5, and R6 are each independently halogen, -O-(C1-C6 alkyl), - OCF3 or -CF3. [0032] In some embodiments, R3, R4, R5, and R6 are each independently halogen or -O-(C1-C6 alkyl). [0033] In some embodiments, R3, R4, R5, and R6 are each independently –Cl, -Br, -F, -O-(C1-C6 alkyl), -OCF3 or -CF3. [0034] In some embodiments, R3, R4, R5, and R6 are each independently –Cl, -Br, or -O-(C1-C6 alkyl). [0035] In some embodiments, R9, R10, R11, and R12 are each independently H, halogen, C1-C6 alkyl, - OH, -O-(C1-C6 alkyl), -CHF2, -CF3, -OCHF2 or -OCF3. [0036] In some embodiments, R9, R10, R11, and R12 are each independently halogen, -O-(C1-C6 alkyl), -OCF3 or -CF3. [0037] In some embodiments, R9, R10, R11, and R12 are each independently halogen or -O-(C1-C6 alkyl). [0038] In some embodiments, R9, R10, R11, and R12 are each independently –Cl, -Br, -F, -O-(C1-C6 alkyl), -OCF3 or -CF3. [0039] In some embodiments, R9, R10, R11, and R12 are each independently –Cl, -Br, or -O-(C1-C6 alkyl). [0040] In some embodiments, at least one of R3, R4, R5, R6 and R7 is not H. [0041] In some embodiments, at least two of R3, R4, R5, R6 and R7 are not H. [0042] In some embodiments, at least one of R9, R10, R11, and R12 is not H. [0043] In some embodiments, at least two of R9, R10, R11, and R12 are not H. [0044] In some embodiments, R3, R4, R6, R7, R10 and R12 are H. [0045] In some embodiments, R3, R4, R6, R9, R10 and R12 are H. [0046] In some embodiments, R5, R9, and R10 are halogen; preferably R5, R9, and R10 are Br. [0047] In some embodiments, R5, R7, R11 are halogen; preferably R5, R9, and R10 are Br. [0048] In some embodiments, R1 is -H; R2 is -H; R3, R4, R5, R6 and R7 are each, independently, H, halogen, C1-C6 alkyl, -OH; and R9, R10, R11, and R12 are each independently H, halogen, C1-C6 alkyl, -OH. [0049] In some embodiments, R1 is -H; R2 is -H; R3, R4, R5, R6 and R7 are each, independently, H or halogen; and R9, R10, R11, and R12 are each independently H or halogen. [0050] In some embodiments, R1 is -H; R2 is -H; R3, R4, R5, R6 and R7 are each, independently H or Br; and R9, R10, R11, and R12 are each independently H or Br. OH OH OH Br , ,
Figure imgf000013_0001
, , , or
Figure imgf000014_0001
[ ] n some em o mens, e compoun as e o owng srucure:
, , , , , ,
Figure imgf000015_0001
,, ,
Figure imgf000016_0001
, , ,
Figure imgf000016_0002
, , , , , , ,
Figure imgf000017_0001
, , , , , , ,
Figure imgf000018_0001
, , , , ,
Figure imgf000019_0001
, , r or
Figure imgf000020_0001
[0058] In some embodiments, the compound has the following structure: Br Br .
Figure imgf000020_0002
[0059] In some embodiments, the compound has the following structure: Br r .
Figure imgf000021_0001
d has the following structure: Br O Br .
Figure imgf000021_0002
, e compound has the following structure:
Figure imgf000021_0003
R11 is -H, -Br, or -NR14R15, R10, R12, and R12 are each independently -H, -Br, -OH, -OR13, or -NR14R15; preferably, R10, R12, and R12 are each independently -H, -Br, -OH, -OCH3, or -N(CH3)2. [0062] In some embodiments, the compound has the following structure: R5 R5 R5 R5 R R R R R R R R6 R7 ,
Figure imgf000021_0004
R5 R5 R4 R6 R4 R6 7 . [0063] pound has the following structure:
Figure imgf000022_0003
Br Br CH3 , [
Figure imgf000022_0001
] n some em o ments, t e compoun avng t e o owng structure: R10 R R11 12 ,
Figure imgf000022_0002
wherein R3, R4, R5, R6 and R7 are each independently, -H, halogen, C1-C6 alkyl, -OH, -O-(C1-C6 alkyl), - CHF2, -CF3, -OCHF2, -OCF3, -CN, -CH2OCH3, -N(CH3)2, -CH2F, -N3 or -CCH. [0065] In some embodiments, R3, R4, R5, R6, and R7 are each independently H, halogen, -CN, alkyl, alkenyl, alkynyl, aryl, heteroaryl, heterocycle, -OCF3, -OH, -OAc, -OR13, -COR13, -CH2OR13. [0066] In some embodiments, R3, R4, R5, R6, and R7 are each independently, H, halogen, -OCF3, heterocycle , or -CH2OR13. [0067] In some embodiments, R3, R4, R5, R6, and R7 are each independently, H or halogen. [0068] In some embodiments, a) R3 is halogen, R4, R5, R6, and R7 are H; b) R4 is halogen, R3, R5, R6, and R7 are H; c) R5 is halogen, R3, R4, R6, and R7 are H; d) R6 is halogen, R3, R4, R5, and R7 are H; e) R7 is halogen, R3, R4, R5, and R6 are H; f) R3 and R4 are halogen, R5, R6, R7 are H; g) R3 and R5 are halogen, R4, R6, R7 are H; h) R3 and R6 are halogen, R4, R5, R7 are H; i) R3 and R7 are halogen, R4, R5, R6 are H; j) R4 and R5 are halogen, R3, R6, R7 are H; k) R4 and R6 are halogen, R3, R5, R7 are H; l) R4 and R7 are halogen, R3, R5, R6 are H; m) R5 and R6 are halogen, R3, R4, R7 are H; n) R5 and R7 are halogen, R3, R4, R6 are H; or o) R6 and R7 are halogen, R3, R4, R5 are H. [0069] In some embodiments, the heterocycle is six-membered heterocycle. [0070] In some embodiments, the heterocycle is five-membered heterocycle. [0071] In some embodiments, the heterocycle is four-membered heterocycle. [0072] In some embodiments, the heterocycle is three-membered heterocycle. [0073] In some embodiments, R3 is OH, R4, R5, R6 and R7 are each independently H or halogen. [0074] In some embodiments, a) R3 is OH, R4, R5, and R6 are H, R7 is halogen; b) R3 is OH, R5, R6, and R7 are H, R4 is halogen; c) R3 is OH, R4, R6, and R7 are H, R5 is halogen; d) R3 is OH, R4, R5, and R7 are H, R6 is halogen; e) R3 is OH, R4 and R5 are halogen, R6 and R7 are H; f) R3 is OH, R4 and R6 are halogen, R5 and R7 are H; g) R3 is OH, R4 and R7 are halogen, R5 and R6 are H; h) R3 is OH, R5 and R6 are halogen, R4 and R7 are H; i) R3 is OH, R5 and R7 are halogen, R4 and R6 are H; or j) R3 is OH, R6 and R7 are halogen, R4 and R5 are H. [0075] In some embodiments, the halogen is F, Cl, Br, and I. [0076] In some embodiments, the halogen is Br. [0077] In some embodiments, R7 is OH, R3, R4, R5 and R6 are each independently H or halogen. [0078] In some embodiments, a) R7 is OH, R4 and R5 are halogen, R3 and R6 are H; b) R7 is OH, R4 and R6 are halogen, R3 and R5 are H; c) R7 is OH, R3 and R4 are halogen, R5 and R6 are H; d) R7 is OH, R3 and R5 are halogen, R4 and R6 are H; e) R7 is OH, R3 and R6 are halogen, R4 and R5 are H; f) R7 is OH, R5 and R6 are halogen, R3 and R4 are H; g) R7 is OH, R3, R4, and R5 are H, R6 is halogen; h) R7 is OH, R3, R4, and R6 are H, R5 is halogen; i) R7 is OH, R4, R5, and R6 are H, R3 is halogen; or j) R7 is OH, R3, R5, and R6 are H, R4 is halogen; [0079] In some embodiments, the halogen is F, Cl, Br, and I. [0080] In some embodiments, the halogen is Br. [0081] In some embodiments, R3, R4, R5, R6, R7 are each independently, H or CH2OR13. [0082] In some embodiments, a) R3 is CH2OR13, R4, R5, R6, and R7 are H; b) R4 is CH2OR13, R3, R5, R6, and R7 are H; c) R5 is CH2OR13, R3, R4, R6, and R7 are H; d) R6 is CH2OR13, R3, R4, R5, and R7 are H; e) R7 is CH2OR13, R3, R4, R5, and R6 are H; f) R3 and R4 are CH2OR13, R5, R6, R7 are H; g) R3 and R5 are CH2OR13, R4, R6, R7 are H; h) R3 and R6 are CH2OR13, R4, R5, R7 are H; i) R3 and R7 are CH2OR13, R4, R5, R6 are H; j) R4 and R5 are CH2OR13, R3, R6, R7 are H; k) R4 and R6 are CH2OR13, R3, R5, R7 are H; l) R4 and R7 are CH2OR13, R3, R5, R6 are H; m) R5 and R6 are CH2OR13, R3, R4, R7 are H; n) R5 and R7 are CH2OR13, R3, R4, R6 are H; or o) R6 and R7 are CH2OR13, R3, R4, R5 are H. [0083] In some embodiments, R5 is CH2OR13 and R13 is branched or unbranched alkyl. [0084] In some embodiments, alkyl is C1-C6 alkyl, branched or unbranched. [0085] In some embodiments, alkyl is methyl, ethyl, propyl, or isopropyl; [0086] In some embodiments, R3, R4, R5, R6, and R7 are each independently, H, halogen or -OCF3. [0087] In some embodiments, a) R3 is halogen, R4, R5, and R6 are H, R7 is -OCF3; b) R3 is halogen, R5, R6, and R7 are H, R4 is -OCF3; c) R3 is halogen, R4, R6, and R7 are H, R5 is -OCF3; d) R3 is halogen, R4, R5, and R7 are H, R6 is -OCF3; e) R4 is halogen, R3, R5, and R6 are H, R7 is -OCF3; f) R4 is halogen, R3, R5, and R7 are H, R6 is -OCF3; g) R4 is halogen, R3, R6, and R7 are H, R5 is -OCF3; h) R4 is halogen, R5, R6, and R7 are H, R3 is -OCF3; i) R6 is halogen, R3, R4, and R5 are H, R7 is -OCF3; j) R6 is halogen, R3, R4, and R7 are H, R5 is -OCF3; k) R6 is halogen, R5, R6, and R7 are H, R3 is -OCF3; l) R6 is halogen, R3, R5, and R7 are H, R4 is -OCF3; m) R7 is halogen, R3, R4, and R5 are H, R6 is -OCF3; n) R7 is halogen, R3, R4, and R6 are H, R5 is -OCF3; o) R7 is halogen, R4, R5, and R6 are H, R3 is -OCF3; or p) R7 is halogen, R3, R5, and R6 are H, R4 is -OCF3. [0088] In some embodiments, the halogen is F, Cl, Br, and I. [0089] In some embodiments, the halogen is F. [0090] In some embodiments, R3, R4, R5, R6, and R7 are each H or heterocycle. [0091] In some embodiments, a) R3 is heterocycle, R4, R5, R6, and R7 are H; b) R4 is heterocycle, R3, R5, R6, and R7 are H; c) R5 is heterocycle, R3, R4, R6, and R7 are H; d) R6 is heterocycle, R3, R4, R5, and R7 are H; or e) R7 is heterocycle, R3, R4, R5, and R6 are H. [0092] In some embodiments, heterocycle is six-membered heterocycle. [0093] In some embodiments, heterocycle is five-membered heterocycle. [0094] In some embodiments, heterocycle is four-membered heterocycle. [0095] In some embodiments, heterocycle is three-membered heterocycle. [0096] In some embodiments, the three-membered heterocycle is aziridine, 2H-azirine, oxirane, thiirane or azirine. [0097] In some embodiments, the azirine is 3-methyl-3-(trifluoromethyl)-3H-diazirine. [0098] In some embodiments, R4, R5, and R6 are each H or heterocycle, R7 is H. [0099] In some embodiments, a) R4 is heterocycle, R3, R5, R6, and R7 are H; b) R5 is heterocycle, R3, R4, R6, and R7 are H; or c) R6 is heterocycle, R3, R4, R5, and R7 are H. [0100] In some embodiments, heterocycle is six-membered heterocycle. [0101] In some embodiments, heterocycle is five-membered heterocycle. [0102] In some embodiments, heterocycle is four-membered heterocycle. [0103] In some embodiments, heterocycle is three-membered heterocycle. [0104] In some embodiments, three-membered heterocycle is aziridine, 2H-azirine, oxirane, thiirane or azirine. [0105] In some embodiments, the azirine is 3-methyl-3-(trifluoromethyl)-3H-diazirine. [0106] In some embodiments, the compound has the following structure: , g structure:
Figure imgf000027_0001
, ,
Figure imgf000027_0002
. [0107]
Figure imgf000027_0003
, ,
Figure imgf000027_0004
g structure: ,
Figure imgf000027_0005
. [0108] lowing structure:
Figure imgf000028_0001
, g structure:
Figure imgf000028_0002
, or
Figure imgf000028_0003
. [0109]
Figure imgf000028_0004
In some embodiments, the compound has the following structure: ,
Figure imgf000028_0005
wherein ring A has the following structure: ,
Figure imgf000028_0006
. [0110] ompound has the following structure:
Figure imgf000029_0001
, or , ,
Figure imgf000029_0002
, , , , , , ,
Figure imgf000030_0001
HO O or [0112]
Figure imgf000031_0001
, [0113]
Figure imgf000031_0002
In some embodiments, the compound has the following structure: , [0114]
Figure imgf000032_0001
,
Figure imgf000032_0002
, , und, is administered to the subject in a capsule. [0116] In some embodiments, the amount of ITC and the amount of the compound, or pharmaceutically acceptable salt of the compound, when taken together are effective to achieve an additive or a greater than additive therapeutic result in treating the subject. [0117] In some embodiments, the amount of ITC and the amount of the compound, or pharmaceutically acceptable salt of the compound, when taken together are effective to achieve a synergism in treating the subject. [0118] In some embodiments, the combination is more effective in treating the subject against fungal infection than when ITC and the compound, or pharmaceutically acceptable salt of the compound, at the same amount is administered alone. [0119] In some embodiments, the amount of the ITC and the amount of the compound, or pharmaceutically acceptable salt of the compound, when administered together, are more effective to treat the subject than when each at the same amount is administered alone. [0120] In some embodiments, the amount of the ITC administered in the combination is less than the fungicidally effective amount of the ITC when the ITC is administered alone. [0121] In some embodiments, the amount of the compound, or pharmaceutically acceptable salt thereof, administered in the combination is less than the fungicidally effective amount of the compound, or pharmaceutically acceptable salt thereof, when the compound, or pharmaceutically acceptable salt thereof, is administered alone. [0122] In some embodiments, the amount of ITC is administered in the range of 1mg/kg to 100 mg/kg subject body weight per day. [0123] In some embodiments, the amount of ITC is administered in the range of 1mg/kg to 70 mg/kg subject body weight per day. [0124] In some embodiments, the amount of ITC is administered in the range of 5mg/kg to 60 mg/kg subject body weight per day. [0125] In some embodiments, the amount of ITC is administered in the range of 10 mg/kg to 20 mg/kg subject body weight per day. [0126] In some embodiments, the amount of the compound, or pharmaceutically acceptable salt thereof, is administered in the range of 1mg/kg to 100 mg/kg subject body weight per day. [0127] In some embodiments, the amount of the compound, or pharmaceutically acceptable salt thereof, is administered in the range of 1mg/kg to 70 mg/kg subject body weight per day. [0128] In some embodiments, the amount of the compound, or pharmaceutically acceptable salt thereof, is administered 5mg/kg to 60 mg/kg subject body weight per day. [0129] In some embodiments, the amount of the compound, or pharmaceutically acceptable salt thereof, is administered10 mg/kg to 20 mg/kg subject body weight per day. [0130] In some embodiments, the ratio between the amount of ITC and the amount of the compound, or pharmaceutically acceptable salt thereof, is from 100:1 to 1:100, 50:1 to 1:50, 20:1 to 1:20, 10:1 to 1:10, 5:1 to 1:5 or 2:1 to 1:2. [0131] In some embodiments, the ratio between the amount of ITC and the amount of the compound, or pharmaceutically acceptable salt thereof, is 2:1. [0132] In some embodiments, the ratio between the amount of ITC and the amount of the compound, or pharmaceutically acceptable salt thereof, is 5:1. [0133] In some embodiments, the ratio between the amount of ITC and the amount of the compound, or pharmaceutically acceptable salt thereof, is from 100:1 to 1:2, 50:1 to 1:2, 20:1 to 1:2, 10:1 to 1:2, 5:1 to 1:2 or 2.5:1 to 1:2.5. [0134] In some embodiments, the ratio between the amount of ITC and the amount of the compound, or pharmaceutically acceptable salt thereof, is from 1:100 to 1:2, 1:50 to 1:2, 1:20 to 1:2, 1:10 to 1:2, 1:5 to 1:2 or 1:2.5 to 1:2. [0135] In some embodiments, the amount of ITC and the amount of the compound, or pharmaceutically acceptable salt thereof, is administered once, twice, or third times per day for a period of time. [0136] In some embodiments, the period is 4-72 weeks. [0137] In some embodiments, the period is 4-32 weeks. [0138] In some embodiments, the subject is a mammal. [0139] In some embodiments, the mammal is cat or human. [0140] In some embodiments, the mammal is cat. [0141] In some embodiments, the mammal is human. [0142] In some embodiments, the ITC and the compound, or pharmaceutically acceptable salt thereof, are administered together. [0143] In some embodiments, the ITC and the compound, or pharmaceutically acceptable salt thereof, are administered in a single dose. [0144] In some embodiments, the ITC and the compound, or pharmaceutically acceptable salt thereof, are administered separately. [0145] In some embodiments, the ITC and the compound, or pharmaceutically acceptable salt thereof, are administered simultaneously. [0146] In some embodiments, the ITC and the compound, or pharmaceutically acceptable salt thereof, are applied contemporaneously. [0147] In some embodiments, the ITC and the compound, or pharmaceutically acceptable salt thereof, are applied successively. [0148] In some embodiments, the method further comprises administering to the subject potassium iodine (KI). [0149] In some embodiments, the method further comprises administering to the subject sylimarin. [0150] In some embodiments, the method further comprises administering to the subject terbinafine. [0151] In some embodiments, the amounts of ITC, KI and the compound, or pharmaceutically acceptable salt thereof, when taken together are effective to achieve an additive or a greater than additive therapeutic result in treating the subject. [0152] In some embodiments, the amounts of ITC, KI and the compound, or pharmaceutically acceptable salt thereof, when taken together are effective to achieve a synergism in treating the subject. [0153] In some embodiments, the combination is more effective in treating the subject against fungal infection than when ITC, KI, and the compound, or pharmaceutically acceptable salt thereof, at the same amount is administered alone. [0154] In some embodiments, the amount of the ITC, KI, and the amount of the compound, or pharmaceutically acceptable salt thereof, when administered together, are more effective to treat the subject than when each at the same amount is administered alone. [0155] In some embodiments, the amount of the ITC and KI administered in the combination is less than the fungicidally effective amount of the ITC and KI when the ITC and KI is administered alone. [0156] In some embodiments, the amount of the compound, or pharmaceutically acceptable salt thereof, administered in the combination is less than the fungicidally effective amount of the compound, or pharmaceutically acceptable salt thereof, when the compound, or pharmaceutically acceptable salt thereof, is administered alone. [0157] In some embodiments, the amount of ITC is administered in the range of 1mg/kg to 200 mg/kg subject body weight per day. [0158] In some embodiments, the amount of ITC is administered in the range of 10mg/kg to 150 mg/kg subject body weight per day. [0159] In some embodiments, the amount of ITC is administered in the range of 50mg/kg to 120mg/kg subject body weight per day. [0160] In some embodiments, the amount of ITC is administered in the range of 80mg/kg to 120mg/kg subject body weight per day. [0161] In some embodiments, the amount of ITC is administered in the range of 90mg/kg to 110mg/kg subject body weight per day. [0162] In some embodiments, the amount of ITC is administered in the range of 10mg/kg to 100 mg/kg subject body weight per day. [0163] In some embodiments, the amount of ITC is administered in the range of 20mg/kg to 100 mg/kg subject body weight per day. [0164] In some embodiments, the amount of ITC is administered in the range of 30mg/kg to 100 mg/kg subject body weight per day. [0165] In some embodiments, the amount of ITC is administered in the range of 40mg/kg to 100 mg/kg subject body weight per day. [0166] In some embodiments, the amount of ITC is administered in the range of 50mg/kg to 100 mg/kg subject body weight per day. [0167] In some embodiments, the amount of ITC is administered in the range of 60mg/kg to 100 mg/kg subject body weight per day. [0168] In some embodiments, the amount of ITC is administered in the range of 70mg/kg to 100 mg/kg subject body weight per day. [0169] In some embodiments, the amount of ITC is administered in the range of 80mg/kg to 100 mg/kg subject body weight per day. [0170] In some embodiments, the amount of ITC is administered in the range of 90mg/kg to 100 mg/kg subject body weight per day. [0171] In some embodiments, the amount of ITC is administered in the range of 1mg/kg to 70 mg/kg subject body weight per day. [0172] In some embodiments, the amount of ITC is administered in the range of 5mg/kg to 60 mg/kg subject body weight per day. [0173] In some embodiments, the amount of ITC is administered in the range of 10 mg/kg to 20 mg/kg subject body weight per day. [0174] In some embodiments, the amount of ITC is administered 50 mg/kg subject body weight per day. [0175] In some embodiments, the amount of ITC is administered 100 mg/kg subject body weight per day. [0176] In some embodiments, the amount of KI is administered in the range of 1mg/kg to 100 mg/kg subject body weight per day. [0177] In some embodiments, the amount of KI is administered in the range of 1mg/kg to 20 mg/kg subject body weight per day. [0178] In some embodiments, the amount of KI is administered in the range of 2 mg/kg to 16 mg/kg subject body weight per day. [0179] In some embodiments, the amount of KI is administered in the range of 2 mg/kg to 10 mg/kg subject body weight per day. [0180] In some embodiments, the amount of KI is administered in the range of 1 mg/kg to 8 mg/kg subject body weight per day. [0181] In some embodiments, the amount of KI is administered in the range of 1 mg/kg to 6 mg/kg subject body weight per day. [0182] In some embodiments, the amount of KI is administered in the range of 2 mg/kg to 5mg/kg subject body weight per day. [0183] In some embodiments, the amount of KI is administered in the range of 5 mg/kg to 10 mg/kg subject body weight per day. [0184] In some embodiments, the amount of KI is 2.5 mg/kg subject body weight per day. [0185] In some embodiments, the amount of the compound, or pharmaceutically acceptable salt thereof, is administered in the range of 1mg/kg to 100 mg/kg subject body weight per day. [0186] In some embodiments, the amount of the compound, or pharmaceutically acceptable salt thereof, is administered in the range of 1mg/kg to 70 mg/kg subject body weight per day. [0187] In some embodiments, the amount of the compound, or pharmaceutically acceptable salt thereof, is administered in the range of 1 mg/kg to 70 mg/kg subject body weight per day. [0188] In some embodiments, the amount of the compound, or pharmaceutically acceptable salt thereof, is administered in the range of 5 mg/kg to 60 mg/kg subject body weight per day. [0189] In some embodiments, the amount of the compound, or pharmaceutically acceptable salt thereof, is administered in the range of 5 mg/kg to 50 mg/kg subject body weight per day. [0190] In some embodiments, the amount of the compound, or pharmaceutically acceptable salt thereof, is administered in the range of 5 mg/kg to 40 mg/kg subject body weight per day. [0191] In some embodiments, the amount of the compound, or pharmaceutically acceptable salt thereof, is administered in the range of 5 mg/kg to 40 mg/kg subject body weight per day. [0192] In some embodiments, the amount of the compound, or pharmaceutically acceptable salt thereof, is administered in the range of 10 mg/kg to 30 mg/kg subject body weight per day. [0193] In some embodiments, the amount of the compound, or pharmaceutically acceptable salt thereof, is administered in the range of 10 mg/kg to 20 mg/kg subject body weight per day. [0194] In some embodiments, the amount of the compound, or pharmaceutically acceptable salt thereof, is administered 20 mg/kg subject body weight per day. [0195] In some embodiments, the amount of sylimarin is administered in the range of 1mg/kg to 100 mg/kg subject body weight per day. [0196] In some embodiments, the amount of sylimarin is administered in the range of 1mg/kg to 70 mg/kg subject body weight per day. [0197] In some embodiments, the amount of sylimarin is administered in the range of 5 mg/kg to 60 mg/kg subject body weight per day. [0198] In some embodiments, the amount of sylimarin is administered in the range of 10 mg/kg to 50 mg/kg subject body weight per day. [0199] In some embodiments, the amount of sylimarin is administered in the range of 10 mg/kg to 40 mg/kg subject body weight per day. [0200] In some embodiments, the amount of sylimarin is administered in the range of 10 mg/kg to 30 mg/kg subject body weight per day. [0201] In some embodiments, the amount of sylimarin is administered in the range of 20 mg/kg to 40 mg/kg subject body weight per day. [0202] In some embodiments, the amount of sylimarin is administered in the range of 25 mg/kg to 35 mg/kg subject body weight per day. [0203] In some embodiments, the amount of sylimarin is administered 30 mg/kg subject body weight per day. [0204] In some embodiments, the amount of terbinafine is administered in the range of 1mg/kg to 100 mg/kg subject body weight per day. [0205] In some embodiments, the amount of terbinafine is administered in the range of 1mg/kg to 70 mg/kg subject body weight per day. [0206] In some embodiments, the amount of terbinafine is administered in the range of 5 mg/kg to 60 mg/kg subject body weight per day. [0207] In some embodiments, the amount of terbinafine is administered in the range of 10 mg/kg to 50 mg/kg subject body weight per day. [0208] In some embodiments, the amount of terbinafine is administered in the range of 10 mg/kg to 40 mg/kg subject body weight per day. [0209] In some embodiments, the amount of terbinafine is administered in the range of 10 mg/kg to 30 mg/kg subject body weight per day. [0210] In some embodiments, the amount of terbinafine is administered in the range of 20 mg/kg to 40 mg/kg subject body weight per day. [0211] In some embodiments, the amount of terbinafine is administered in the range of 25 mg/kg to 35 mg/kg subject body weight per day. [0212] In some embodiments, the amount of terbinafine is administered 30 mg/kg subject body weight per day. [0213] In some embodiments, the ratio between the amount of ITC and the amount of the compound, or pharmaceutically acceptable salt thereof, is from 100:1 to 1:100, 50:1 to 1:50, 20:1 to 1:20, 10:1 to 1:10, 5:1 to 1:5 or 2:1 to 1:2. [0214] In some embodiments, the ratio between the amount of ITC and the amount of the compound, or pharmaceutically acceptable salt thereof, is from 100:1 to 1:2, 50:1 to 1:2, 20:1 to 1:2, 10:1 to 1:2, 5:1 to 1:2 or 2:1 to 1:2. [0215] In some embodiments, the ratio between the amount of ITC and the amount of the compound, or pharmaceutically acceptable salt thereof, is from 2:1 to 1:100, 2:1 to 1:50, 2:1 to 1:20, 2:1 to 1:10, 2:1 to 1:5 or 2:1 to 1:2. [0216] In some embodiments, the ratio between the amount of ITC and the amount of the compound, or pharmaceutically acceptable salt thereof, is 2:1. [0217] In some embodiments, the ratio between the amount of ITC and the amount of the compound, or pharmaceutically acceptable salt thereof, is 5:1. [0218] In some embodiments, the ratio between the amount of ITC and KI is from 100:1 to 1:100, 50:1 to 1:50, 20:1 to 1:20, 10:1 to 1:10, 5:1 to 1:5 or 2:1 to 1:2. [0219] In some embodiments, the ratio between the amount of ITC and KI is from 100:1 to 1:2, 50:1 to 1:2, 20:1 to 1:2, 10:1 to 1:2, 5:1 to 1:2 or 2:1 to 1:2. [0220] In some embodiments, the ratio between the amount of ITC and KI is from 2:1 to 1:100, 2:1 to 1:50, 2:1 to 1:20, 2:1 to 1:10, 2:1 to 1:5 or 2:1 to 1:2. [0221] In some embodiments, the ratio between the amount of ITC and KI is 20:1. [0222] In some embodiments, the ratio between the amount of KI and the compound, or pharmaceutically acceptable salt thereof, is from 100:1 to 1:100, 50:1 to 1:50, 20:1 to 1:20, 10:1 to 1:10, 5:1 to 1:5 or 2:1 to 1:2. [0223] In some embodiments, the ratio between the amount of KI and the compound, or pharmaceutically acceptable salt thereof, is from 100:1 to 1:2, 50:1 to 1:2, 20:1 to 1:2, 10:1 to 1:2, 5:1 to 1:2 or 2:1 to 1:2. [0224] In some embodiments, the ratio between the amount of KI and the compound, or pharmaceutically acceptable salt thereof, is from 2:1 to 1:100, 2:1 to 1:50, 2:1 to 1:20, 2:1 to 1:10, 2:1 to 1:5 or 2:1 to 1:2. [0225] In some embodiments, the ITC, KI and the compound, or pharmaceutically acceptable salt thereof, are administered together. [0226] In some embodiments, the ITC, KI and the compound, or pharmaceutically acceptable salt thereof, are administered in a single dose. [0227] In some embodiments, the ITC, KI and the compound, or pharmaceutically acceptable salt thereof, are administered separately. [0228] In some embodiments, the ITC, KI and the compound, or pharmaceutically acceptable salt thereof, are administered simultaneously. [0229] In some embodiments, the ITC, KI and the compound, or pharmaceutically acceptable salt thereof, are applied contemporaneously. [0230] In some embodiments, the ITC, KI and the compound, or pharmaceutically acceptable salt thereof, are applied successively. [0231] In some embodiments, the amount of ITC, KI and the compound, or pharmaceutically acceptable salt thereof, is administered once twice, or third times per day for a period of time. [0232] In some embodiments, the period is 4-72 weeks. [0233] In some embodiments, the period is 4-32 weeks. [0234] In some embodiments, the period is 4 weeks. [0235] In some embodiments, the period is 3 months. [0236] In some embodiments, the period is 6 months. [0237] In some embodiments, the period is 8 months. [0238] In some embodiments, the subject is a mammal. [0239] In some embodiments, the mammal is cat or human. [0240] In some embodiments, the fungal infection is caused by Cryptococcus Neoformans, Cryptococcus gattii, Candida albicans, Candida krusei, Candida glabrata, Candida parapsilosis, Candida guilliermondii, Aspergillus fumigatus, Rhizopus oryzae, Rhizopus spp., Blastomyces dermatitis, Histoplasma capsulatum, Coccidioides spp., Paecilomyces variotii, Pneumocystis murina, Pneumocystis jiroveci, Histoplasma capsulatum, Aspergillus spp., S. brasiliensis, S. schenckii, S. globosa, S. mexicana, S. chilensis, S. luriei, or S. pallida. [0241] In some embodiments, the fungal infection is caused by is Cryptococcus Neoformans or Sporothrix brasiliensis. [0242] In some embodiments, the fungal infection is caused by Sporothrix brasiliensis. [0243] The present invention provides a combination comprising an amount of itraconazole (ITC) and an amount of a compound, or a pharmaceutically acceptable salt thereof, wherein the compound has the following structure: ,
Figure imgf000041_0001
R1 is -H, alkyl, alkenyl, or alkynyl; R2 is -H, alkyl, alkenyl, or alkynyl; R9, R10, R11, and R12 are each independently, H, halogen, -CN, -CF3, -OCF3, -NO2, alkyl, alkenyl, alkynyl, aryl, heteroaryl, cycloalkyl, heterocycloalkyl, -OH, -OAc, -OR13, -COR13, -SH, -SR13, - SO2R13, -SO2NR14R15, -NH2, -NHR13, -NR14R15, -NHCOR12, or -CONR14R15; or R9 and R10 are each independently -H, halogen, C1-C6 alkyl, -OH, -O-(C1-C6 alkyl), -CHF2, -CF3, -OCHF2 or -OCF3, and R11 and R12 combine to form a fused aryl or fused heteroaryl, which are each unsubstituted or substituted, or R9 and R12 are each independently -H, halogen, C1-C6 alkyl, -OH, -O-(C1-C6 alkyl), -CHF2, -CF3, -OCHF2 or -OCF3, and R10 and R11 combine to form a fused aryl or fused heteroaryl, which are each unsubstituted or substitute, or R11 and R12 are each independently -H, halogen, C1-C6 alkyl, -OH, -O-(C1-C6 alkyl), -CHF2, -CF3, -OCHF2 or -OCF3, and R3 and R4 combine to form a fused aryl or fused heteroaryl, which are each unsubstituted or substituted; wherein each occurrence of R13 is independently alkyl, alkenyl, alkynyl, aryl, or heteroaryl; wherein each occurrence of R14 and R15 is independently –H, alkyl, alkenyl, alkynyl, aryl, or heteroaryl; wherein when R14 is methyl, R15 is not methyl; and wherein at least one of R9, R10, R11, and R12 is not H; for use in treating a subject afflicted with a fungal infection by administering the combination to the subject. [0244] In some embodiments, the amount of the compound, or pharmaceutically acceptable salt thereof, improves the fungicidal efficacy of ITC compared to when the same amount of the ITC is administered not in combination with the amount of the compound, or pharmaceutically acceptable salt thereof. [0245] In some embodiments, the amount of ITC improves the fungicidal efficacy of the compound, or pharmaceutically acceptable salt thereof, compared to when the same amount of the compound, or pharmaceutically acceptable salt thereof, is applied not in combination with the amount of ITC. [0246] In some embodiments, a substantially similar level of fungicidal efficacy is achieved by using a lesser amount of the ITC and/or the compound, or pharmaceutically acceptable salt thereof. [0247] In some embodiments, the amount of the compound, or pharmaceutically acceptable salt thereof, is effective to increase sensitivity of the fungus to the amount of ITC compared to the sensitivity of the fungus to the amount of the ITC when it is administered not in combination with the amount of the compound, or pharmaceutically acceptable salt thereof. [0248] In some embodiments, the amount of the ITC is effective to increase sensitivity of the fungus to the amount of the compound, or pharmaceutically acceptable salt thereof, compared to the sensitivity of the fungus to the amount of the compound, or pharmaceutically acceptable salt thereof, when it is administered not in combination with the amount of ITC. [0249] In some embodiments, the amount of the compound, or pharmaceutically acceptable salt thereof, is effective to increase bioavailability of the amount of ITC compared to the bioavailability of the amount of the ITC when it is administered not in combination with the amount of the compound, or pharmaceutically acceptable salt thereof. [0250] In some embodiments, the amount of ITC is effective to increase bioavailability of the amount of the compound, or pharmaceutically acceptable salt thereof, compared to the bioavailability of the amount of the compound, or pharmaceutically acceptable salt thereof, when it is administered not in combination with the amount of ITC. [0251] In some embodiments, the combination is more effective in treating the subject against fungal infection than when ITC and the compound, or pharmaceutically acceptable salt thereof, at the same amount is administered alone. [0252] In some embodiments, the combination prolongs the period of protection against fungal infection and/or control of fungal infection than when the amount of ITC and the amount of the compound, or pharmaceutically acceptable salt thereof, are administered alone. [0253] In some embodiments, the combination reduces the amount of time needed to achieve a level of fungal control than when the amount of ITC and the amount of the compound, or pharmaceutically acceptable salt thereof, are administered alone. [0254] The present invention provides a pharmaceutical composition in unit dosage form, useful in treating a subject afflicted with fungal infection, which comprises: (a) an amount of ITC; (b) an amount of the compound described herein, or a pharmaceutically acceptable salt thereof; (c) optionally an amount of KI; wherein the respective amounts of said compound, or a pharmaceutically acceptable salt thereof, said ITC and said KI, if present, in said composition are effective, upon concomitant administration to said subject of one or more of said unit dosage forms of said composition, to treat the subject. [0255] The present invention provides a therapeutic package for dispensing to, or for use in dispensing to, a subject afflicted with fungal infection, which comprises: (c) one or more unit doses, each such unit dose comprising: iv) an amount of ITC; v) an amount of the compound described herein, or a pharmaceutically acceptable salt thereof; vi) optionally an amount of KI; wherein the respective amounts of said compound, or a pharmaceutically acceptable salt thereof, said ITC and said KI, if present, in said unit doses are effective, upon concomitant administration to said subject, to treat the subject, and (d) a finished pharmaceutical container therefor, said container containing said unit dose or unit doses, said container further containing or comprising labeling directing the use of said package in the treatment of said subject. [0256] In some embodiments, the pharmaceutical composition, unit dose, or unit dosage form comprises: (a) 25-75 mg of the compound, or a pharmaceutically acceptable salt thereof, 10-100 mg of ITC, 1-20 mg of KI, if present. (b) 25-75 mg of the compound, or a pharmaceutically acceptable salt thereof, 10-100 mg of ITC, 1-20 mg of KI, if present and 11-50 mg of sylimarin, if present. [0257] In some embodiments, the pharmaceutical composition, unit dose, or unit dosage form comprises: (a) 25 mg of the compound, or a pharmaceutically acceptable salt thereof, 25 mg of ITC, 2.5 mg of KI and 30 mg sylimarin; (b) 20 mg of the compound, or a pharmaceutically acceptable salt thereof, 50 mg of ITC, and 2.5 mg of KI; and (c) 20 mg of the compound or a pharmaceutically acceptable salt thereof, and 100 mg of ITC. [0258] In some embodiments, the unit dosage form is administered to the subject in a capsule. [0259] The synthetic schemes of compounds described herein can be found in International Publication No. WO 2016/094307, International Publication No. WO 2018/232298, and International Publication No. WO 2023/060161, the entire contents of which are hereby incorporated by reference. Definitions [0260] Unless otherwise defined, all technical and/or scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which the invention pertains. Although methods and materials similar or equivalent to those described herein can be used in the practice or testing of embodiments of the invention, exemplary methods and/or materials are described below. In case of conflict, the patent specification, including definitions, will control. In addition, the materials, methods, and examples are illustrative only and are not intended to be necessarily limiting. [0261] In the discussion unless otherwise stated, adjectives such as “substantially” and “about” modifying a condition or relationship characteristic of a feature or features of an embodiment of the invention, are understood to mean that the condition or characteristic is defined to within tolerances that are acceptable for operation of the embodiment for an application for which it is intended. In embodiments, about means within a standard deviation using measurements generally acceptable in the art. In embodiments, about means a range extending to +/- 10% of the specified value. In embodiments, about includes the specified value. Unless otherwise indicated, the word “or” in the specification and claims is considered to be the inclusive “or” rather than the exclusive or, and indicates at least one of and any combination of items it conjoins. [0262] It should be understood that the terms “a” and “an” as used above and elsewhere herein refer to “one or more” of the enumerated components. It will be clear to one of ordinary skill in the art that the use of the singular includes the plural unless specifically stated otherwise. Therefore, the terms “a,” “an” and “at least one” are used interchangeably in this application. [0263] For purposes of better understanding the present teachings and in no way limiting the scope of the teachings, unless otherwise indicated, all numbers expressing quantities, percentages or proportions, and other numerical values used in the specification and claims, are to be understood as being modified in all instances by the term “about.” Accordingly, unless indicated to the contrary, the numerical parameters set forth in the following specification and attached claims are approximations that may vary depending upon the desired properties sought to be obtained. At the very least, each numerical parameter should at least be construed in light of the number of reported significant digits and by applying ordinary rounding techniques. [0264] In the description and claims of the present application, each of the verbs, “comprise,” “include” and “have” and conjugates thereof, are used to indicate that the object or objects of the verb are not necessarily a complete listing of components, elements or parts of the subject or subjects of the verb. Other terms as used herein are meant to be defined by their well-known meanings in the art. [0265] As used herein, a “symptom” associated with a fungal infection includes any clinical or laboratory manifestation associated with the fungal infection and is not limited to what the subject can feel or observe. [0266] As used herein, “treating”, e.g. of a fungal infection, encompasses inducing prevention, inhibition, regression, or stasis of the disease or a symptom or condition associated with the infection. [0267] The compounds of the present invention include all hydrates, solvates, and complexes of the compounds used by this invention. If a chiral center or another form of an isomeric center is present in a compound of the present invention, all forms of such isomer or isomers, including enantiomers and diastereomers, are intended to be covered herein. Compounds containing a chiral center may be used as a racemic mixture, an enantiomerically enriched mixture, or the racemic mixture may be separated using well-known techniques and an individual enantiomer may be used alone. The compounds described in the present invention are in racemic form or as individual enantiomers. The enantiomers can be separated using known techniques, such as those described in Pure and Applied Chemistry 69, 1469–1474, (1997) IUPAC. In cases in which compounds have unsaturated carbon-carbon double bonds, both the cis (Z) and trans (E) isomers are within the scope of this invention. [0268] The compounds of the subject invention may have spontaneous tautomeric forms. In cases wherein compounds may exist in tautomeric forms, such as keto-enol tautomers, each tautomeric form is contemplated as being included within this invention whether existing in equilibrium or predominantly in one form. [0269] In the compound structures depicted herein, hydrogen atoms are not shown for carbon atoms having less than four bonds to non-hydrogen atoms. However, it is understood that enough hydrogen atoms exist on said carbon atoms to satisfy the octet rule. [0270] This invention also provides isotopic variants of the compounds disclosed herein, including wherein the isotopic atom is 2H and/or wherein the isotopic atom 13C. Accordingly, in the compounds provided herein hydrogen can be enriched in the deuterium isotope. It is to be understood that the invention encompasses all such isotopic forms. [0271] It is understood that the structures described in the embodiments of the methods hereinabove can be the same as the structures of the compounds described hereinabove. [0272] It is understood that where a numerical range is recited herein, the present invention contemplates each integer between, and including, the upper and lower limits, unless otherwise stated. [0273] Except where otherwise specified, if the structure of a compound of this invention includes an asymmetric carbon atom, it is understood that the compound occurs as a racemate, racemic mixture, and isolated single enantiomer. All such isomeric forms of these compounds are expressly included in this invention. Except where otherwise specified, each stereogenic carbon may be of the R or S configuration. It is to be understood accordingly that the isomers arising from such asymmetry (e.g., all enantiomers and diastereomers) are included within the scope of this invention, unless indicated otherwise. Such isomers can be obtained in substantially pure form by classical separation techniques and by stereochemically controlled synthesis, such as those described in "Enantiomers, Racemates and Resolutions" by J. Jacques, A. Collet and S. Wilen, Pub. John Wiley & Sons, NY, 1981. For example, the resolution may be carried out by preparative chromatography on a chiral column. [0274] The subject invention is also intended to include all isotopes of atoms occurring on the compounds disclosed herein. Isotopes include those atoms having the same atomic number but different mass numbers. By way of general example and without limitation, isotopes of hydrogen include tritium and deuterium. Isotopes of carbon include C-13 and C-14. [0275] It will be noted that any notation of a carbon in structures throughout this application, when used without further notation, are intended to represent all isotopes of carbon, such as 12C, 13C, or 14C. Furthermore, any compounds containing 13C or 14C may specifically have the structure of any of the compounds disclosed herein. [0276] It will also be noted that any notation of a hydrogen in structures throughout this application, when used without further notation, are intended to represent all isotopes of hydrogen, such as 1H, 2H, or 3H. Furthermore, any compounds containing 2H or 3H may specifically have the structure of any of the compounds disclosed herein. [0277] Isotopically-labeled compounds can generally be prepared by conventional techniques known to those skilled in the art using appropriate isotopically-labeled reagents in place of the non-labeled reagents employed. [0278] In the compounds used in the method of the present invention, the substituents may be substituted or unsubstituted, unless specifically defined otherwise. [0279] In the compounds used in the method of the present invention, alkyl, heteroalkyl, monocycle, bicycle, aryl, heteroaryl and heterocycle groups can be further substituted by replacing one or more hydrogen atoms with alternative non-hydrogen groups. These include, but are not limited to, halo, hydroxy, mercapto, amino, carboxy, cyano, carbamoyl and aminocarbonyl and aminothiocarbonyl. [0280] It is understood that substituents and substitution patterns on the compounds used in the method of the present invention can be selected by one of ordinary skill in the art to provide compounds that are chemically stable and that can be readily synthesized by techniques known in the art from readily available starting materials. If a substituent is itself substituted with more than one group, it is understood that these multiple groups may be on the same carbon or on different carbons, so long as a stable structure results. [0281] In choosing the compounds used in the method of the present invention, one of ordinary skill in the art will recognize that the various substituents, i.e. R1, R2, etc. are to be chosen in conformity with well-known principles of chemical structure connectivity. [0282] As used herein, "alkyl" is intended to include both branched and straight-chain saturated aliphatic hydrocarbon groups having the specified number of carbon atoms. Thus, C1-Cn as in “C1–Cn alkyl" is defined to include groups having 1, 2......, n-1 or n carbons in a linear or branched arrangement, and specifically includes methyl, ethyl, propyl, butyl, pentyl, hexyl, heptyl, isopropyl, isobutyl, sec-butyl and so on. An embodiment can be C1-C12 alkyl, C2-C12 alkyl, C3-C12 alkyl, C4-C12 alkyl and so on. ”Alkoxy" represents an alkyl group as described above attached through an oxygen bridge. [0283] The term "alkenyl" refers to a non-aromatic hydrocarbon radical, straight or branched, containing at least 1 carbon to carbon double bond, and up to the maximum possible number of non- aromatic carbon-carbon double bonds may be present. Thus, C2-Cn alkenyl is defined to include groups having 1, 2...., n-1 or n carbons. For example, "C2-C6 alkenyl" means an alkenyl radical having 2, 3, 4, 5, or 6 carbon atoms, and at least 1 carbon-carbon double bond, and up to, for example, 3 carbon-carbon double bonds in the case of a C6 alkenyl, respectively. Alkenyl groups include ethenyl, propenyl, butenyl and cyclohexenyl. As described above with respect to alkyl, the straight, branched or cyclic portion of the alkenyl group may contain double bonds and may be substituted if a substituted alkenyl group is indicated. An embodiment can be C2-C12 alkenyl, C3-C12 alkenyl, C4-C12 alkenyl and so on. [0284] The term "alkynyl" refers to a hydrocarbon radical straight or branched, containing at least 1 carbon to carbon triple bond, and up to the maximum possible number of non-aromatic carbon-carbon triple bonds may be present. Thus, C2-Cn alkynyl is defined to include groups having 1, 2...., n-1 or n carbons. For example, "C2-C6 alkynyl" means an alkynyl radical having 2 or 3 carbon atoms, and 1 carbon-carbon triple bond, or having 4 or 5 carbon atoms, and up to 2 carbon-carbon triple bonds, or having 6 carbon atoms, and up to 3 carbon-carbon triple bonds. Alkynyl groups include ethynyl, propynyl and butynyl. As described above with respect to alkyl, the straight or branched portion of the alkynyl group may contain triple bonds and may be substituted if a substituted alkynyl group is indicated. An embodiment can be a C2- Cn alkynyl. An embodiment can be C2-C12 alkynyl, C3-C12 alkynyl, C4-C12 alkynyl and so on [0285] “Alkylene”, “alkenylene” and “alkynylene” shall mean, respectively, a divalent alkane, alkene and alkyne radical, respectively. It is understood that an alkylene, alkenylene, and alkynylene may be straight or branched. An alkylene, alkenylene, and alkynylene may be unsubstituted or substituted. [0286] As used herein, "heteroalkyl" includes both branched and straight-chain saturated aliphatic hydrocarbon groups having the specified number of carbon atoms and at least 1 heteroatom within the chain or branch. [0287] As used herein, "heterocycle" or "heterocyclyl" as used herein is intended to mean a 5- to 10- membered nonaromatic ring containing from 1 to 4 heteroatoms selected from the group consisting of O, N and S, and includes bicyclic groups. "Heterocyclyl" therefore includes, but is not limited to the following: imidazolyl, piperazinyl, piperidinyl, pyrrolidinyl, morpholinyl, thiomorpholinyl, tetrahydropyranyl, dihydropiperidinyl, tetrahydrothiophenyl and the like. If the heterocycle contains a nitrogen, it is understood that the corresponding N-oxides thereof are also encompassed by this definition. [0288] As herein, "cycloalkyl" shall mean cyclic rings of alkanes of three to eight total carbon atoms, or any number within this range (i.e., cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl or cyclooctyl). [0289] As used herein, "monocycle" includes any stable polyatomic carbon ring of up to 10 atoms and may be unsubstituted or substituted. Examples of such non-aromatic monocycle elements include but are not limited to: cyclobutyl, cyclopentyl, cyclohexyl, and cycloheptyl. Examples of such aromatic monocycle elements include but are not limited to: phenyl. [0290] As used herein, "bicycle" includes any stable polyatomic carbon ring of up to 10 atoms that is fused to a polyatomic carbon ring of up to 10 atoms with each ring being independently unsubstituted or substituted. Examples of such non-aromatic bicycle elements include but are not limited to: decahydronaphthalene. Examples of such aromatic bicycle elements include but are not limited to: naphthalene. [0291] As used herein, "aryl" is intended to mean any stable monocyclic, bicyclic or polycyclic carbon ring of up to 10 atoms in each ring, wherein at least one ring is aromatic, and may be unsubstituted or substituted. Examples of such aryl elements include phenyl, p-toluenyl (4-methylphenyl), naphthyl, tetrahydro-naphthyl, indanyl, biphenyl, phenanthryl, anthryl or acenaphthyl. In cases where the aryl substituent is bicyclic and one ring is non-aromatic, it is understood that attachment is via the aromatic ring. [0292] As used herein, the term “polycyclic” refers to unsaturated or partially unsaturated multiple fused ring structures, which may be unsubstituted or substituted. [0293] The term “arylalkyl” refers to alkyl groups as described above wherein one or more bonds to hydrogen contained therein are replaced by a bond to an aryl group as described above. It is understood that an “arylalkyl” group is connected to a core molecule through a bond from the alkyl group and that the aryl group acts as a substituent on the alkyl group. Examples of arylalkyl moieties include, but are not limited to, benzyl (phenylmethyl), p-trifluoromethylbenzyl (4-trifluoromethylphenylmethyl), 1-phenylethyl, 2- phenylethyl, 3-phenylpropyl, 2-phenylpropyl and the like. [0294] The term "heteroaryl", as used herein, represents a stable monocyclic, bicyclic or polycyclic ring of up to 10 atoms in each ring, wherein at least one ring is aromatic and contains from 1 to 4 heteroatoms selected from the group consisting of O, N and S. Bicyclic aromatic heteroaryl groups include phenyl, pyridine, pyrimidine or pyridizine rings that are (a) fused to a 6-membered aromatic (unsaturated) heterocyclic ring having one nitrogen atom; (b) fused to a 5- or 6-membered aromatic (unsaturated) heterocyclic ring having two nitrogen atoms; (c) fused to a 5-membered aromatic (unsaturated) heterocyclic ring having one nitrogen atom together with either one oxygen or one sulfur atom; or (d) fused to a 5- membered aromatic (unsaturated) heterocyclic ring having one heteroatom selected from O, N or S. Heteroaryl groups within the scope of this definition include but are not limited to: benzoimidazolyl, benzofuranyl, benzofurazanyl, benzopyrazolyl, benzotriazolyl, benzothiophenyl, benzoxazolyl, carbazolyl, carbolinyl, cinnolinyl, furanyl, indolinyl, indolyl, indolazinyl, indazolyl, isobenzofuranyl, isoindolyl, isoquinolyl, isothiazolyl, isoxazolyl, naphthpyridinyl, oxadiazolyl, oxazolyl, oxazoline, isoxazoline, oxetanyl, pyranyl, pyrazinyl, pyrazolyl, pyridazinyl, pyridopyridinyl, pyridazinyl, pyridyl, pyrimidyl, pyrrolyl, quinazolinyl, quinolyl, quinoxalinyl, tetrazolyl, tetrazolopyridyl, thiadiazolyl, thiazolyl, thienyl, triazolyl, azetidinyl, aziridinyl, 1,4-dioxanyl, hexahydroazepinyl, dihydrobenzoimidazolyl, dihydrobenzofuranyl, dihydrobenzothiophenyl, dihydrobenzoxazolyl, dihydrofuranyl, dihydroimidazolyl, dihydroindolyl, dihydroisooxazolyl, dihydroisothiazolyl, dihydrooxadiazolyl, dihydrooxazolyl, dihydropyrazinyl, dihydropyrazolyl, dihydropyridinyl, dihydropyrimidinyl, dihydropyrrolyl, dihydroquinolinyl, dihydrotetrazolyl, dihydrothiadiazolyl, dihydrothiazolyl, dihydrothienyl, dihydrotriazolyl, dihydroazetidinyl, methylenedioxybenzoyl, tetrahydrofuranyl, tetrahydrothienyl, acridinyl, carbazolyl, cinnolinyl, quinoxalinyl, pyrrazolyl, indolyl, benzotriazolyl, benzothiazolyl, benzoxazolyl, isoxazolyl, isothiazolyl, furanyl, thienyl, benzothienyl, benzofuranyl, quinolinyl, isoquinolinyl, oxazolyl, isoxazolyl, indolyl, pyrazinyl, pyridazinyl, pyridinyl, pyrimidinyl, pyrrolyl, tetra- hydroquinoline. In cases where the heteroaryl substituent is bicyclic and one ring is non-aromatic or contains no heteroatoms, it is understood that attachment is via the aromatic ring or via the heteroatom containing ring, respectively. If the heteroaryl contains nitrogen atoms, it is understood that the corresponding N-oxides thereof are also encompassed by this definition. [0295] The term “alkylheteroaryl” refers to alkyl groups as described above wherein one or more bonds to hydrogen contained therein are replaced by a bond to an heteroaryl group as described above. It is understood that an “alkylheteroaryl” group is connected to a core molecule through a bond from the alkyl group and that the heteroaryl group acts as a substituent on the alkyl group. Examples of alkylheteroaryl moieties include, but are not limited to, -CH2-(C5H4N), -CH2-CH2-(C5H4N) and the like. [0296] The term "heterocycle" or “heterocyclyl” refers to a mono- or poly-cyclic ring system which can be saturated or contains one or more degrees of unsaturation and contains one or more heteroatoms. Preferred heteroatoms include N, O, and/or S, including N-oxides, sulfur oxides, and dioxides. Preferably the ring is three to ten-membered and is either saturated or has one or more degrees of unsaturation. The heterocycle may be unsubstituted or substituted, with multiple degrees of substitution being allowed. Such rings may be optionally fused to one or more of another "heterocyclic" ring(s), heteroaryl ring(s), aryl ring(s), or cycloalkyl ring(s). Examples of heterocycles include, but are not limited to, tetrahydrofuran, pyran, 1,4-dioxane, 1,3-dioxane, piperidine, piperazine, pyrrolidine, morpholine, thiomorpholine, tetrahydrothiopyran, tetrahydrothiophene, 1,3-oxathiolane, and the like. [0297] The alkyl, alkenyl, alkynyl, aryl, heteroaryl and heterocyclyl substituents may be substituted or unsubstituted, unless specifically defined otherwise. In the compounds of the present invention, alkyl, alkenyl, alkynyl, aryl, heterocyclyl and heteroaryl groups can be further substituted by replacing one or more hydrogen atoms with alternative non-hydrogen groups. These include, but are not limited to, halo, hydroxy, mercapto, amino, carboxy, cyano and carbamoyl. [0298] As used herein, the term “halogen” refers to F, Cl, Br, and I. [0299] The terms “substitution”, “substituted” and “substituent” refer to a functional group as described above in which one or more bonds to a hydrogen atom contained therein are replaced by a bond to non-hydrogen or non-carbon atoms, provided that normal valencies are maintained and that the substitution results in a stable compound. Substituted groups also include groups in which one or more bonds to a carbon(s) or hydrogen(s) atom are replaced by one or more bonds, including double or triple bonds, to a heteroatom. Examples of substituent groups include the functional groups described above, and halogens (i.e., F, Cl, Br, and I); alkyl groups, such as methyl, ethyl, n-propyl, isopropryl, n-butyl, tert-butyl, and trifluoromethyl; hydroxyl; alkoxy groups, such as methoxy, ethoxy, n-propoxy, and isopropoxy; aryloxy groups, such as phenoxy; arylalkyloxy, such as benzyloxy (phenylmethoxy) and p- trifluoromethylbenzyloxy (4-trifluoromethylphenylmethoxy); heteroaryloxy groups; sulfonyl groups, such as trifluoromethanesulfonyl, methanesulfonyl, and p-toluenesulfonyl; nitro, nitrosyl; mercapto; sulfanyl groups, such as methylsulfanyl, ethylsulfanyl and propylsulfanyl; cyano; amino groups, such as amino, methylamino, dimethylamino, ethylamino, and diethylamino; and carboxyl. Where multiple substituent moieties are disclosed or claimed, the substituted compound can be independently substituted by one or more of the disclosed or claimed substituent moieties, singly or pluraly. By independently substituted, it is meant that the (two or more) substituents can be the same or different. [0300] It is understood that substituents and substitution patterns on the compounds of the instant invention can be selected by one of ordinary skill in the art to provide compounds that are chemically stable and that can be readily synthesized by techniques known in the art, as well as those methods set forth below, from readily available starting materials. If a substituent is itself substituted with more than one group, it is understood that these multiple groups may be on the same carbon or on different carbons, so long as a stable structure results. [0301] In choosing the compounds of the present invention, one of ordinary skill in the art will recognize that the various substituents, i.e. R1, R2, etc. are to be chosen in conformity with well-known principles of chemical structure connectivity. [0302] The various R groups attached to the aromatic rings of the compounds disclosed herein may be added to the rings by standard procedures, for example those set forth in Advanced Organic Chemistry: Part B: Reaction and Synthesis, Francis Carey and Richard Sundberg, (Springer) 5th ed. Edition. (2007), the content of which is hereby incorporated by reference. [0303] The compounds used in the method of the present invention may be prepared by techniques well known in organic synthesis and familiar to a practitioner ordinarily skilled in the art. However, these may not be the only means by which to synthesize or obtain the desired compounds. [0304] The compounds used in the method of the present invention may be prepared by techniques described in Vogel’s Textbook of Practical Organic Chemistry, A.I. Vogel, A.R. Tatchell, B.S. Furnis, A.J. Hannaford, P.W.G. Smith, (Prentice Hall) 5th Edition (1996), March's Advanced Organic Chemistry: Reactions, Mechanisms, and Structure, Michael B. Smith, Jerry March, (Wiley-Interscience) 5th Edition (2007), and references therein, which are incorporated by reference herein. However, these may not be the only means by which to synthesize or obtain the desired compounds. [0305] Another aspect of the invention comprises a compound used in the method of the present invention as a pharmaceutical composition. [0306] In some embodiments, a pharmaceutical composition comprising the compound of the present invention and a pharmaceutically acceptable carrier. [0307] As used herein, the term “pharmaceutically active agent” means any substance or compound suitable for administration to a subject and furnishes biological activity or other direct effect in the treatment, cure, mitigation, diagnosis, or prevention of disease, or affects the structure or any function of the subject. Pharmaceutically active agents include, but are not limited to, substances and compounds described in the Physicians’ Desk Reference (PDR Network, LLC; 64th edition; November 15, 2009) and “Approved Drug Products with Therapeutic Equivalence Evaluations” (U.S. Department Of Health And Human Services, 30th edition, 2010), which are hereby incorporated by reference. Pharmaceutically active agents which have pendant carboxylic acid groups may be modified in accordance with the present invention using standard esterification reactions and methods readily available and known to those having ordinary skill in the art of chemical synthesis. Where a pharmaceutically active agent does not possess a carboxylic acid group, the ordinarily skilled artisan will be able to design and incorporate a carboxylic acid group into the pharmaceutically active agent where esterification may subsequently be carried out so long as the modification does not interfere with the pharmaceutically active agent’s biological activity or effect. [0308] The compounds used in the method of the present invention may be in a salt form. As used herein, a “salt” is a salt of the instant compounds which has been modified by making acid or base salts of the compounds. In the case of compounds used to treat an infection or disease caused by a pathogen, the salt is pharmaceutically acceptable. Examples of pharmaceutically acceptable salts include, but are not limited to, mineral or organic acid salts of basic residues such as amines; alkali or organic salts of acidic residues such as phenols. The salts can be made using an organic or inorganic acid. Such acid salts are chlorides, bromides, sulfates, nitrates, phosphates, sulfonates, formates, tartrates, maleates, malates, citrates, benzoates, salicylates, ascorbates, and the like. Phenolate salts are the alkaline earth metal salts, sodium, potassium or lithium. The term "pharmaceutically acceptable salt" in this respect, refers to the relatively non-toxic, inorganic and organic acid or base addition salts of compounds of the present invention. These salts can be prepared in situ during the final isolation and purification of the compounds of the invention, or by separately reacting a purified compound of the invention in its free base or free acid form with a suitable organic or inorganic acid or base, and isolating the salt thus formed. Representative salts include the hydrobromide, hydrochloride, sulfate, bisulfate, phosphate, nitrate, acetate, valerate, oleate, palmitate, stearate, laurate, benzoate, lactate, phosphate, tosylate, citrate, maleate, fumarate, succinate, tartrate, napthylate, mesylate, glucoheptonate, lactobionate, and laurylsulphonate salts and the like. (See, e.g., Berge et al. (1977) "Pharmaceutical Salts", J. Pharm. Sci.66:1-19). [0309] The compounds of the present invention may also form salts with basic amino acids such a lysine, arginine, etc. and with basic sugars such as N-methylglucamine, 2-amino-2-deoxyglucose, etc. and any other physiologically non-toxic basic substance. [0310] As used herein, “administering” an agent may be performed using any of the various methods or delivery systems well known to those skilled in the art. The administering can be performed, for example, orally, parenterally, intraperitoneally, intravenously, intraarterially, transdermally, sublingually, intramuscularly, rectally, transbuccally, intranasally, liposomally, via inhalation, vaginally, intraoccularly, via local delivery, subcutaneously, intraadiposally, intraarticularly, intrathecally, into a cerebral ventricle, intraventicularly, intratumorally, into cerebral parenchyma or intraparenchchymally. [0311] The compounds used in the method of the present invention may be administered in various forms, including those detailed herein. The treatment with the compound may be a component of a combination therapy or an adjunct therapy, i.e. the subject or patient in need of the drug is treated or given another drug for the disease in conjunction with one or more of the instant compounds. This combination therapy can be sequential therapy where the patient is treated first with one drug and then the other or the two drugs are given simultaneously. These can be administered independently by the same route or by two or more different routes of administration depending on the dosage forms employed. [0312] As used herein, a "pharmaceutically acceptable carrier" is a pharmaceutically acceptable solvent, suspending agent or vehicle, for delivering the instant compounds to the animal or human. The carrier may be liquid or solid and is selected with the planned manner of administration in mind. Liposomes are also a pharmaceutically acceptable carrier as are slow-release vehicles. [0313] The dosage of the compounds administered in treatment will vary depending upon factors such as the pharmacodynamic characteristics of a specific chemotherapeutic agent and its mode and route of administration; the age, sex, metabolic rate, absorptive efficiency, health and weight of the recipient; the nature and extent of the symptoms; the kind of concurrent treatment being administered; the frequency of treatment with; and the desired therapeutic effect. [0314] A dosage unit of the compounds used in the method of the present invention may comprise a single compound or mixtures thereof with additional antitumor agents. The compounds can be administered in oral dosage forms as tablets, capsules, pills, powders, granules, elixirs, tinctures, suspensions, syrups, and emulsions. The compounds may also be administered in intravenous (bolus or infusion), intraperitoneal, subcutaneous, or intramuscular form, or introduced directly, e.g. by injection, topical application, or other methods, into or topically onto a site of disease or lesion, all using dosage forms well known to those of ordinary skill in the pharmaceutical arts. [0315] The compounds used in the method of the present invention can be administered in admixture with suitable pharmaceutical diluents, extenders, excipients, or in carriers such as the novel programmable sustained-release multi-compartmental nanospheres (collectively referred to herein as a pharmaceutically acceptable carrier) suitably selected with respect to the intended form of administration and as consistent with conventional pharmaceutical practices. The unit will be in a form suitable for oral, nasal, rectal, topical, intravenous or direct injection or parenteral administration. The compounds can be administered alone or mixed with a pharmaceutically acceptable carrier. This carrier can be a solid or liquid, and the type of carrier is generally chosen based on the type of administration being used. The active agent can be co-administered in the form of a tablet or capsule, liposome, as an agglomerated powder or in a liquid form. Examples of suitable solid carriers include lactose, sucrose, gelatin and agar. Capsule or tablets can be easily formulated and can be made easy to swallow or chew; other solid forms include granules, and bulk powders. Tablets may contain suitable binders, lubricants, diluents, disintegrating agents, coloring agents, flavoring agents, flow-inducing agents, and melting agents. Examples of suitable liquid dosage forms include solutions or suspensions in water, pharmaceutically acceptable fats and oils, alcohols or other organic solvents, including esters, emulsions, syrups or elixirs, suspensions, solutions and/or suspensions reconstituted from non-effervescent granules and effervescent preparations reconstituted from effervescent granules. Such liquid dosage forms may contain, for example, suitable solvents, preservatives, emulsifying agents, suspending agents, diluents, sweeteners, thickeners, and melting agents. Oral dosage forms optionally contain flavorants and coloring agents. Parenteral and intravenous forms may also include minerals and other materials to make them compatible with the type of injection or delivery system chosen. [0316] Techniques and compositions for making dosage forms useful in the present invention are described in the following references: 7 Modern Pharmaceutics, Chapters 9 and 10 (Banker & Rhodes, Editors, 1979); Pharmaceutical Dosage Forms: Tablets (Lieberman et al., 1981); Ansel, Introduction to Pharmaceutical Dosage Forms 2nd Edition (1976); Remington's Pharmaceutical Sciences, 17th ed. (Mack Publishing Company, Easton, Pa., 1985); Advances in Pharmaceutical Sciences (David Ganderton, Trevor Jones, Eds., 1992); Advances in Pharmaceutical Sciences Vol. 7. (David Ganderton, Trevor Jones, James McGinity, Eds., 1995); Aqueous Polymeric Coatings for Pharmaceutical Dosage Forms (Drugs and the Pharmaceutical Sciences, Series 36 (James McGinity, Ed., 1989); Pharmaceutical Particulate Carriers: Therapeutic Applications: Drugs and the Pharmaceutical Sciences, Vol 61 (Alain Rolland, Ed., 1993); Drug Delivery to the Gastrointestinal Tract (Ellis Horwood Books in the Biological Sciences. Series in Pharmaceutical Technology; J. G. Hardy, S. S. Davis, Clive G. Wilson, Eds.); Modem Pharmaceutics Drugs and the Pharmaceutical Sciences, Vol 40 (Gilbert S. Banker, Christopher T. Rhodes, Eds.). All of the aforementioned publications are incorporated by reference herein. [0317] Tablets may contain suitable binders, lubricants, disintegrating agents, coloring agents, flavoring agents, flow-inducing agents, and melting agents. For instance, for oral administration in the dosage unit form of a tablet or capsule, the active drug component can be combined with an oral, non-toxic, pharmaceutically acceptable, inert carrier such as lactose, gelatin, agar, starch, sucrose, glucose, methyl cellulose, magnesium stearate, dicalcium phosphate, calcium sulfate, mannitol, sorbitol and the like. Suitable binders include starch, gelatin, natural sugars such as glucose or beta-lactose, corn sweeteners, natural and synthetic gums such as acacia, tragacanth, or sodium alginate, carboxymethylcellulose, polyethylene glycol, waxes, and the like. Lubricants used in these dosage forms include sodium oleate, sodium stearate, magnesium stearate, sodium benzoate, sodium acetate, sodium chloride, and the like. Disintegrators include, without limitation, starch, methyl cellulose, agar, bentonite, xanthan gum, and the like. [0318] The compounds used in the method of the present invention may also be administered in the form of liposome delivery systems, such as small unilamellar vesicles, large unilamellar vesicles, and multilamellar vesicles. Liposomes can be formed from a variety of phospholipids such as lecithin, sphingomyelin, proteolipids, protein-encapsulated vesicles or from cholesterol, stearylamine, or phosphatidylcholines. The compounds may be administered as components of tissue-targeted emulsions. [0319] The compounds used in the method of the present invention may also be coupled to soluble polymers as targetable drug carriers or as a prodrug. Such polymers include polyvinylpyrrolidone, pyran copolymer, polyhydroxylpropylmethacrylamide-phenol, polyhydroxyethylasparta-midephenol, or polyethyleneoxide-polylysine substituted with palmitoyl residues. Furthermore, the compounds may be coupled to a class of biodegradable polymers useful in achieving controlled release of a drug, for example, polylactic acid, polyglycolic acid, copolymers of polylactic and polyglycolic acid, polyepsilon caprolactone, polyhydroxy butyric acid, polyorthoesters, polyacetals, polydihydropyrans, polycyanoacylates, and crosslinked or amphipathic block copolymers of hydrogels. [0320] Gelatin capsules may contain the active ingredient compounds and powdered carriers, such as lactose, starch, cellulose derivatives, magnesium stearate, stearic acid, and the like. Similar diluents can be used to make compressed tablets. Both tablets and capsules can be manufactured as immediate release products or as sustained release products to provide for continuous release of medication over a period of hours. Compressed tablets can be sugar-coated or film-coated to mask any unpleasant taste and protect the tablet from the atmosphere, or enteric coated for selective disintegration in the gastrointestinal tract. [0321] For oral administration in liquid dosage form, the oral drug components are combined with any oral, non-toxic, pharmaceutically acceptable inert carrier such as ethanol, glycerol, water, and the like. Examples of suitable liquid dosage forms include solutions or suspensions in water, pharmaceutically acceptable fats and oils, alcohols or other organic solvents, including esters, emulsions, syrups or elixirs, suspensions, solutions and/or suspensions reconstituted from non-effervescent granules and effervescent preparations reconstituted from effervescent granules. Such liquid dosage forms may contain, for example, suitable solvents, preservatives, emulsifying agents, suspending agents, diluents, sweeteners, thickeners, and melting agents. [0322] Liquid dosage forms for oral administration can contain coloring and flavoring to increase patient acceptance. In general, water, asuitable oil, saline, aqueous dextrose (glucose), and related sugar solutions and glycols such as propylene glycol or polyethylene glycols are suitable carriers for parenteral solutions. Solutions for parenteral administration preferably contain a water soluble salt of the active ingredient, suitable stabilizing agents, and if necessary, buffer substances. Antioxidizing agents such as sodium bisulfite, sodium sulfite, or ascorbic acid, either alone or combined, are suitable stabilizing agents. Also used are citric acid and its salts and sodium EDTA. In addition, parenteral solutions can contain preservatives, such as benzalkonium chloride, methyl- or propyl-paraben, and chlorobutanol. Suitable pharmaceutical carriers are described in Remington's Pharmaceutical Sciences, Mack Publishing Company, a standard reference text in this field. [0323] The compounds used in the method of the present invention may also be administered in intranasal form via use of suitable intranasal vehicles, or via transdermal routes, using those forms of transdermal skin patches well known to those of ordinary skill in that art. To be administered in the form of a transdermal delivery system, the dosage administration will generally be continuous rather than intermittent throughout the dosage regimen. [0324] Parenteral and intravenous forms may also include minerals and other materials such as solutol and/or ethanol to make them compatible with the type of injection or delivery system chosen. [0325] The compounds and compositions of the present invention can be administered in oral dosage forms as tablets, capsules, pills, powders, granules, elixirs, tinctures, suspensions, syrups, and emulsions. The compounds may also be administered in intravenous (bolus or infusion), intraperitoneal, subcutaneous, or intramuscular form, or introduced directly, e.g. by topical administration, injection or other methods, to the afflicted area, such as a wound, including ulcers of the skin, all using dosage forms well known to those of ordinary skill in the pharmaceutical arts. [0326] Specific examples of pharmaceutically acceptable carriers and excipients that may be used to formulate oral dosage forms of the present invention are described in U.S. Pat. No. 3,903,297 to Robert, issued Sept.2, 1975. Techniques and compositions for making dosage forms useful in the present invention are described-in the following references: 7 Modern Pharmaceutics, Chapters 9 and 10 (Banker & Rhodes, Editors, 1979); Pharmaceutical Dosage Forms: Tablets (Lieberman et al., 1981); Ansel, Introduction to Pharmaceutical Dosage Forms 2nd Edition (1976); Remington's Pharmaceutical Sciences, 17th ed. (Mack Publishing Company, Easton, Pa., 1985); Advances in Pharmaceutical Sciences (David Ganderton, Trevor Jones, Eds., 1992); Advances in Pharmaceutical Sciences Vol 7. (David Ganderton, Trevor Jones, James McGinity, Eds., 1995); Aqueous Polymeric Coatings for Pharmaceutical Dosage Forms (Drugs and the Pharmaceutical Sciences, Series 36 (James McGinity, Ed., 1989); Pharmaceutical Particulate Carriers: Therapeutic Applications: Drugs and the Pharmaceutical Sciences, Vol 61 (Alain Rolland, Ed., 1993); Drug Delivery to the Gastrointestinal Tract (Ellis Horwood Books in the Biological Sciences. Series in Pharmaceutical Technology; J. G. Hardy, S. S. Davis, Clive G. Wilson, Eds.); Modem Pharmaceutics Drugs and the Pharmaceutical Sciences, Vol 40 (Gilbert S. Banker, Christopher T. Rhodes, Eds.). All of the aforementioned publications are incorporated by reference herein. [0327] The active ingredient can be administered orally in solid dosage forms, such as capsules, tablets, powders, and chewing gum; or in liquid dosage forms, such as elixirs, syrups, and suspensions, including, but not limited to, mouthwash and toothpaste. It can also be administered parentally, in sterile liquid dosage forms. [0328] Solid dosage forms, such as capsules and tablets, may be enteric-coated to prevent release of the active ingredient compounds before they reach the small intestine. Materials that may be used as enteric coatings include, but are not limited to, sugars, fatty acids, proteinaceous substances such as gelatin, waxes, shellac, cellulose acetate phthalate (CAP), methyl acrylate-methacrylic acid copolymers, cellulose acetate succinate, hydroxy propyl methyl cellulose phthalate, hydroxy propyl methyl cellulose acetate succinate (hypromellose acetate succinate), polyvinyl acetate phthalate (PVAP), and methyl methacrylate- methacrylic acid copolymers. [0329] The compounds and compositions of the invention can be coated onto stents for temporary or permanent implantation into the cardiovascular system of a subject. [0330] Variations on those general synthetic methods will be readily apparent to those of ordinary skill in the art and are deemed to be within the scope of the present invention. [0331] Each embodiment disclosed herein is contemplated as being applicable to each of the other disclosed embodiments. Thus, all combinations of the various elements described herein are within the scope of the invention. [0332] This invention will be better understood by reference to the Experimental Details which follow, but those skilled in the art will readily appreciate that the specific experiments detailed are only illustrative of the invention as described more fully in the claims which follow thereafter. General [0333] For the foregoing embodiments, each embodiment disclosed herein is contemplated as being applicable to each of the other disclosed embodiments. [0334] As used herein, all headings are simply for organization and are not intended to limit the disclosure in any manner. The content of any individual section may be equally applicable to all sections. All combinations of the various elements disclosed herein are within the scope of the invention. [0335] Additional objects, advantages, and novel features of the present invention will become apparent to one ordinarily skilled in the art upon examination of the following examples, which are not intended to be limiting. Additionally, each of the various embodiments and aspects of the present invention as delineated hereinabove and as claimed in the claims section below finds experimental support in the following examples. [0336] It is appreciated that certain features of the invention, which are, for clarity, described in the context of separate embodiments, may also be provided in combination in a single embodiment. Conversely, various features of the invention, which are, for brevity, described in the context of a single embodiment, may also be provided separately or in any suitable sub-combination or as suitable in any other described embodiment of the invention. Certain features described in the context of various embodiments are not to be considered essential features of those embodiments, unless the embodiment is inoperative without those elements. [0337] Examples are provided below to facilitate a more complete understanding of the invention. The following examples illustrate the exemplary modes of making and practicing the invention. However, the scope of the invention is not limited to specific embodiments disclosed in these Examples, which are for purposes of illustration only. EXAMPLES [0338] MATERIALS AND METHODS [0339] Reagents. Itraconazole (ITC, ≥98% Thin-Layer Chromatography) and RPMI 1640 (supplemented with L-glutamine, 2% (w/v) of glucose and without sodium bicarbonate were purchased from Sigma-Aldrich, USA. Brain Heart Infusion (BHI) was acquired from Oxoid, Brazil. [0340] Large scale synthesis of D13. Large scale synthesis (1,000 gr) of D13 (NED-59254-02) was commissioned and performed by New England Discovery Partner (NEDP) and a Material Safety Data Sheet (MSDS) was provided (see supplementary material). The compound was subjected to HPLC, LC-MS and 1H-NMR analysis (data not shown). When shipped, it was placed in dry ice. Upon arrival, the powder was stored at -20C. Under this storage condition, D13 is active at least for 3 years. Small scale synthesis of D13 (100 mg) was also performed. This small batch of D13 was then compared with the big batch of D13 (from NEDP) by HPLC, LC-MS, 1H-NMR, and by in vitro susceptibility analysis to make sure D13 from NEDP was identical in chemical structure and in activity to the D13 produced. [0341] The drugs, as powder, were encapsulated. The capsules were given to the owner who opened them on top of the cat wet food and the drug powder mixed in the food. This oral treatment was given daily and administered by the owner. [0342] Sporothrix strains and culture conditions. Sporothrix brasiliensis ATCC 5110/MYA4823 was obtained from the American Type Culture Collection (Manassas, VA 20108 USA). All strains were cultured in BHI broth at 37°C with orbital agitation (150 rpm) for 3 days and then cryopreserved at -80°C in BHI broth with 20% glycerol. [0343] Clinical isolates were obtained as follow. Exudate from a cutaneous or mucosal lesion were collected by impression smear on a glass slide for cytological examination. The slide was airdried, stained using the Quick Panoptic method (Instant Prov Kit; Newprov, Pinhais, Brazil), and then analyzed under an optical microscope at x1,000 magnification (Pereira SA, 2011) of the presence of yeasts-like cells suggestive of Sporothrix spp. Exudates from the same lesion were also seeded on to Sabouraud dextrose agar and Mycobiotic agar (Difco) plates, incubated at 25oC for up to 4 weeks for fungal growth. Suspected isolates were sub-cultured on potato dextrose agar medium (Difco) at 25oC for macroscopic and microscopic morphological studies. Dimorphism was demonstrated by conversion to the yeast-like form on brain heart infusion agar medium (Difco) at 37oC. Sporothrix spp. was isolated in mycological culture and S. brasiliensis was identified using the molecular tool T3B PCR fingerprinting (de Oliveira MM, 2012). Sporothrix brasiliensis was identified in 8 out of 10 cats. From two cats, it was not able to identify the fungal species because the samples were highly contaminated by bacteria. [0344] Anti-Sporothrix tests. Antifungal assays were conducted according to the Clinical and Laboratory Standards Institute (CLSI) M27-A3 protocol, with necessary adjustments for cell suspension. Yeasts were cultured in BHI broth at 37 °C/150 rpm for 3 days. Subsequently, the cells were washed in phosphate-buffered saline (PBS) and adjusted to a concentration of 5 x 105 yeasts/mL in RPMI 1640. D13 and ITC were diluted in RPMI ranging from 16 to 0.06 µg/mL, and 100 µL volumes were added to 96-well plates. Then, 100 µL of the yeast suspension was added to each well. Cells were incubated at 37 °C for 3 days. RPMI 1640 was used as control. Fungal growth was quantified using a BioTeK ELx808 microplate reader (Netherlands) through spectrophotometric measurements taken at a wavelength of 530 nm. The Minimal Inhibitory Concentration (MIC50 and MIC90) was determined as the lowest concentrations of D13 that reduced microbial growth by 50% and 90%, respectively, compared to the control conditions. [0345] Combination drugs. The experimental procedure was conducted using 96-well plates, following the checkerboard test outlined in previous studies (Artunduaga Bonilla JJ, 2021). Succinctly, serial two-fold dilutions of each drug (D13 and ITC) were prepared in RPMI. Yeasts (5 x 105 cells/mL) was added into the 96-well microplates in 100 µL volumes, after which the plates were incubated at 37 °C for 3 days. The optical density (OD) was measured at 530 nm using a Biotek ELx808 microplate reader. The fractional inhibitory concentration index (FICi) was calculated as (MIC combined/MIC drug A alone) + (MIC combined/MIC drug B alone). Synergistic drug interactions were identified as synergistic when the FICi was less than 0.5, indifferent interactions when the FICi was between 0.5 and 4.0, and antagonistic interactions when the FICi exceeded 4 ( Odds FC. 2003 and Zhao YJ, 2019,). Alternatively, the Bliss synergistic scores was calculated using the SynergyFinder tool (synergyfinder.fimm.fi/synergy) (Ianevski A, 2017). A score below -10 was interpreted as an antagonistic interaction, while scores ranging from -10 to 10 indicated an additive interaction. Scores greater than 10 denoted a synergistic interaction between the two drugs. [0346] Ex vivo Biofilm. The anti-biofilm effect of D13 was visualized by scanning electron microscopy (SEM) and quantified by CFU determination. Claws collected from healthy cats were previously sterilized by immersion in 90% ethanol / 3h, washed 3x with PBS and transferred to tubes with 200 μL of RPMI / 1% PenStrep containing 1 x106 cells/mL of S. brasiliensis ATCC 5110. Cells were incubated at 37 °C for 3 days. Afterwards, claws were washed 3x with PBS in order to remove the cells not adhered to the biofilm. The biofilm formation was checked using an optical microscope (Olympus CX-31) and then 200 μL of D13 (10 μg/mL) or ITC (10 μg/mL) previously diluted in RPMI 1640 was added. After 3 days of incubation at 37 °C, the claws were washed with PBS and the cells fixed with 2.5% glutaraldehyde and 4% formaldehyde (in 0.1 M cacodylate buffer, pH 7.2) for 1 hour at room temperature. The claws were then washed with cacodylate buffer and dehydrated using ethanol gradient. Samples were coated with a 20 nm layer of gold and visualized using a scanning electron microscope (ZEISS EVO MA 10, Germany) operated at 10 Kv. [0347] To determine the CFU, claws from each treatment and control were collected and weighed prior to fixation stage. The claws were suspended in 200 μL of RPMI 1640/1% PenStrep, subjected to sonication for 1 minute, and vortexed. Aliquots of 20 μL were plated onto BHI plates. Following an incubation of 2-3 days at 37°C, the CFU count was performed. Two independent experiments were conducted. [0348] Ethics statement. Procedures performed on patients were approved by the Animal Ethics Committee/Fiocruz (LW-13/19). [0349] Statistical analysis. Data were analyzed using GraphPad Prism 7 software, employing ordinary one-way ANOVA followed by Tukey's multiple comparisons test. Statistical significance was determined by a p-value of less than 0.05. [0350] Cases Histories. Based on the compelling results obtained in the previous study (21-24), as well as on the promising in vitro data observed in this study, the inventors submitted to the Animal Ethics Committee at Fiocruz a pilot clinical study using D13 in cats affected with sporotrichosis. The study was approved as a “compassionate drug use”, and the inventors could add D13 to the SOC only when the SOC alone failed. The study enrolled a total of 10 cats. All cases included were confirmed as sporotrichosis by isolation of Sporothrix spp. in culture from skin ulcers, and S. brasiliensis was identified in two cases (Cases 1 and 3). Example 1 [0351] Case 1. The patient was a 4-year-old neutered male crossbred cat, weighing 1.6 kg. The cat was being already treated with ITC (100 mg/cat/day) for eight months before referral. Clinical examination revealed multiple ulcerated cutaneous lesions and nodules distributed over the head, nasal region, hind and forelimbs, swelling of both nostrils, dyspnea and loss of the normal nasal planum architecture. The cat was in serious overall condition and presented hyporexia. Due to poor clinical response, KI (2.5 mg/kg/day) was initiated, in combination with ITC in a lower dose (25 mg/cat/day). Oral sylimarin (30 mg/kg/day) was also prescribed. After one month, the dose of ITC was increased up to 50 mg/cat/day as the animal gained weight, while KI and sylimarin were maintained at the same dose. Nevertheless, the cutaneous and nasal mucosal lesions persisted after three months of ITC and KI therapy. Thus, D13 (20 mg/kg/day) was added to the ITC (50 mg/cat/day) and KI (2.5 mg/kg/day) therapeutic regimen. Follow-up clinical examinations and analysis of hematological and biochemical parameters, such as urea, creatinine, alanine transaminase (ALT) and aspartate aminotransferase (AST), were performed monthly by the reference center for fungal diseases in animals in Rio de Janeiro. After three months of treatment, hyporexia had been noted. After 14 days, mild elevations in ALT (ALT 130.4 U/L; Reference Interval 6-83 U/L) and AST (AST 93.9 U/L; Reference Interval 10-80 U/L) were noted. These laboratory abnormalities in ALT and AST levels were transient, and returned to the pre-treatment levels when measured at day 28 of treatment. The clinical cure occurred after eight months of combination therapy (Figure 3). Example 2 [0352] Case 2. The patient was a 1-year-old non-neutered male crossbred cat, weighing 4.1 kg, examined at a reference center for fungal diseases in animals in Rio de Janeiro, Brazil. The cat presented ulcerated cutaneous lesions on the nasal bridge, lips, left ear, and right forelimb. Swelling of both nostrils, sneezing, nasal discharge and enlarged mandibular lymph nodes were also observed. Its overall condition was good. Upon identification of S. brasiliensis, the cat was treated with ITC (100 mg/cat/day). After 12 weeks of therapy, hyporexia was noted, and no improvement of any cutaneous lesion was observed. In addition, during ITC treatment three new lesions appeared: one on the nasal bridge near the other lesion and the others two on the right pinna. This indicated a therapeutic failure. Thus, D13 (20 mg/kg/day) was added to ITC (100 mg/cat/day). During the 12 weeks of combination therapy, no clinical adverse reactions were observed. However, mild to moderate elevations in ALT between days 30 (ALT 103 U/L; Reference Interval 10-83 U/L) and day 60 (ALT 266 U/L; Reference Interval 10-83 U/L), were noted. This abnormality in ALT level was transient, and returned to the pre-treatment level when measured at day 90 of treatment (25). The clinical cure (complete healing of the skin lesions) occurred after 6 months of therapy, when the cat was discharged (Figure 4). Example3 [0353] Case 3. The patient was a 1-year-old non-neutered male crossbred cat, weighing 4 kg. Clinical examination revealed multiple ulcerated cutaneous lesions on the nasal bridge, lips, left ear, and right forelimb. Swelling of both nostrils, sneezing, nasal discharge and enlarged mandibular lymph nodes were also observed. The cat was in good overall condition. ITC (100 mg/cat/day) was prescribed and after three months of therapy, hyporexia was noted. Also, ITC failed to resolve the cutaneous lesion on the nasal bridge. In addition, during ITC treatment three new lesions appeared: one on the nasal bridge near the other lesion and the others two on the right pinna. Thus, D13 (20 mg/kg/day) was added to ITC (100 mg/cat/day). During the three months of combination therapy, no clinical adverse reactions or abnormal laboratory findings were observed. After 30 days, mild to moderate elevations in ALT (ALT 103 U/L; Reference Interval 10-83 U/L) and at day 60 (ALT 266 U/L; Reference Interval 10-83 U/L), were noted. This abnormality in ALT level was transient, and returned to the pre-treatment level when measured at day 90 of treatment. The clinical cure occurred after three months of combination therapy, when the cat was discharged (Figure 5). Example 4 [0354] Case 4. The patient was a 2-year-old castrated male crossbred cat, weighing 4.9 kg, and it was examined at the same reference center as above. The cat presented ulcerated cutaneous lesions on the nasal bridge, left mandible, dorsal thoracic region and one nodule in the medial corner of the left eye. An enlarged right mandibular lymph node was the only extracutaneous sign observed. The cat`s overall condition was good. ITC (100 mg/cat/day) was prescribed orally once a day. After 30 days of ITC treatment, the cat was evaluated. Although there were no clinical adverse reactions or abnormal laboratory findings, the cutaneous lesions persisted, indicating therapeutic failure. Thus, D13 at 20 mg/kg/day was added to ITC (100 mg/cat/day). The clinical cure occurred after 3 months of therapy, when the cat was discharged (Figure 6). No clinical adverse reactions or abnormal laboratory findings were noted during the combination therapy. Example 5 [0355] Case 5. The patient was a 3-year-old non-neutered male crossbred cat, weighing 4.2 kg, and it was examined at the same reference center as above. The cat presented multiple ulcerated cutaneous lesions on the nasal bridge, head, right forelimb and tail. Swelling of both nostrils, sneezing, nasal discharge and enlarged mandibular lymph nodes were also observed. The cat was in good overall condition. ITC (100 mg/cat/day) was prescribed and after four weeks of therapy, it was observed the persistence of the cutaneous lesions and the respiratory signs. Thus, D13 (20 mg/kg/day) was added to ITC (100 mg/cat/day). During the four weeks of combination therapy, weight loss and mild elevation in ALT (ALT 112 U/L; Reference Interval 10-83 U/L) were observed. There was an improvement (Figure 7) in the cutaneous lesions, but the respiratory signs persisted. [0356] Among the other five included cats, all of them had cutaneous lesions and four had cutaneous and mucosal lesions. Respiratory signs (nasal discharge, sneezing and dyspnea) were observed in four cats. Three cats had serious overall condition. Unfortunately, three cats were lost during follow-up (treatment abandonment), whereas the other two cats had a progressive evolution of sporotrichosis. Both died after 8 weeks and one of them died despite 72 weeks of treatment. No ALT elevation was observed in these 5 cats during the observation period. Table A. Summary of dosage in Case 1- Case 5* Case 1 Case 2 Case 3 Case 4 Case 5
Figure imgf000064_0001
Dose Calculations and Examples [0357] The dose by factor method is an empirical approach and use the no observed adverse effect levels (NOAEL) of drug from preclinical toxicological studies to estimate human equivalent dose (HED). Here, the dose selection is based on minimum risk of toxicity, instead of choosing one with minimum pharmacologic activity in humans. In similar drug approach, the existing pharmacokinetics data for another drug of the same pharmacological category may be used. On the other hand, pharmacokinetically guided approach utilizes the drug activity instead of scaling of dose among species. In case of comparative approach, different methods are utilized to determine initial dose, and the data are compared and optimize to get an initial dose. (Nair AB, Jacob S, 2016) [0358] The dose by factor method applies an exponent for body surface area (0.67), which account for difference in metabolic rate, to convert doses between animals and humans. Thus, human equivalent dose (HED) is determined by the equation (Nair AB, Jacob S, 2016): HED (mg / kg = Animal NOAEL mg/kg) ×(Weightanimal [kg]/Weighthuman [kg])(1–0.67) Eq. (1) [0359] For example, for a newly developed drug molecule, the no observed adverse effect levels (NOAEL) value in rat weighing approximately 150 g is 18 mg/kg. To calculate the starting dose for human studies, use Equation 1. (Nair AB, Jacob S, 2016) HED (mg / kg = 18 × (0.15 / 60)(0.33)= 2.5 mg / kg [0360] Thus, for a 60 kg human, the dose is 150 mg. This HED value is further divided by a factor value of 10; thus, the initial dose in entry into man studies is 15 mg. (Nair AB, Jacob S, 2016) [0361] Dose is equally related to body weight although it is not the lone factor which influences the scaling for dose calculation. The correction factor (Km) is estimated by dividing the average body weight (kg) of species to its body surface area (m2). For example, the average human body weight is 60 kg, and the body surface area is 1.62 m2. Therefore, the Km factor for human is calculated by dividing 60 by 1.62, which is 37 (Table 1 in Nair AB, Jacob S, 2016). The Km factor values of various animal species (Table 1 in Nair AB, Jacob S, 2016) is used to estimate the HED as HED (mg / kg) = Animal does (mg / kg) × (Animal Km/ Human Km) Eq. (2). [0362] As the Km factor for each species is constant, the Km ratio is used to simplify calculations. Hence, Equation 2 (Nair AB, Jacob S, 2016) is modified as: HED (mg / kg) = Animal does (mg / kg) × Kmratio Eq. (3) . [0363] The Km ratio values provided in Table 1 (Nair AB, Jacob S, 2016) is easily obtained by dividing human Km factor by animal Km factor or vice versa. For instance, the Km ratio values for rat is 6.2 and 0.162, obtained by dividing 37 (human Km factor) by 6 (animal Km factor) and vice versa, respectively. Thus, usually to obtain the HED values (mg/kg), one can either divide or multiply the animal dose (mg/kg) by the Km ratio provided in Table 1. For example, for a particular drug, the NOAEL in rats is 50 mg/kg. Using Equation 3, HED is calculated either by multiplying or dividing the animal dose with the Km ratio values given in Table 1 (Nair AB, Jacob S, 2016). Accordingly, divide the rat dose (50 mg/kg) by 6.2 or multiply by 0.162, the HED is –8.1 mg/kg. (Nair AB, Jacob S, 2016) [0364] However, it must be borne in mind that the km factor varies across animal species and increases proportional to W2/3 within a species as body weight increases. For example, the km value in rats varies from 5.2 (100 g rat), 6 (150 g rat), and 7 (250 g rat). Therefore, calculation of HED for a drug of NOAEL in rats is 50 mg/kg with an average weight of 250 g is as below: ^ HED(mg/kg)=50(mg/kg)×^ ଷ^^ =9.5 mg/kg in humans. km factor value of 6 for rats with average weight of 150 g Table 1 (Nair AB,
Figure imgf000066_0001
Jacob S, 2016), the dose varies accordingly as seen below: ^ HED(mg/kg)=50(mg/kg)× ^ ଷ^^ =8.1 mg/kg in humans. g/m2) of dose of animals or human is carried out using the Km
Figure imgf000066_0002
factor Table 1 (Nair AB, Jacob S, 2016) as: mg / m2 = Km × mg / kg Eq. (4). [0367] However, conversion between species based on mg/m2 is not supported for drugs administered by topical, nasal, subcutaneous, or intramuscular routes as well as proteins administered parenterally with molecular weight >100,000 Daltons. [0368] Similar to the HED estimation, the animal equivalent dose (AED) can also be calculated on the basis of body surface area by either dividing or multiplying the human dose (mg/kg) by the Km ratio provided in Table 2 (Nair AB, Jacob S, 2016). AED is calculated by minor modification of Equation 3 as: AED (mg / kg) = Human does (mg / kg) × Km ratio Eq. (5) [0369] For example, if the maximum dose of a particular drug in human is 10 mg/kg, the AED is calculated by multiplying the HED by 6.2 or dividing by 0.162; AED is 62 mg/kg. [0370] For parenteral administration, HED conversion (mg/kg) is also based on body surface area normalization. The conversion can be made by dividing the NOAEL in appropriate species by the conversion factor. Guidelines for maximum injection volumes, by species, site location, and gauge size are summarized in Table 3 (Nair AB, Jacob S, 2016). Injection volume of parenteral formulation is calculated by following equation. [0371] Suppose the concentration of the formulation (say nanoparticles) is 10 mg/mL, its AED is 62 mg/kg, the rat weight is 250 g and is administered through intraperitoneal route, and then the injection volume is calculated as ^.ଶହ^^^^௫^ଶ^^^ ion volume (ml) = ೖ ^ Inject ^ ^^ ൌ 1.55 ^^^^.
Figure imgf000066_0003
[0372] The injection volume observed here (1.55 mL) is well below the maximum injection volume (5–10 mL) for rat through intraperitoneal route and the injection site is lower left quadrant. Results [0373] Enrollment. The present inventions provide a pilot clinical study. The inventors did not inflict sporotrichosis to cats. Cats affected with sporotrichosis were taken by their owner to the Laboratory of Clinical Research on Dermatozoonoses in Domestic Animals at Oswaldo Cruz Foundation (Fiocruz), in Rio de Janeiro, Brazil, to seek diagnosis and treatment. Upon confirming the diagnosis of sporotrichosis, cats were treated using the standard of care (SOC). They mainly received itraconazole (ITC) alone or in combination with potassium iodide (KI) (Table 1). One cat (Case 2) also received terbinafine (TRB). If the cat did not respond to the SOC (see below), a possibility to add D13 was proposed to the owner of the cat as “compassionate drug use”. If the owner agreed, D13 was added to the therapeutic protocol. The Animal Ethics Committee at Fiocruz granted approval for D13 exclusively to be added to the SOC, when the SOC failed. Failure was defined as no improvement of the skin lesion(s) or/and appearance of new skin lesion(s) after at least 4 weeks of treatment of SOC. Because the animal protocol was approved for using D13 as adjuvant therapy (to be added to the SOC), it was not possible to include a control group taking D13 in monotherapy because the Animal Ethics Committee did not approve the use D13 as a monotherapy. In addition, because the cats that did not respond to the SOC were very sick, it was not humanly ethical to use some of these animals continuing taking the SOC (without D13) as a control group.
[0374] Table 1. Part A. Clinical information and summary table of the 10 cases of sporotrichosis treated with the experimental drug acylhydrazone D13 in various combinations with the standard of care. Case Sex Age W (Kg) Con Lesions Resp signs is
Figure imgf000068_0001
Table 1. Part B. Clinical information and summary table of the 10 cases of sporotrichosis treated with the experimental drug acylhydrazone D13 in various combinations with the standard of care. Case Prior treatment New ther prot Treat time LAB Out the
Figure imgf000069_0001
identification, these clinical samples were lost and no longer available for MIC and synergistic testing. All other clinical isolates were available (see Table 1 and Table 2). M, male; F, female; m, months; Kg, kilogram; Con, health condition at the initiation of the standard of care (SOC); S, serious; G, good; Cut, cutaneous; muc, mucosal; Resp, respiratory; Dys, dyspnea; Dis, discharge; Ther, therapeutic; New ther prot, New therapeutic protocol; ITC, itraconazole; TRB, terbinafine; KI, potassium iodine; Treat, treatment; w, weeks; LAB; laboratory testing (blood work); ALT, alanine aminotransferase; Out, outcome; Imp, improved, Aband, abandonment. [0375] In vitro antifungal activity. Upon clinical suspicion of sporotrichosis, swab samples were collected and placed onto Sabouraud-dextrose agar. Plates were incubated for up to 5 days at room temperature (~25C), fungal growth was monitored and fungal identification was performed by standard methods. Fungal isolates were tested for in vitro susceptibility against ITC and D13. From the 10 cats, only 8 clinical isolates were obtained. Two isolates (from Case 4 and from Case 10) were highly contaminated by bacteria and were lost. [0376] D13 displayed high efficacy in inhibiting the growth of all tested isolates (Table 2), with MIC50 ranging from 0.12 μg/ml to 4 μg/ml. As for ITC, only two clinical isolates (Case 1 and Case 2) showed low MIC50 of 0.5 μg/ml. All other isolates had higher MIC50 ranging from 4 μg/ml to 32 μg/ml (Table 2), suggesting that these isolates were, in general, more resistant to ITC. [0377] Table 2. Anti-Sporothrix activity of D13 and Itraconazole (Itra), against clinical isolates recovered from 8 cats. Clinical isolates D13 (µg/ml) ITC (µg/ml)
Figure imgf000070_0001
the growth observed in absence of drug, respectively. ITC, Itraconazole. Tests were performed at least twice. [0378] Synergistic studies. Because D13 was intended to be administered solely as a combination therapy, it was assessed the synergist effect of D13 when combined with ITC. Synergistic activity was examined using the checkerboard method and the Bliss method. Using the checkerboard assay, the combination of D13 and ITC resulted in synergistic activity or in no interaction (Table 3). Using the Bliss method, the combination of D13 and ITC resulted to have additive activity to all tested clinical isolates (Table 3). [0379] Table 3. Synergistic activity illustrated as fractional inhibitory concentration (FIC) index of Itraconazole combined with D13 against Sporothrix clinical isolated from cats. Checkerboard Method Bliss Method Clinical isolates Ʃ FICi Activity Score Activity Cat 1 0.65 ± 0.12 Indifferent 1.72 Additive Cat 2 0.52 ± 0.01 Indifferent -7.98 Additive Cat 3 0.98 ± 0.04 Indifferent -7.43 Additive Cat 5 0.66 ± 0.10 Indifferent -9.12 Additive Cat 6 0.68 ± 0.16 Indifferent -5.98 Additive Cat 7 0.32 ± 0.07 Synergistic -8.56 Additive Cat 8 1.01 ± 0.02 Indifferent -0.38 Additive Cat 9 0.29 ± 0.03 Synergistic -8.79 Additive [0380] For the checkerboard method: synergistic drug interactions were identified as synergistic when the FICi was less than 0.5, no interaction or indifferent when the FICi was between 0.5 and 4.0, and antagonistic interactions when the FICi exceeded 4.0. For the Bliss method, scores greater than 10 denote a synergistic interaction, scores ranging from -10 to 10 indicate an additive interaction, and scores below - 10 denote an antagonistic interaction. Data represent geometric means ± standard deviations of at least three independent experiments. [0381] D13 is effective against S. brasiliensis biofilm. Recognizing the crucial role of biofilm as an important reservoir of fungal cells, which become more resistant to antifungals, the effect of D13 on biofilm formation was examined by Sporothrix on cat's claw. This is particularly important because this fungus is frequently transmitted from cats to humans by scratches. If Sporothrix forms biofilm in the cat’s claw its potential for transmission is significantly increased. [0382] For this experiment, a reference clinical strain Sporothrix brasiliensis ATCC 5110/MYA4823 was used and a clinical isolate 4 - 17522, obtained from worse case (case 1). The untreated biofilm exhibited a surface densely populated mostly composed of intact yeasts, as showed in Figure 1. In contrast, the biofilm treated with D13 showed a substantial reduction in fungal growth, as clearly evidenced by scanning electron microcopy (SEM) images captured at different magnifications (Figure 1). Analysis of the colony forming units (CFU) of the tissue confirmed a significant decrease in fungal load in the D13- treated biofilm compared to the control group (p < 0.0001) (Figure 2). The effect of D13 was similar against the referent strain and against the clinical isolate. [0383] Most importantly, treatment with D13 exhibited a pronounced reduction in fungal load when compared to the biofilm treated with ITC (p < 0.05) (Figure 1 and Figure 2), providing further confirmation that D13 appears to be more effective than ITC in inhibiting biofilm formation and growth of S. brasiliensis.
REFERENCES 1. Poester VR, Mattei AS, Madrid IM, Pereira JTB, Klafke GB, Sanchotene KO, Brandolt TM, Xavier MO. 2018. Sporotrichosis in Southern Brazil, towards an epidemic? Zoonoses Public Health 65:815- 821. 2. Della Terra PP, Rodrigues AM, Fernandes GF, Nishikaku AS, Burger E, de Camargo ZP. 2017. Exploring virulence and immunogenicity in the emerging pathogen Sporothrix brasiliensis. PLoS Negl Trop Dis 11:e0005903. 3. de Carvalho JA, Beale MA, Hagen F, Fisher MC, Kano R, Bonifaz A, Toriello C, Negroni R, Rego RSM, Gremiao IDF, Pereira SA, de Camargo ZP, Rodrigues AM. 2021. Trends in the molecular epidemiology and population genetics of emerging Sporothrix species. Stud Mycol 100:100129. 4. Boechat JS, Oliveira MME, Gremiao IDF, Almeida-Paes R, Machado ACS, Zancope-Oliveira RM, Oliveira RVC, Morgado DS, Correa ML, Figueiredo ABF, Menezes RC, Pereira SA.2022. Sporothrix brasiliensis and Feline Sporotrichosis in the Metropolitan Region of Rio de Janeiro, Brazil (1998- 2018). J Fungi (Basel) 8. 5. Han HS, Kano R.2021. Feline sporotrichosis in Asia. Braz J Microbiol 52:125-134. 6. Gremiao ID, Menezes RC, Schubach TM, Figueiredo AB, Cavalcanti MC, Pereira SA. 2015. Feline sporotrichosis: epidemiological and clinical aspects. Med Mycol 53:15-21. 7. Miranda LHM, Santiago MA, Schubach TMP, Morgado FN, Pereira SA, Oliveira RVC, Conceicao- Silva F. 2016. Severe feline sporotrichosis associated with an increased population of CD8low cells and a decrease in CD4(+) cells. Med Mycol 54:29-39. 8. de Souza EW, Borba CM, Pereira SA, Gremiao IDF, Langohr IM, Oliveira MME, de Oliveira RVC, da Cunha CR, Zancope-Oliveira RM, de Miranda LHM, Menezes RC.2018. Clinical features, fungal load, coinfections, histological skin changes, and itraconazole treatment response of cats with sporotrichosis caused by Sporothrix brasiliensis. Sci Rep 8:9074. 9. Schubach TM, Schubach A, Okamoto T, Barros MB, Figueiredo FB, Cuzzi T, Fialho-Monteiro PC, Reis RS, Perez MA, Wanke B. 2004. Evaluation of an epidemic of sporotrichosis in cats: 347 cases (1998-2001). J Am Vet Med Assoc 224:1623-9. 10. de Miranda LHM, Silva JN, Gremiao IDF, Menezes RC, Almeida-Paes R, Dos Reis EG, de Oliveira RVC, de Araujo D, Ferreiro L, Pereira SA. 2018. Monitoring Fungal Burden and Viability of Sporothrix spp. in Skin Lesions of Cats for Predicting Antifungal Treatment Response. J Fungi (Basel) 4. 11. da Rocha R, Schubach TMP, Pereira SA, Dos Reis EG, Carvalho BW, Gremiao IDF.2018. Refractory feline sporotrichosis treated with itraconazole combined with potassium iodide. J Small Anim Pract 59:720-721. 12. Nakasu CCT, Waller SB, Ripoll MK, Ferreira MRA, Conceicao FR, Gomes ADR, Osorio LDG, de Faria RO, Cleff MB. 2021. Feline sporotrichosis: a case series of itraconazole-resistant Sporothrix brasiliensis infection. Braz J Microbiol 52:163-171. 13. Gremiao IDF, Miranda LHM, Pereira-Oliveira GR, Menezes RC, Machado ACS, Rodrigues AM, Pereira SA.2022. Advances and challenges in the management of feline sporotrichosis. Rev Iberoam Micol 39:61-67. 14. Silva FS, Cunha SCS, Moraes VA, Leite JS, Ferreira AMR.2022. Refractory feline sporotrichosis: a comparative analysis on the clinical, histopathological, and cytopathological aspects. Brazilian Journal of Veterinary Research 42:e06923 15. Poester VR, Basso RP, Stevens DA, Munhoz LS, de Souza Rabello VB, Almeida-Paes R, Zancope- Oliveira RM, Zanchi M, Benelli JL, Xavier MO. 2022. Treatment of Human Sporotrichosis Caused by Sporothrix brasiliensis. J Fungi (Basel) 8. 16. Gremiao IDF, Martins da Silva da Rocha E, Montenegro H, Carneiro AJB, Xavier MO, de Farias MR, Monti F, Mansho W, de Macedo Assuncao Pereira RH, Pereira SA, Lopes-Bezerra LM. 2021. Guideline for the management of feline sporotrichosis caused by Sporothrix brasiliensis and literature revision. Braz J Microbiol 52:107-124. 17. Xavier MO, Poester VR, Trapaga MR, Stevens DA. 2023. Sporothrix brasiliensis: Epidemiology, Therapy, and Recent Developments. J Fungi (Basel) 9. 18. Poester VR, Munhoz LS, Stevens DA, Melo AM, Trapaga MR, Flores MM, Larwood DJ, Xavier MO. 2023. Nikkomycin Z for the treatment of experimental sporotrichosis caused by Sporothrix brasiliensis. Mycoses 66:898-905. 19. Como JA, Dismukes WE. 1994. Oral azole drugs as systemic antifungal therapy. N Engl J Med 330:263-72. 20. Johnson MD, Perfect JR.2010. Use of Antifungal Combination Therapy: Agents, Order, and Timing. Curr Fungal Infect Rep 4:87-95. 21. Mor V, Rella A, Farnoud AM, Singh A, Munshi M, Bryan A, Naseem S, Konopka JB, Ojima I, Bullesbach E, Ashbaugh A, Linke MJ, Cushion M, Collins M, Ananthula HK, Sallans L, Desai PB, Wiederhold NP, Fothergill AW, Kirkpatrick WR, Patterson T, Wong LH, Sinha S, Giaever G, Nislow C, Flaherty P, Pan X, Cesar GV, de Melo Tavares P, Frases S, Miranda K, Rodrigues ML, Luberto C, Nimrichter L, Del Poeta M.2015. Identification of a New Class of Antifungals Targeting the Synthesis of Fungal Sphingolipids. MBio 6:e00647. 22. Lazzarini C, Haranahalli K, Rieger R, Ananthula HK, Desai PB, Ashbaugh A, Linke MJ, Cushion M, Ruzsicska B, Haley J, Ojima I, Del Poeta M.2018. Acylhydrazones as antifungal agents targeting the synthesis of fungal sphingolipids. Antimicrob Agents Chemother 62:e00156-18. 23. Artunduaga Bonilla JJ, Honorato L, Haranahalli K, Gremiao IDF, Pereira SA, Guimaraes A, Baptista ARS, de MTP, Rodrigues ML, Miranda K, Ojima I, Del Poeta M, Nimrichter L. 2021. Antifungal activity of Acylhydrazone derivatives against Sporothrix spp. Antimicrob Agents Chemother doi:10.1128/AAC.02593-20. 24. Haranahalli K, Lazzarini C, Sun Y, Zambito J, Pathiranage S, McCarthy JB, Mallamo J, Del Poeta M, Ojima I. 2019. SAR Studies on Aromatic Acylhydrazone-Based Inhibitors of Fungal Sphingolipid Synthesis as Next-Generation Antifungal Agents. J Med Chem 62:e00156-18. 25. Center SA. 2007. Interpretation of liver enzymes. Vet Clin North Am Small Anim Pract 37:297-333, vii. 26. de Souza CP, Lucas R, Ramadinha RH, Pires TB.2016. Cryosurgery in association with itraconazole for the treatment of feline sporotrichosis. J Feline Med Surg 18:137-43. 27. Lazzarini C, Haranahalli K, McCarthy JB, Mallamo J, Ojima I, Del Poeta M. 2020. Preclinical evaluation of acylhydrazone SB-AF-1002 as a novel broad-spectrum antifungal agent. Antimicrob Agents Chemother 64:e00946-20. 28. Reis EG, Schubach TM, Pereira SA, Silva JN, Carvalho BW, Quintana MS, Gremiao ID. 2016. Association of itraconazole and potassium iodide in the treatment of feline sporotrichosis: a prospective study. Med Mycol 54:684-90. 29. Pereira SA, Menezes RC, Gremiao ID, Silva JN, Honse Cde O, Figueiredo FB, da Silva DT, Kitada AA, dos Reis EG, Schubach TM. 2011. Sensitivity of cytopathological examination in the diagnosis of feline sporotrichosis. J Feline Med Surg 13:220-3. 30. de Oliveira MM, Sampaio P, Almeida-Paes R, Pais C, Gutierrez-Galhardo MC, Zancope-Oliveira RM. 2012. Rapid identification of Sporothrix species by T3B fingerprinting. J Clin Microbiol 50:2159-62. 31. Odds FC. 2003. Synergy, antagonism, and what the chequerboard puts between them. J Antimicrob Chemother 52:1. 32. Zhao YJ, Liu WD, Shen YN, Li DM, Zhu KJ, Zhang H. 2019. The efflux pump inhibitor tetrandrine exhibits synergism with fluconazole or voriconazole against Candida parapsilosis. Mol Biol Rep 46:5867-5874. 33. Ianevski A, He L, Aittokallio T, Tang J. 2017. SynergyFinder: a web application for analyzing drug combination dose-response matrix data. Bioinformatics 33:2413-2415. 34. Nair AB, Jacob S. A simple practice guide for dose conversion between animals and human. J Basic Clin Pharm. 2016 Mar;7(2):27-31. doi: 10.4103/0976-0105.177703. PMID: 27057123; PMCID: PMC4804402.

Claims

CLAIMS What is claimed is: 1. A method of treating fungal infection in a subject, wherein the method comprises administering to the subject a combination comprising an amount of itraconazole (ITC) and an amount of a compound, or a pharmaceutically acceptable salt thereof, wherein the compound has the following structure: ,
Figure imgf000076_0001
R1 is -H, alkyl, alkenyl, or alkynyl; R2 is -H, alkyl, alkenyl, or alkynyl; R9, R10, R11, and R12 are each independently, H, halogen, -CN, -CF3, -OCF3, -NO2, alkyl, alkenyl, alkynyl, aryl, heteroaryl, cycloalkyl, heterocycloalkyl, -OH, -OAc, -OR13, -COR13, -SH, -SR13, - SO2R13, -SO2NR14R15, -NH2, -NHR13, -NR14R15, -NHCOR12, or -CONR14R15; or R9 and R10 are each independently -H, halogen, C1-C6 alkyl, -OH, -O-(C1-C6 alkyl), -CHF2, -CF3, -OCHF2 or -OCF3, and R11 and R12 combine to form a fused aryl or fused heteroaryl, which are each unsubstituted or substituted, or R9 and R12 are each independently -H, halogen, C1-C6 alkyl, -OH, -O-(C1-C6 alkyl), -CHF2, -CF3, -OCHF2 or -OCF3, and R10 and R11 combine to form a fused aryl or fused heteroaryl, which are each unsubstituted or substitute, or R11 and R12 are each independently -H, halogen, C1-C6 alkyl, -OH, -O-(C1-C6 alkyl), -CHF2, -CF3, -OCHF2 or -OCF3, and R3 and R4 combine to form a fused aryl or fused heteroaryl, which are each unsubstituted or substituted; wherein each occurrence of R13 is independently alkyl, alkenyl, alkynyl, aryl, or heteroaryl; wherein each occurrence of R14 and R15 is independently –H, alkyl, alkenyl, alkynyl, aryl, or heteroaryl; wherein when R14 is methyl, R15 is not methyl; and wherein at least one of R9, R10, R11, and R12 is not H; so as to thereby treat the subject against fungal infection.
2. The method of claim 1, wherein in the compound (a) (i) when R9 and R11 are each –H and R10 and R12 are each –Br, then A is other than para- bromophenyl, meta-bromophenyl, ortho-tolyl or 3-quinolinyl; and (ii) when R9, R11 and R12 are each –H and R10 is –Br, then A is other than 3,5-dibromo- ortho-hydroxyphenyl, para-bromophenyl, meta-bromophenyl or ortho-tolyl; (b) the aryl or heteroaryl is substituted with halogen, C1-C6 alkyl, -OH, -O-(C1-C6 alkyl), -CHF2, - CF3, -OCHF2 or -OCF3; or (c) the fused aryl or fused heteroaryl is substituted with halogen, C1-C6 alkyl, -OH, -O-(C1-C6 alkyl), -CHF2, -CF3, -OCHF2 or -OCF3. 3. The method of any one of claims 1-2, wherein the compound has the following structure: R10 R R O 9 R11 2 ,
Figure imgf000077_0001
each independently, -H, halogen, C1-C6 alkyl, -OH, -O-(C1-C6 alkyl), -CHF2, -CF3, -OCHF2, -OCF3, -CN, -CH2OCH3, -N(CH3)2, -CH2F, -N3 or -CCH. 4. The method of any one of claims 2-3, wherein in the compound (a) R1 and R2 are each H; (b) R3, R4, R5, R6 and R7 are each independently -H, halogen, C1-C6 alkyl, -OH, -O-(C1-C6 alkyl), -CHF2, -CF3, -OCHF2 or -OCF3; preferably, R3, R4, R5, and R6 are each independently halogen, -O-(C1-C6 alkyl), -OCF3 or -CF3; more preferably, R3, R4, R5, and R6 are each independently halogen or -O-(C1-C6 alkyl); more preferably, R3, R4, R5, and R6 are each independently –Cl, - Br, -F, -O-(C1-C6 alkyl), -OCF3 or -CF3; more preferably, R3, R4, R5, and R6 are each independently –Cl, -Br, or -O-(C1-C6 alkyl); (c) R9, R10, R11, and R12 are each independently H, halogen, C1-C6 alkyl, -OH, -O-(C1-C6 alkyl), - CHF2, -CF3, -OCHF2 or -OCF3; preferably, R9, R10, R11, and R12 are each independently halogen, -O-(C1-C6 alkyl), -OCF3 or -CF3; more preferably, R9, R10, R11, and R12 are each independently halogen or -O-(C1-C6 alkyl); more preferably, R9, R10, R11, and R12 are each independently –Cl, -Br, -F, -O-(C1-C6 alkyl), -OCF3 or -CF3; more preferably, R9, R10, R11, and R12 are each independently –Cl, -Br, or -O-(C1-C6 alkyl); (d) at least one of R3, R4, R5, R6 and R7 is not H; (e) at least two of R3, R4, R5, R6 and R7 are not H; (f) at least one of R9, R10, R11, and R12 is not H; (g) at least two of R9, R10, R11, and R12 are not H; (h) R3, R4, R6, R7, R10 and R12 are H; (i) R3, R4, R6, R9, R10 and R12 are H; (j) R5, R9, and R10 are halogen; preferably R5, R9, and R10 are Br; or (k) R5, R7, R11 are halogen; preferably R5, R9, and R10 are Br, 5. The method of any one of claims 3-4; wherein in the compound R1 is -H; R2 is -H; R3, R4, R5, R6 and R7 are each, independently, H, halogen, C1-C6 alkyl, -OH; and R9, R10, R11, and R12 are each independently H, halogen, C1-C6 alkyl, -OH; preferably, R1 is -H; R2 is -H; R3, R4, R5, R6 and R7 are each, independently, H or halogen; and R9, R10, R11, and R12 are each independently H or halogen; more preferably, R1 is -H; R2 is -H; R3, R4, R5, R6 and R7 are each, independently H or Br; and R9, R10, R11, and R12 are each independently H or Br.
6. The method of claim 3, wherein in the compound OH OH OH Br Cl Br Cl , . , , ,
Figure imgf000079_0001
9. The method of claim 3, wherein in the compound R3 R4 .
Figure imgf000080_0001
he compound has the following structure: , , , , ,
Figure imgf000080_0002
, ,, ,
Figure imgf000081_0001
. , , ,
Figure imgf000081_0002
, , , , , , ,
Figure imgf000082_0001
HO Br O , , , , , , ,
Figure imgf000083_0001
, , , , ,
Figure imgf000084_0001
, , , or
Figure imgf000085_0001
13. The method of claim 1, wherein the compound has the following structure: Br O Br .
Figure imgf000086_0001
ture: Br O r . 15. mpound has the following structure:
Figure imgf000086_0002
R10 R10 R R O 9 11 R Br O 9 R11 12
Figure imgf000086_0003
R11 is -H, -Br, or -NR14R15, R10, R12, and R12 are each independently -H, -Br, -OH, -OR13, or -NR14R15; preferably, R10, R12, and R12 are each independently -H, -Br, -OH, -OCH3, or -N(CH3)2. 16. The method of claim 1, wherein the compound has the following structure: R5 R5 R5 R5 R R R4 R6 R R R4 R6 R7 ,
Figure imgf000086_0004
R5 R5 R4 R6 R4 R6 7 . 17. e compound has the following structure:
Figure imgf000087_0001
Br Br CH3 ,
Figure imgf000087_0002
. e met o o cam , w eren n t e compoun , 3, 4, 5, 6, an 7 are eac n epen ently H, halogen, -CN, alkyl, alkenyl, alkynyl, aryl, heteroaryl, heterocycle, -OCF3, -OH, -OAc, -OR13, -COR13, -CH2OR13, preferably, R3, R4, R5, R6, and R7 are each independently, H, halogen, -OCF3, heterocycle , or -CH2OR13, more preferably, R3, R4, R5, R6, and R7 are each independently, H or halogen; more preferably, [A] a) R3 is halogen, R4, R5, R6, and R7 are H; b) R4 is halogen, R3, R5, R6, and R7 are H; c) R5 is halogen, R3, R4, R6, and R7 are H; d) R6 is halogen, R3, R4, R5, and R7 are H; e) R7 is halogen, R3, R4, R5, and R6 are H; f) R3 and R4 are halogen, R5, R6, R7 are H; g) R3 and R5 are halogen, R4, R6, R7 are H; h) R3 and R6 are halogen, R4, R5, R7 are H; i) R3 and R7 are halogen, R4, R5, R6 are H; j) R4 and R5 are halogen, R3, R6, R7 are H; k) R4 and R6 are halogen, R3, R5, R7 are H; l) R4 and R7 are halogen, R3, R5, R6 are H; m) R5 and R6 are halogen, R3, R4, R7 are H; n) R5 and R7 are halogen, R3, R4, R6 are H; or o) R6 and R7 are halogen, R3, R4, R5 are H; preferably, the heterocycle is six-membered heterocycle, more preferably, the heterocycle is five- membered heterocycle, more preferably, the heterocycle is four-membered heterocycle, more preferably, the heterocycle is three-membered heterocycle; [B] R3 is OH, R4, R5, R6 and R7 are each independently H or halogen, preferably, a) R3 is OH, R4, R5, and R6 are H, R7 is halogen; b) R3 is OH, R5, R6, and R7 are H, R4 is halogen; c) R3 is OH, R4, R6, and R7 are H, R5 is halogen; d) R3 is OH, R4, R5, and R7 are H, R6 is halogen; e) R3 is OH, R4 and R5 are halogen, R6 and R7 are H; f) R3 is OH, R4 and R6 are halogen, R5 and R7 are H; g) R3 is OH, R4 and R7 are halogen, R5 and R6 are H; h) R3 is OH, R5 and R6 are halogen, R4 and R7 are H; i) R3 is OH, R5 and R7 are halogen, R4 and R6 are H; or j) R3 is OH, R6 and R7 are halogen, R4 and R5 are H; preferably, the halogen is F, Cl, Br, and I, preferably, the halogen is Br; [C] R7 is OH, R3, R4, R5 and R6 are each independently H or halogen, preferably, a) R7 is OH, R4 and R5 are halogen, R3 and R6 are H; b) R7 is OH, R4 and R6 are halogen, R3 and R5 are H; c) R7 is OH, R3 and R4 are halogen, R5 and R6 are H; d) R7 is OH, R3 and R5 are halogen, R4 and R6 are H; e) R7 is OH, R3 and R6 are halogen, R4 and R5 are H; f) R7 is OH, R5 and R6 are halogen, R3 and R4 are H; g) R7 is OH, R3, R4, and R5 are H, R6 is halogen; h) R7 is OH, R3, R4, and R6 are H, R5 is halogen; i) R7 is OH, R4, R5, and R6 are H, R3 is halogen; or j) R7 is OH, R3, R5, and R6 are H, R4 is halogen; preferably, the halogen is F, Cl, Br, and I, preferably, the halogen is Br; [D] R3, R4, R5, R6, R7 are each independently, H or CH2OR13, preferably, a) R3 is CH2OR13, R4, R5, R6, and R7 are H; b) R4 is CH2OR13, R3, R5, R6, and R7 are H; c) R5 is CH2OR13, R3, R4, R6, and R7 are H; d) R6 is CH2OR13, R3, R4, R5, and R7 are H; e) R7 is CH2OR13, R3, R4, R5, and R6 are H; f) R3 and R4 are CH2OR13, R5, R6, R7 are H; g) R3 and R5 are CH2OR13, R4, R6, R7 are H; h) R3 and R6 are CH2OR13, R4, R5, R7 are H; i) R3 and R7 are CH2OR13, R4, R5, R6 are H; j) R4 and R5 are CH2OR13, R3, R6, R7 are H; k) R4 and R6 are CH2OR13, R3, R5, R7 are H; l) R4 and R7 are CH2OR13, R3, R5, R6 are H; m) R5 and R6 are CH2OR13, R3, R4, R7 are H; n) R5 and R7 are CH2OR13, R3, R4, R6 are H; or o) R6 and R7 are CH2OR13, R3, R4, R5 are H; preferably, R5 is CH2OR13 and R13 is branched or unbranched alkyl, more preferably, alkyl is C1-C6 alkyl, branched or unbranched, more preferably, alkyl is methyl, ethyl, propyl, or isopropyl; [E] R3, R4, R5, R6, and R7 are each independently, H, halogen or -OCF3, preferably, a) R3 is halogen, R4, R5, and R6 are H, R7 is -OCF3; b) R3 is halogen, R5, R6, and R7 are H, R4 is -OCF3; c) R3 is halogen, R4, R6, and R7 are H, R5 is -OCF3; d) R3 is halogen, R4, R5, and R7 are H, R6 is -OCF3; e) R4 is halogen, R3, R5, and R6 are H, R7 is -OCF3; f) R4 is halogen, R3, R5, and R7 are H, R6 is -OCF3; g) R4 is halogen, R3, R6, and R7 are H, R5 is -OCF3; h) R4 is halogen, R5, R6, and R7 are H, R3 is -OCF3; i) R6 is halogen, R3, R4, and R5 are H, R7 is -OCF3; j) R6 is halogen, R3, R4, and R7 are H, R5 is -OCF3; k) R6 is halogen, R5, R6, and R7 are H, R3 is -OCF3; l) R6 is halogen, R3, R5, and R7 are H, R4 is -OCF3; m) R7 is halogen, R3, R4, and R5 are H, R6 is -OCF3; n) R7 is halogen, R3, R4, and R6 are H, R5 is -OCF3; o) R7 is halogen, R4, R5, and R6 are H, R3 is -OCF3; or p) R7 is halogen, R3, R5, and R6 are H, R4 is -OCF3. preferably, the halogen is F, Cl, Br, and I, more preferably, the halogen is F; [F] R3, R4, R5, R6, and R7 are each H or heterocycle, preferably, a) R3 is heterocycle, R4, R5, R6, and R7 are H; b) R4 is heterocycle, R3, R5, R6, and R7 are H; c) R5 is heterocycle, R3, R4, R6, and R7 are H; d) R6 is heterocycle, R3, R4, R5, and R7 are H; or e) R7 is heterocycle, R3, R4, R5, and R6 are H; preferably, the heterocycle is six-membered heterocycle, more preferably, heterocycle is five- membered heterocycle, more preferably, heterocycle is four-membered heterocycle, more preferably, heterocycle is three-membered heterocycle; preferably, the three-membered heterocycle is aziridine, 2H-azirine, oxirane, thiirane or azirine, more preferably, the azirine is 3-methyl-3-(trifluoromethyl)-3H-diazirine;or [G] R4, R5, and R6 are each H or heterocycle, R7 is H, preferably, a) R4 is heterocycle, R3, R5, R6, and R7 are H; b) R5 is heterocycle, R3, R4, R6, and R7 are H; or c) R6 is heterocycle, R3, R4, R5, and R7 are H preferably, the heterocycle is six-membered heterocycle, more preferably, heterocycle is five- membered heterocycle, more preferably, heterocycle is four-membered heterocycle, more preferably, heterocycle is three-membered heterocycle; preferably, the three-membered heterocycle is aziridine, 2H-azirine, oxirane, thiirane or azirine, preferably, the azirine is 3-methyl-3-(trifluoromethyl)-3H-diazirine. 19. The method of claim 1, wherein the compound has the following structure: [A] , structure:
Figure imgf000091_0001
, ,
Figure imgf000091_0002
.;
Figure imgf000091_0003
wherein ring A has the following structure: ,
Figure imgf000092_0001
;
Figure imgf000092_0002
, or
Figure imgf000092_0003
r
Figure imgf000092_0004
[D] ,
Figure imgf000093_0001
g structure: ,
Figure imgf000093_0002
. 20. e compound has the following structure:
Figure imgf000093_0003
, or
Figure imgf000093_0004
21. The method of claim 1, wherein the compound has the following structure: , , , , , , , ,
Figure imgf000094_0001
, . 22. [
Figure imgf000095_0001
A] ,
Figure imgf000095_0002
[C] , 23.
Figure imgf000096_0001
(a) the amount of ITC and the amount of the compound, or pharmaceutically acceptable salt of the compound, when taken together are effective to achieve an additive or a greater than additive therapeutic result in treating the subject; and/or (b) the amount of ITC and the amount of the compound, or pharmaceutically acceptable salt of the compound, when taken together are effective to achieve a synergism in treating the subject. 24. The method of any one of claims 1-22, wherein (a) the combination is more effective in treating the subject against fungal infection than when ITC and the compound, or pharmaceutically acceptable salt of the compound, at the same amount is administered alone; (b) the amount of the ITC and the amount of the compound, or pharmaceutically acceptable salt of the compound, when administered together, are more effective to treat the subject than when each at the same amount is administered alone; (c) the amount of the ITC administered in the combination is less than the fungicidally effective amount of the ITC when the ITC is administered alone, and/or the amount of the compound, or pharmaceutically acceptable salt of the compound, administered in the combination is less than the fungicidally effective amount of the compound, or pharmaceutically acceptable salt of the compound, when the compound, or pharmaceutically acceptable salt of the compound, is administered alone. 25. The method of any one of claims 1-24, wherein (a) the amount of ITC is administered in the range of 1mg/kg to 100mg/kg subject body weight per day; preferably in the range of 1mg/kg to 70 mg/kg subject body weight per day; more preferably, in the range of 5mg/kg to 60 mg/kg subject body weight per day; more preferably, in the range of 10 mg/kg to 20 mg/kg subject body weight per day, and/or (b) the amount of the compound, or pharmaceutically acceptable salt of the compound, is administered in the range of 1mg/kg to 100mg/kg subject body weight per day; preferably in the range of 1mg/kg to 70 mg/kg subject body weight per day, more preferably, in the range of 5mg/kg to 60 mg/kg subject body weight per day; more preferably, in the range of 10 mg/kg to 20 mg/kg subject body weight per day. 26. The method of claim 25, wherein the ratio between the amount of ITC and the amount of the compound, or pharmaceutically acceptable salt of the compound, is from 100:1 to 1:100, 50:1 to 1:50, 20:1 to 1:20, 10:1 to 1:10, 5:1 to 1:5 or 2:1 to 1:2. 27. The method of claim 26, wherein (a) the ratio between the amount of ITC and the amount of the compound, or pharmaceutically acceptable salt of the compound, is from 100:1 to 1:2, 50:1 to 1:2, 20:1 to 1:2, 10:1 to 1:2, 5:1 to 1:2 or 2.5:1 to 1:2.5; (b) the ratio between the amount of ITC and the amount of the compound, or pharmaceutically acceptable salt of the compound, is from 1:100 to 1:2, 1:50 to 1:2, 1:20 to 1:2, 1:10 to 1:2, 1:5 to 1:2 or 1:2.5 to 1:2; and/or (c) the amount of ITC and the amount of the compound, or pharmaceutically acceptable salt of the compound, is administered once, twice, or third times per day for a period of time, preferably, the period is 4-72 weeks; more preferably, the period is 4-32 weeks. 28. The method of claim 1, wherein the subject is a mammal; preferably, the mammal is cat or human. 29. The method of any one of claims 1-28, wherein (a) the ITC and the compound, or pharmaceutically acceptable salt of the compound, are administered together; preferably, the ITC and the compound, or pharmaceutically acceptable salt of the compound, are administered in a single dose; (b) the ITC and the compound, or pharmaceutically acceptable salt of the compound, are administered separately; (c) the ITC and the compound, or pharmaceutically acceptable salt of the compound, are administered simultaneously; (d) the ITC and the compound, or pharmaceutically acceptable salt of the compound, are applied contemporaneously, or (e) the ITC and the compound, or pharmaceutically acceptable salt of the compound, are applied successively. 30. The method of any one of claims 1-29, further comprises administering to the subject an amount of potassium iodine (KI). 31. The method of claim 30, wherein (a) the amounts of ITC, KI and the compound, or pharmaceutically acceptable salt of the compound, when taken together are effective to achieve an additive or a greater than additive therapeutic result in treating the subject; and/or (b) the amounts of ITC, KI and the compound, or pharmaceutically acceptable salt of the compound, when taken together are effective to achieve a synergism in treating the subject. 32. The method of any one of claims 30-31, wherein (a) the combination is more effective in treating the subject against fungal infection than when ITC, KI, and the compound, or pharmaceutically acceptable salt thereof, at the same amount is administered alone; (b) the amount of the ITC, KI, and the amount of the compound, or pharmaceutically acceptable salt thereof, when administered together, are more effective to treat the subject than when each at the same amount is administered alone; (c) the amount of the ITC and KI administered in the combination is less than the fungicidally effective amount of the ITC and KI when the ITC and KI is administered alone, and/or wherein the amount of the compound, or pharmaceutically acceptable salt thereof, administered in the combination is less than the fungicidally effective amount of the compound, or pharmaceutically acceptable salt thereof, when the compound, or pharmaceutically acceptable salt thereof, is administered alone. 33. The method of any one of claims 30-32, wherein (a) The amount of ITC is administered in the range of 1mg/kg to 100 mg/kg subject body weight per day; preferably, in the range of 1mg/kg to 70 mg/kg subject body weight per day; more preferably, in the range of 5mg/kg to 60 mg/kg subject body weight per day; more preferably, in the range of 10 mg/kg to 20 mg/kg subject body weight per day; (b) the amount of KI is administered in the range of 1mg/kg to 100 mg/kg subject body weight per day; preferably, in the range of 1mg/kg to 20 mg/kg subject body weight per day; more preferably, in the range of 2mg/kg to 16 mg/kg subject body weight per day; more preferably, in the range of 5 mg/kg to 10 mg/kg subject body weight per day; and/or (c) the amount of the compound is administered in the range of 1mg/kg to 100 mg/kg subject body weight per day; preferably, in the range of 1mg/kg to 70 mg/kg subject body weight per day, more preferably, in the range of 5mg/kg to 60 mg/kg subject body weight per day; more preferably, in the range of 10 mg/kg to 20 mg/kg subject body weight per day. 34. The method of claim 33, wherein (a) the ratio between the amount of ITC and the amount of the compound, or pharmaceutically acceptable salt thereof, is from 100:1 to 1:100, 50:1 to 1:50, 20:1 to 1:20, 10:1 to 1:10, 5:1 to 1:5 or 2:1 to 1:2; (b) the ratio between the amount of ITC and the amount of the compound, or pharmaceutically acceptable salt thereof, is from 100:1 to 1:2, 50:1 to 1:2, 20:1 to 1:2, 10:1 to 1:2, 5:1 to 1:2 or 2:1 to 1:2; (c) the ratio between the amount of ITC and the amount of the compound, or pharmaceutically acceptable salt thereof, is from 2:1 to 1:100, 2:1 to 1:50, 2:1 to 1:20, 2:1 to 1:10, 2:1 to 1:5 or 2:1 to 1:2; (d) the ratio between the amount of ITC and KI is from 100:1 to 1:100, 50:1 to 1:50, 20:1 to 1:20, 10:1 to 1:10, 5:1 to 1:5 or 2:1 to 1:2; (e) the ratio between the amount of ITC and KI is from 100:1 to 1:2, 50:1 to 1:2, 20:1 to 1:2, 10:1 to 1:2, 5:1 to 1:2 or 2:1 to 1:2; (f) the ratio between the amount of ITC and KI is from 2:1 to 1:100, 2:1 to 1:50, 2:1 to 1:20, 2:1 to 1:10, 2:1 to 1:5 or 2:1 to 1:2; (g) the ratio between the amount of KI and the compound, or pharmaceutically acceptable salt thereof, is from 100:1 to 1:100, 50:1 to 1:50, 20:1 to 1:20, 10:1 to 1:10, 5:1 to 1:5 or 2:1 to 1:2 (h) the ratio between the amount of KI and the compound, or pharmaceutically acceptable salt thereof, is from 100:1 to 1:2, 50:1 to 1:2, 20:1 to 1:2, 10:1 to 1:2, 5:1 to 1:2 or 2:1 to 1:2; or (i) the ratio between the amount of KI and the compound, or pharmaceutically acceptable salt thereof, is from 2:1 to 1:100, 2:1 to 1:50, 2:1 to 1:20, 2:1 to 1:10, 2:1 to 1:5 or 2:1 to 1:2.
35. The method of any one of claims 30-34, wherein (a) the ITC, KI and the compound, or pharmaceutically acceptable salt thereof, are administered together; preferably, the ITC, KI and the compound, or pharmaceutically acceptable salt thereof, are administered in a single dose; (b) the ITC, KI and the compound, or pharmaceutically acceptable salt thereof are administered separately; (c) the ITC, KI and the compound, or pharmaceutically acceptable salt thereof, are administered simultaneously; (d) the ITC, KI and the compound , or pharmaceutically acceptable salt thereof, are applied contemporaneously, or (e) the ITC, KI and the compound, or pharmaceutically acceptable salt thereof, are applied successively. 36. The method of any one of claims 30-35, wherein (a) the amount of ITC, KI and the compound, or pharmaceutically acceptable salt thereof, is administered once twice, or third times per day for a period of time; preferably, the period is 4-72 weeks; more preferably, the period is 4-32 weeks; and/or (b) the subject is a mammal; preferably, the mammal is cat or human. 37. The method of any one of claims 1-36; wherein the fungal infection is caused by Cryptococcus Neoformans, Cryptococcus gattii, Candida albicans, Candida krusei, Candida glabrata, Candida parapsilosis, Candida guilliermondii, Aspergillus fumigatus, Rhizopus oryzae, Rhizopus spp., Blastomyces dermatitis, Histoplasma capsulatum, Coccidioides spp., Paecilomyces variotii, Pneumocystis murina, Pneumocystis jiroveci, Histoplasma capsulatum, Aspergillus spp., S. brasiliensis, S. schenckii, S. globosa, S. mexicana, S. chilensis, S. luriei, or S. pallida; preferably, the fungal infection is caused by is Cryptococcus Neoformans or Sporothrix brasiliensis; more preferably, the fungal infection is caused by Sporothrix brasiliensis. 38. A combination comprising an amount of itraconazole (ITC) and an amount of a compound, or a pharmaceutically acceptable salt thereof, wherein the compound has the following structure: ,
Figure imgf000100_0001
wherein R1 is -H, alkyl, alkenyl, or alkynyl; R2 is -H, alkyl, alkenyl, or alkynyl; R9, R10, R11, and R12 are each independently, H, halogen, -CN, -CF3, -OCF3, -NO2, alkyl, alkenyl, alkynyl, aryl, heteroaryl, cycloalkyl, heterocycloalkyl, -OH, -OAc, -OR13, -COR13, -SH, -SR13, - SO2R13, -SO2NR14R15, -NH2, -NHR13, -NR14R15, -NHCOR12, or -CONR14R15; or R9 and R10 are each independently -H, halogen, C1-C6 alkyl, -OH, -O-(C1-C6 alkyl), -CHF2, -CF3, -OCHF2 or -OCF3, and R11 and R12 combine to form a fused aryl or fused heteroaryl, which are each unsubstituted or substituted, or R9 and R12 are each independently -H, halogen, C1-C6 alkyl, -OH, -O-(C1-C6 alkyl), -CHF2, -CF3, -OCHF2 or -OCF3, and R10 and R11 combine to form a fused aryl or fused heteroaryl, which are each unsubstituted or substitute, or R11 and R12 are each independently -H, halogen, C1-C6 alkyl, -OH, -O-(C1-C6 alkyl), -CHF2, -CF3, -OCHF2 or -OCF3, and R3 and R4 combine to form a fused aryl or fused heteroaryl, which are each unsubstituted or substituted; wherein each occurrence of R13 is independently alkyl, alkenyl, alkynyl, aryl, or heteroaryl; wherein each occurrence of R14 and R15 is independently –H, alkyl, alkenyl, alkynyl, aryl, or heteroaryl; wherein when R14 is methyl, R15 is not methyl; and wherein at least one of R9, R10, R11, and R12 is not H; for use in treating a subject afflicted with a fungal infection by administering the combination to the subject . 39. The combination of claim 38, wherein the combination is an improved combination in that: (a) the amount of the compound, or pharmaceutically acceptable salt thereof, improves the fungicidal efficacy of ITC compared to when the same amount of the ITC is administered not in combination with the amount of the compound, or pharmaceutically acceptable salt thereof; or the amount of ITC improves the fungicidal efficacy of the compound compared to when the same amount of the compound, or pharmaceutically acceptable salt thereof, is applied not in combination with the amount of ITC; (b) a substantially similar level of fungicidal efficacy is achieved by using a lesser amount of the ITC and/or the compound, or pharmaceutically acceptable salt thereof; (c) the amount of the compound, or pharmaceutically acceptable salt thereof, is effective to increase sensitivity of the fungus to the amount of ITC compared to the sensitivity of the fungus to the amount of the ITC when it is administered not in combination with the amount of the compound, or pharmaceutically acceptable salt thereof; or the amount of the ITC is effective to increase sensitivity of the fungus to the amount of the compound, or pharmaceutically acceptable salt thereof, compared to the sensitivity of the fungus to the amount of the compound, or pharmaceutically acceptable salt thereof, when it is administered not in combination with the amount of ITC; (d) the amount of the compound is effective to increase bioavailability of the amount of ITC compared to the bioavailability of the amount of the ITC when it is administered not in combination with the amount of the compound; or the amount of ITC is effective to increase bioavailability of the amount of the compound compared to the bioavailability of the amount of the compound when it is administered not in combination with the amount of ITC; (e) the combination is more effective in treating the subject against fungal infection than when ITC and the compound, or pharmaceutically acceptable salt thereof, at the same amount is administered alone, (f) the combination prolongs the period of protection against fungal infection and/or control of fungal infection than when the amount of ITC and the amount of the compound, or pharmaceutically acceptable salt thereof, are administered alone; and/or (g) the combination reduces the amount of time needed to achieve a level of fungal control than when the amount of ITC and the amount of the compound, or pharmaceutically acceptable salt thereof, are administered alone. 40. The combination of claim 38, wherein the compound has the following structure: Br r .
Figure imgf000102_0001
41. A pharmaceutical composition in unit dosage form, useful in treating a subject afflicted with fungal infection, which comprises: (a) an amount of ITC; (b) an amount of the compound from any of claims 38-40, or a pharmaceutically acceptable salt thereof; and (c) optionally an amount of KI; wherein the respective amounts of said compound, or a pharmaceutically acceptable salt thereof, said ITC and said KI, if present, in said composition are effective, upon concomitant administration to said subject of one or more of said unit dosage forms of said composition, to treat the subject. 42. A therapeutic package for dispensing to, or for use in dispensing to, a subject afflicted with fungal infection, which comprises: (a) one or more unit doses, each such unit dose comprising: i) an amount of ITC; ii) an amount of the compound from any of claims 38-40, or a pharmaceutically acceptable salt thereof; and iii) optionally an amount of KI; wherein the respective amounts of said compound, or a pharmaceutically acceptable salt thereof, said ITC and said KI, if present, in said unit doses are effective, upon concomitant administration to said subject, to treat the subject, and (b) a finished pharmaceutical container therefor, said container containing said unit dose or unit doses, said container further containing or comprising labeling directing the use of said package in the treatment of said subject. 43. The package, pharmaceutical composition, or therapeutic package of claim 42, wherein the pharmaceutical composition, unit dose, or unit dosage form comprises: (a) 25-75 mg of the compound, or a pharmaceutically acceptable salt thereof, 10-100 mg of ITC, 1-20 mg of KI, if present; and (b) 25-75 mg of the compound, or a pharmaceutically acceptable salt thereof, 10-100 mg of ITC, 1-20 mg of KI, if present and 11-50 mg of sylimarin, if present. 44. The package, pharmaceutical composition, or therapeutic package of claim 42, wherein the pharmaceutical composition, unit dose, or unit dosage form comprises: (a) 25 mg of the compound, or a pharmaceutically acceptable salt thereof, 25 mg of ITC, 2.5 mg of KI and 30 mg sylimarin; (b) 20 mg of the compound, or a pharmaceutically acceptable salt thereof, 50 mg of ITC, and 2.5 mg of KI; and (c) 20 mg of the compound or a pharmaceutically acceptable salt thereof, and 100 mg of ITC.
PCT/US2024/054777 2023-11-06 2024-11-06 Combination therapy of itraconazole and acylhydrazone-based antifungalagents for the treatment of sporotrichosis in cats Pending WO2025101647A1 (en)

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US202363596461P 2023-11-06 2023-11-06
US63/596,461 2023-11-06
US202463570884P 2024-03-28 2024-03-28
US63/570,884 2024-03-28

Publications (2)

Publication Number Publication Date
WO2025101647A1 true WO2025101647A1 (en) 2025-05-15
WO2025101647A9 WO2025101647A9 (en) 2025-06-12

Family

ID=95696273

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2024/054777 Pending WO2025101647A1 (en) 2023-11-06 2024-11-06 Combination therapy of itraconazole and acylhydrazone-based antifungalagents for the treatment of sporotrichosis in cats

Country Status (1)

Country Link
WO (1) WO2025101647A1 (en)

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20120010075A1 (en) * 2009-01-14 2012-01-12 Dow Agrosciences Llc Synergistic fungicidal compositions including hydrazone derivatives and copper
US20150374719A1 (en) * 2014-06-26 2015-12-31 Brigham Young University Methods for treating fungal infections
US20220394973A1 (en) * 2017-06-16 2022-12-15 The Research Foundation For The State University Of New York Anti-fungals compounds targeting the synthesis of fungal sphingolipids

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20120010075A1 (en) * 2009-01-14 2012-01-12 Dow Agrosciences Llc Synergistic fungicidal compositions including hydrazone derivatives and copper
US20150374719A1 (en) * 2014-06-26 2015-12-31 Brigham Young University Methods for treating fungal infections
US20220394973A1 (en) * 2017-06-16 2022-12-15 The Research Foundation For The State University Of New York Anti-fungals compounds targeting the synthesis of fungal sphingolipids

Also Published As

Publication number Publication date
WO2025101647A9 (en) 2025-06-12

Similar Documents

Publication Publication Date Title
US11858880B2 (en) Anti-fungals compounds targeting the synthesis of fungal sphingolipids
EP2717692B1 (en) Tetrahydrocannabinol-11-oic acids for use in treating fibrotic diseases
RU2727520C2 (en) Antifungal agents
KR102528435B1 (en) Biphenyl sulfonamide compounds for the treatment of kidney diseases or disorders
KR102347721B1 (en) PPAR compounds for use in the treatment of fibrotic diseases
US11174267B2 (en) Spiroindolinones and therapeutic uses thereof
US20160113937A1 (en) Discovery of fda-approved drugs as inhibitors of fatty acid binding protein 4 using molecular docking screening
US20210379040A1 (en) Combination treatment of liver disorders
EP2636409A1 (en) Combined pharmaceutical composition as antifungal agent
US20250109096A1 (en) Anti-fungals targeting the synthesis of fungal shingolipids
WO2021163633A1 (en) Mono and combination therapies with ulk1/2 inhibitors
JP5587488B1 (en) Evaluation method and index substance of preparation containing luliconazole
WO2020142547A1 (en) Methods and formulations to treat mitochondrial dysfunction
WO2025101647A1 (en) Combination therapy of itraconazole and acylhydrazone-based antifungalagents for the treatment of sporotrichosis in cats
JP2018520104A (en) Antifungal 4- (4- (4-(((3R, 5R) -5-((1H-1,2,4-triazol-1-yl) methyl) -5- (2,4-difluorophenyl) ) Tetrahydrofuran-3-yl) methoxy) -3-methylphenyl) piperazin-1-yl) -N- (2-hydroxycyclohexyl) benzamide, or a pharmaceutically acceptable salt thereof.
JP2025531219A (en) Compositions and methods for treating depression in women
EP4392025A1 (en) Anti-fungals compounds targeting the synthesis of fungal sphingolipids
EP4598906A2 (en) Sterylglucosidase inhibiting compositions and method of using
AU2010352387B2 (en) Diyne compositions
JP5926756B2 (en) Evaluation method and index substance of preparation containing luliconazole
JP2015113342A (en) Evaluation method and index material of preparation containing luliconazole

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 24889522

Country of ref document: EP

Kind code of ref document: A1