WO2023001905A1 - Novel antifungal compounds - Google Patents

Novel antifungal compounds Download PDF

Info

Publication number
WO2023001905A1
WO2023001905A1 PCT/EP2022/070389 EP2022070389W WO2023001905A1 WO 2023001905 A1 WO2023001905 A1 WO 2023001905A1 EP 2022070389 W EP2022070389 W EP 2022070389W WO 2023001905 A1 WO2023001905 A1 WO 2023001905A1
Authority
WO
WIPO (PCT)
Prior art keywords
alkyl
compound
compound according
spp
equiv
Prior art date
Application number
PCT/EP2022/070389
Other languages
French (fr)
Inventor
Philipp Knechtle
Frank Otto Gombert
Rainer Riedl
Urs LINDENMANN
Walter Krebs
Dennis WIPFLI
Ina ALBERT
Sandro WEGMANN
Andrina SCHMID
Original Assignee
Selmod Gmbh
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 Selmod Gmbh filed Critical Selmod Gmbh
Publication of WO2023001905A1 publication Critical patent/WO2023001905A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D307/00Heterocyclic compounds containing five-membered rings having one oxygen atom as the only ring hetero atom
    • C07D307/02Heterocyclic compounds containing five-membered rings having one oxygen atom as the only ring hetero atom not condensed with other rings
    • C07D307/34Heterocyclic compounds containing five-membered rings having one oxygen atom as the only ring hetero atom not condensed with other rings having two or three double bonds between ring members or between ring members and non-ring members
    • C07D307/38Heterocyclic compounds containing five-membered rings having one oxygen atom as the only ring hetero atom not condensed with other rings having two or three double bonds between ring members or between ring members and non-ring members with substituted hydrocarbon radicals attached to ring carbon atoms
    • C07D307/54Radicals substituted by carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals
    • 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
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C311/00Amides of sulfonic acids, i.e. compounds having singly-bound oxygen atoms of sulfo groups replaced by nitrogen atoms, not being part of nitro or nitroso groups
    • C07C311/50Compounds containing any of the groups, X being a hetero atom, Y being any atom
    • C07C311/52Y being a hetero atom
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07FACYCLIC, CARBOCYCLIC OR HETEROCYCLIC COMPOUNDS CONTAINING ELEMENTS OTHER THAN CARBON, HYDROGEN, HALOGEN, OXYGEN, NITROGEN, SULFUR, SELENIUM OR TELLURIUM
    • C07F9/00Compounds containing elements of Groups 5 or 15 of the Periodic System
    • C07F9/02Phosphorus compounds
    • C07F9/547Heterocyclic compounds, e.g. containing phosphorus as a ring hetero atom
    • C07F9/6564Heterocyclic compounds, e.g. containing phosphorus as a ring hetero atom having phosphorus atoms, with or without nitrogen, oxygen, sulfur, selenium or tellurium atoms, as ring hetero atoms
    • C07F9/6571Heterocyclic compounds, e.g. containing phosphorus as a ring hetero atom having phosphorus atoms, with or without nitrogen, oxygen, sulfur, selenium or tellurium atoms, as ring hetero atoms having phosphorus and oxygen atoms as the only ring hetero atoms
    • C07F9/6574Esters of oxyacids of phosphorus
    • C07F9/65742Esters of oxyacids of phosphorus non-condensed with carbocyclic rings or heterocyclic rings or ring systems

Definitions

  • the present invention relates to novel compounds and pharmaceutical compositions comprising the same that can be used in the prevention, treatment and alleviation of a fungal infection.
  • the instant compounds and pharmaceutical compositions are particularly useful in treating, preventing and alleviating fungal infections caused by Candida fungi (including, but not limited to C. albicans, C. parapsilosis and C. krusei) and Aspergillus fungi (including, but not limited to A. fumigatus, A. flavus and A. niger).
  • Fungal infections can contribute to and complicate many diseases, including pulmonary diseases and respiratory tract diseases.
  • the incidence of fungal infections is steadily rising as a consequence of antibiotic treatments, an immune-suppressed or immune- compromised population (mainly caused by cancer treatment, HIV, allergy-treatments, transplantations and general surgery) and an aging population.
  • Candida species account for 80% of infections in general medicine, 40% in HIV populations and 90% in both cancer therapy and surgical-non transplant cases.
  • Candida is now the 4th largest cause of nosocomial blood stream infections. Mortality from systemic fungal infections remains high despite the development of new antifungal agents, and Candida bloodstream infections in the United States are associated with a 40% crude mortality rate. Since 1980, the mortality due to Aspergillus fumigatus is continually increasing.
  • Candida fungi are the fourth most common cause of nosocomial bloodstream infections. While C. albicans was the predominant cause of Candida bloodstream infections in the early 80s, C. glabrata has emerged as the second most common cause of said infections in various parts of the world, including the United States. In addition to Aspergillus and Candida infections, other fungal pathogens such as Scedosporium spp. are becoming increasingly important. Their susceptibility to existing antifungals is limited and their mortality rate is >70% in patients with hematological malignancies. In other patients the mortality rates vary between 30 and 80%.
  • Onychomycosis is a fungal infection of the nails which is estimated to affect up to 25% of the geriatric and diabetic populations and 2 to 13% of the general US population. Common risk factors include age, male gender, diabetes, nail trauma, and chronic Tinea pedis (fungal infection of the foot). Onychomycosis has significant cosmetic, psychological and social implications. In some patient subsets it has serious medical consequences (e.g., foot amputations in diabetics).
  • polyenes e.g., Amphotericin B
  • imidazoles and triazoles e.g., Fluconazole or Itraconazole
  • cytochrome P450 14a-demethylase which is required for ergosterol synthesis and homeostasis of fungal cell membrane
  • allylamines e.g., Terbinafine
  • echinocandins e.g., Caspofungin
  • Amphotericin B is nephrotoxic. Terbinafine is associated with liver damage. Furthermore, there is generalized intolerance to azoles, for example, up to 20% of females with vaginal candidiosis cannot tolerate Fluconazole. Moreover, many pathogenic strains are insensitive or resistant to currently available antifungal drugs and resistance development is a concern. Thus, mortality due to fungal infections remains high.
  • Caspofungin resistance is relatively rare with ca. 8% of C. tropicalis and ca. 2% of C. glabrata isolates having been classified as resistant (corresponding MIC values of > 2mg/L). Given the observation that 2001 to 2004 surveillance studies identified > 99.5% of patients as Caspofungin sensitive, it is disconcerting how rapidly echinocandin resistance is spreading. Furthermore, there have been reported cases of reduced C. glabrata susceptibility developing during Caspofungin therapy.
  • the target of Caspofungin is the enzyme 1,3-P-D-glucan synthase, encoded by one of several FKS genes, depending on the species.
  • WO 01/25197 refers to certain substantially pure ene-diyne compounds bearing a carboxylic group or a moiety that can be hydrolyzed to a carboxylic group, which are useful as antifungal agents.
  • WO 2011/006061 refers to certain substantially pure cis-isomer compounds bearing an ene- diyne moiety that are useful as Olel protein inhibitors and can be applied as antifungal agents.
  • WO 2011/134538 discloses specific diyne compounds substituted with an optionally substituted carbon chain bearing a -COO- group and with an optionally substituted heteroaromatic ring that can be used in fungicidal formulations.
  • the natural diyne-furan fatty acid compound of formula: is described as an inhibitor of fungal ⁇ -9 fatty acid desaturation, which shows efficacy in a model of skin dermatophytosis (Antimicrobial Agents and Chemotherapy, 2014, 58, 455-456).
  • the compounds should exhibit broad antifungal activity, in other words be suitable for treating fungal infections caused by diverse fungi.
  • the present inventors have surprisingly found that this object can be solved by the compounds having the formula (I).
  • the compounds of the present invention are useful in treating infections caused by Candida fungi such as C. albicans, C. parapsilosis, C. tropicalis and C. glabrata.
  • the compounds of the present invention are also useful in treating the infections caused by Aspergillus fungi such as A. fumigatus, A. flavus, A. niger and A. terreus.
  • Candida parapsilosis responsible for 30% cases of Candida infections in the south of Europe.
  • Candida parapsilosis is also the main Candida species, apart from Candida albicans, isolated from patients in South America. Due to mutations in Ergl 1 gene and overexpression of transporter proteins, Candida parapsilosis is resistant against treatment with fluconazole, an oral antifungal medication considered to be a standard of care.
  • compounds of formula (I) are active against Candida parapsilosis, and have surprisingly shown improved efficacy against Candida parapsilosis as compared with the state of the art compound EV-086. Furthermore, the compounds of formula (I) have surprisingly shown improved efficacy against Aspergillus fungi, for example, against A. flavus and A. fumigatus, as compared to the state-of-the-art compound EV-086.
  • the present invention can be summarized in the following aspects.
  • the present invention relates to a compound having the formula (I):
  • Z represents an alkylene moiety having 6 to 12 carbon atoms and optionally containing 1 or 2 double bonds, wherein
  • one or more non-adjacent -CH 2 - groups can be independently replaced by -O-, -S- -S(O)-, -C(O)-N(H)-, or-C(O)-N(C 1-4 alkyl)- and/or
  • one or more hydrogen atoms can be independently replaced by -F, -OH or C 1-4 alkyl, and/or
  • one or more -CH 2 -CH 2 - moieties can be independently replaced by a catenary 3- to 6- membered heterocyclic ring, and/or
  • one or more -CH 2 -CH 2 -CH 2 - moieties can be independently replaced by a catenary 3- to 6-membered heterocyclic ring;
  • R 3 is selected from
  • an optionally substituted heterocyclic or carbocyclic group having 5 to 10 ring atoms wherein the optional substituent is independently selected from -C 1-4 alkyl, -OH, -OC 1-4 alkyl, -Hal, -NH2, -NH(C 1-4 alkyl), -N(C 1-4 alkyl)2, and -NO2, or
  • the present invention relates to the compound of formula (I), wherein R 3 is optionally substituted as defined in claim 1 and is selected from furane, thiophene, norbomane, cyclopropyl, pyrrole, oxazole, isoxazole, thiazole, isothiazole, pyrazole, and imidazole, preferably R 3 is furane.
  • the present invention relates to the compound of formula (I), wherein the compound is selected from:
  • Another aspect of the present invention relates to a compound of formula (I): R 1 -Z-C ⁇ C-C ⁇ C-R 3
  • Z represents an alkylene moiety having 6 to 12 carbon atoms and optionally containing 1 or 2 double bonds, wherein (i) one or more non-adjacent -CH 2 - groups can be independently replaced by -O-, -S-, - S(O)-, -C(O)-N(H)-, or -C(O)-N(C 1-4 alkyl)- and/or
  • one or more hydrogen atoms can be independently replaced by -F, -OH or C 1-4 alkyl, and/or
  • one or more -CH 2 -CH 2 - moieties can be independently replaced by a catenary 3- to 6- membered heterocyclic ring, and/or
  • one or more -CH 2 -CH 2 -CH 2 - moieties can be independently replaced by a catenary 3- to 6-membered heterocyclic ring;
  • R 3 represents an aliphatic group containing 1 to 4 carbon atoms, wherein one divalent carbon atom can be replaced by oxygen or sulfur and wherein the alkyl group R 3 can be optionally substituted by one or more Hal.
  • the present invention relates to the compound of formula (I), wherein R 1 is selected from -COOH, -COONa and -COOK.
  • the present invention relates to the compound of formula (I), wherein R 3 is selected from -CH 2 CH 2 CH 3, -CH 2 OCH 3 , -CH 2 SCH 3 , -CHFCH 2 CH 3 , -CF 2 CH 2 CH 3 , preferably R 3 is -CH 2 CH 2 CH 3 .
  • the present invention relates to the compound of formula (I), wherein R 3 is cyclopropyl or cyclobutyl.
  • the present invention relates to the compound of the present invention, wherein the compound has a minimum inhibitory concentration of at most 1.0 mg/L against Candida albicans.
  • Another aspect of the present invention relates to the compound of the present invention for use as a medicament.
  • a yet another aspect of the present invention relates to a pharmaceutical composition
  • a pharmaceutical composition comprising the compound of the present invention and optionally a pharmaceutically acceptable carrier.
  • Another aspect of the present inventions relates to the compound of the present invention for use in the treatment, alleviation or prevention of a fungal infection.
  • a yet another aspect of the present invention relates to a use of the compound of the present invention for the preparation of a medicament for treating, alleviating or preventing a fungal infection.
  • Another aspect of the present invention relates to a method of treating, alleviating or preventing a fungal infection, the method comprising administering an effective amount of the compound of the invention to a subject in need thereof.
  • the fungal infection can be caused by a fungus selected from Candida spp. (for example C. albicans, C. auri, C. krusei, C. glabrata, C. tropicalis, C. parapsilosis, C. guilliermondii, C. haemulonii, C. lusitaniae, C. lipolytica, C. norvegensis, C. viswanathii, C. kefyr or C. dubliniensis), Aspergillus spp. (for example A.fumigatus, A.flavus, A. niger or A.
  • Candida spp. for example C. albicans, C. auri, C. krusei, C. glabrata, C. tropicalis, C. parapsilosis, C. guilliermondii, C. haemulonii, C. lusitaniae, C. lipolytica, C. norve
  • C. neoformans for example var. neoformans or var. gattii
  • C. bidus for example var. laurentii, or C.fusarium
  • Zygomycetes such as Saksenaea vasiformis
  • Malassezia spp. for example M. furfur or M. globosa
  • Hyalohyphomycetes for example Scedosporium spp., such as S. prolificans or S. apiospermum
  • Dermatophytes for example Trichophyton spp.
  • T. mentagrophytes, T. rubrum or T. tonsurans Epidermophyton floccosum
  • Microsporum spp for example M. cookei, M. canis, M. vanbreuseghemii, M. gallinae or M. gypseum) or Trichosporon terrestre
  • Blastomyces dermatitidis Sporothrix schenkii
  • Chromomycotic fungi for example Fonsecaea pedrosoi, F. compacta, Cladophylophora carrionii or Phial ophora verrucosa
  • Madurella spp. for example M. mycetomatis or M.
  • the fungal infection can be caused by a fungus selected from Candida fungi (including, but not limited to C. albicans, C. parapsylosis and C. krusei) and Aspergillus fungi (including, but not limited to A. fumigatus, A. flavus and A. niger).
  • Candida fungi including, but not limited to C. albicans, C. parapsylosis and C. krusei
  • Aspergillus fungi including, but not limited to A. fumigatus, A. flavus and A. niger.
  • Solvates, hydrates as well as anhydrous forms of the salt are also encompassed by the invention.
  • the solvent included in the solvates is not particularly limited and can be any pharmaceutically acceptable solvent. Examples include water and C 1-4 alcohols (such as methanol or ethanol).
  • Table 1 Summary of the studies of antifungal properties of the exemplary compounds 1 to 14 of the present invention against selected fungi. It is noted that compound 3 is shown as a reference example.
  • Figure 2 The results of the measurement of the plasma stability of the antifungal compounds 5, 8, 10, 13 and EV-086.
  • the ID of the compound is shown on the x-axis and the recovery rate on the y-axis.
  • the error bars indicate the standard errors of the measured triplicates. For each compound there are four bars which show the recovery rates of the measurements of the four timepoints 0, 60, 120 and 180 minutes (T0 to T3, respectively).
  • the compound according to formula (la) as described herein, is preferably a compound according to formula (Ic)
  • R 3 represents an aliphatic group containing 1 to 4 carbon atoms, wherein one divalent carbon atom can be replaced by oxygen or sulfur and wherein the alkyl group R 3 can be optionally substituted by one or more Hal.
  • R 3 is selected from -CH 2 CH 2 CH 3. -CH 2 OCH 3 , -CH 2 SCH 3 , -CHFCH 2 CH 3 , -CF 2 CH 2 CH 3 . More preferably, R 3 is -CH 2 CH 2 CH 3 .
  • R 3 represents an aliphatic group containing 1 to 4 carbon atoms, wherein one divalent carbon atom can be replaced by oxygen or sulfur and wherein the alkyl group R 3 can be optionally substituted by one or more Hal.
  • R 3 is selected from -CH 2 CH 2 CH 3, -CH 2 OCH 3 , -CH 2 SCH 3 , -CHFCH 2 CH 3 , -CF2CH 2 CH 3 . More preferably, R 3 is -CH 2 CH 2 CH 3 .
  • Sterilization of infusion or injection solutions can be accomplished by any number of art recognized techniques including but not limited to addition of preservatives like anti -bacterial or anti -fungal agents, e.g., parabene, chlorobutanol, phenol, sorbic acid or thimersal. Further, isotonic agents, such as sugars or salts, in particular sodium chloride may be incorporated in infusion or injection solutions.
  • preservatives like anti -bacterial or anti -fungal agents, e.g., parabene, chlorobutanol, phenol, sorbic acid or thimersal.
  • isotonic agents such as sugars or salts, in particular sodium chloride may be incorporated in infusion or injection solutions.
  • sterile injectable solutions containing one or several of the compounds of the invention is accomplished by incorporating the respective compound in the required amount in the appropriate solvent with various ingredients enumerated above as required followed by sterilization. To obtain a sterile powder the above solutions are vacuum-dried or freeze-dried as necessary.
  • Preferred diluents of the present invention are water, physiologically acceptable buffers, physiologically acceptable buffer salt solutions or salt solutions.
  • Preferred carriers are cocoa butter and vitebesole.
  • a compound of the invention may be administered in the form of a dry powder inhaler or an aerosol spray from a pressurized container, pump, spray or nebulizer with the use of a suitable propellant, e.g., dichlorodifluoromethane, trichlorofluorom ethane, dichlorotetrafluoroethane, a hydrofluoroalkane such as 1, 1,1,2- tetrafluoroethane (HFA 134ATM) or 1,1,1,2,3,3,3-heptafluoropropane (HFA 227EATM), carbon dioxide, or another suitable gas.
  • a suitable propellant e.g., dichlorodifluoromethane, trichlorofluorom ethane, dichlorotetrafluoroethane, a hydrofluoroalkane such as 1, 1,1,2- tetrafluoroethane (HFA 134ATM) or 1,1,1,2,3,3,3-
  • binders such as lactose, mannitol, crystalline sorbitol, dibasic phosphates, calcium phosphates, sugars, microcrystalline cellulose, carboxymethyl cellulose, hydroxyethyl cellulose, polyvinyl pyrrolidone and the like;
  • lubricants such as magnesium stearate, talc, calcium stearate, zinc stearate, stearic acid, hydrogenated vegetable oil, leucine, glycerids and sodium stearyl fumarates,
  • disintegrants such as starches, croscaramellose, sodium methyl cellulose, agar, bentonite, alginic acid, carboxymethyl cellulose, polyvinyl pyrrolidone and the like.
  • the compounds of the present invention are particularly useful for treating, ameliorating, or preventing fungal infections.
  • the compounds of the present invention are employed to treat fungal infections.
  • the subject to be treated is not particularly restricted and can be any vertebrate, such as birds and mammals (including humans).
  • the fungal infection that can be treated, ameliorated or prevented with the compounds of the present invention is not particularly limited and can be caused by a fungus selected from Candida spp. (for example C. albicans, C. auris, C. krusei, C. glabrata, C. tropicalis, C. parapsilosis, C. guilliermondii, C. haemulonii, C. lusitaniae, C. lipolytica, C.
  • Hyalohyphomycetes for example Scedosporium spp., such as S. prolificans or S. apiospermum
  • Dermatophytes for example Trichophyton spp. (for example T. mentagrophytes, T. rubrum or T. tonsurans), Epidermophyton floccosum
  • Microsporum spp for example M. cookei, M. canis, M. vanbreuseghemii, M. gallinae or M.
  • the fungal infection that can be treated, ameliorated or prevented with the compounds of the present invention is caused by a fungus selected from Candida spp. (for example C.
  • albicans C. auris, C. krusei, C. glabrata, C. tropicalis, C. parapsilosis, C. guilliermondii, C. haemulonii, C. lusitaniae, C. lipolytica, C. norvegensis, C. viswanathii, C. kefyr or C. dubliniensis), and Aspergillus spp. (for example A. fumigatus, A. flavus, A. niger or A. terreus).
  • the compounds having the formula (I) can be used in combination with one or more other medicaments.
  • the type of the other medicaments is not particularly limited and will depend on the disorder to be treated.
  • the other medicament will be a further medicament which is useful in treating, ameloriating or preventing a fungal infection, more preferably a further medicament which is useful in treating, ameloriating or preventing a fungal infection that is caused by a fungus selected from Candida spp. (for example C. albicans, C. auris, C. krusei, C. glabrata, C. tropicalis, C. parapsilosis, C. guilliermondii, C. haemulonii, C. lusitaniae, C.
  • Candida spp. for example C. albicans, C. auris, C. krusei, C. glabrata, C. tropicalis, C. parapsilosis, C. guilliermondii, C.
  • the fungal infection that can be treated, ameliorated or prevented with the compounds of the present invention is caused by a fungus selected from Candida spp. (for example C.
  • albicans C. auris, C. krusei, C. glabrata, C. tropicalis, C. parapsilosis, C. guilliermondii, C. haemulonii, C. lusitaniae, C. lipolytica, C. norvegensis, C. viswanathii, C. kefyr or C. dubliniensis), and Aspergillus spp. (for example A. fumigatus, A. flavus, A. niger or A. terreus).
  • compositions of the invention may be for treatment of a disseminated infection or a local infection.
  • the infection by a fungus may preferably be an infection involving at least partly infection of tissue, organs or cells with hypoxic conditions, preferably the infection may be infection of tissues, organs or cells with hypoxic conditions.
  • the infection may at least partly involve infection of one or more inner organs, tissues or cells of a mammal, preferably of a human being. More preferably, the infection may be infection of one or more inner organs, tissues or cells of a mammal, preferably of a human being.
  • the hypoxic condition is preferably an oxygen partial pressure (Po 2 ) of at the most 140 mmHg, preferably at the most 110 mmHg, such as at the most 80 mmHg.
  • Such conditions may in general be found in inner organs, for example in the liver, pancreas, gut, duodenum, skeletal muscles, brain, kidney or peritoneal cavity.
  • the infection by the fungus may also involve, at least partly, infection of a body surface.
  • Infection of a body surface may, for example, be infection of skin, nails or mucosal membranes of body surfaces.
  • the infection may be infection of a body surface, for example infection of skin, nails or mucosal membranes of body surfaces.
  • body surfaces may include the oral cavity, the genital organs, nose or eyes.
  • the infection by a fungus may be an infection by one species of fungus or an infection by more than one fungal species, such as 2, for example 3, such as 4, for example 5, such as more than 5 different fungal species.
  • the fungus may be any fungus, but usually it is a pathogenic fungus, such as a fungus pathogenic in the individual to be treated.
  • the individual to be treated is a human being, and then the fungus is a fungus pathogenic in human beings.
  • the fungus may also be selected from the Aspergillus spp., preferably from A. fumigatus, A. flavus, A. niger and A. terreus.
  • the fungus may also be selected from Cryptococcus spp., preferably from C. neoformans, C. bidus, C. laurentii, and C. fusarium.
  • the C. neoformans is preferably selected from var. neoformans and var. gattii.
  • the fungus may also be selected from zygomycetes, preferably the fungus is Saksenaea vasiformis.
  • the fungus may also be selected from Malassezia spp., preferably from M. furfur and globosa.
  • the fungus may also be selected from Dermatophytes. In particular this is the case when the infection is partly or entirely an infection of the skin.
  • the Dermatophyte may preferably be selected from Trichophyton spp., Epidermophyton floccosum, Microsporum spp and Trichosporon terrestre.
  • the Trichophyton spp. may preferably be selected from T. mentagrophytes, T. rubrum and T. tonsurans.
  • the Microsporum spp may preferably be selected from M. cookei, M. canis, M. vanbreuseghemii, M. gallinae and M. gypseum.
  • the fungus may also be selected from Chromomycotic fungi, preferably from Fonsecaea pedrosoi, F. compacta, Cladophylophora carrionii and Phialophora verrucosa.
  • the fungus may also be selected from Madurella spp., preferably from mycetomatis and M. griseum.
  • Combinations of more than one fungus can also be treated.
  • the compounds of the present invention are capable of inhibiting transcriptional activation of Olel protein.
  • the Olel protein is a A9-fatty acid desaturase, which converts stearic acid to oleic acid. Deletion of the Olel gene induces an oleic acid auxotrophy, which is lethal to the fungal cell.
  • Such activity has been previously demonstrated for the compound EV-086 that belongs to the prior art.
  • delivery of a compound into a fungal cell may represent a problem depending on, e.g., the solubility of the compound or its capabilities to cross the fungal cell membrane.
  • the present invention not only shows that the claimed compounds according to formula (I) have in vivo antifungal activity against common Candida albicans fungus, but that the claimed compounds show an improved in vivo antifungal activity against fungi that are resistant to fluconazole, including Candida parapsilosis, when compared to EV-086.
  • the compounds according to the formula (I) are capable of treating infections by fungi which are resistant to one or more conventional antifungal agents, in particular antifungal agents, which are not capable of inhibiting conversion of a saturated fatty acid to a A9-monounsaturated fatty acid in a fungus.
  • the fungi may be resistant for any reason.
  • particular species of fungus may be resistant to treatment with that particular antifungal agent.
  • the fungus may have acquired resistance, i.e. in general the fungal species is not resistant to treatment with the particular antifungal agent, but this particular fungus has become resistant.
  • the fungus has acquired resistance to one or more conventional antifungal agents.
  • the compounds according to formula (I) according to the invention and pharmaceutical compositions comprising the same are useful for treating infection by a fungus, which is resistant to one or more other antifungal agents.
  • the compounds according to formula (I) according to the invention and pharmaceutical compositions comprising the same are useful for treating an infection by a fungus, which is resistant to one or more antifungal agents capable of at least one of: a) inhibiting ergosterol biosynthesis; b) binding to ergosterol; c) inhibiting 1,3-P-glucan synthase; d) inhibiting epoxidase; e) inhibiting leucyl-tRNA synthetase; and/or f) inhibition of elongation factor 2.
  • the compounds according to formula (I) according to the invention and pharmaceutical compositions comprising the same are useful for treating infection by a fungus, which is resistant to one or more antifungal agents selected from the group consisting of polyene antifungal agents, azole antifungal agents, allylamine antifungal agents and echinocandins.
  • antifungal agents selected from the group consisting of polyene antifungal agents, azole antifungal agents, allylamine antifungal agents and echinocandins.
  • polyenes include Natamycin, Rimocidin, Filipin, Nystatin, Amphotericin B or Candicin.
  • Non-limiting examples of allylamine antifungals include Terbinafme, Amorolfme, Naftifme or Butenafme.
  • Non-limiting examples of echinocandins include Anidulafungin, Caspofungin or Micafungin.
  • compositions comprising compounds according to formula (I) of the invention may also be useful for treating infection by a fungus, which is resistant to one or more antifungal agents selected from the group consisting of benzoic acid, ciclopirox, tolnaftate, undecylenic acid, flucytosine, griseofulvin, haloprogin and sodium bicarbonate.
  • antifungal agents selected from the group consisting of benzoic acid, ciclopirox, tolnaftate, undecylenic acid, flucytosine, griseofulvin, haloprogin and sodium bicarbonate.
  • Compounds of formula (I) may demonstrate activity against more than one type of organism and are therefore particularly suitable and effective for administration to patients with more than one type of infection (e.g., a patient may have two types of fungal infections).
  • the pharmaceutical compositions for treating fungal infections according to the invention may in addition to one or more compounds according to formula (I) also comprise additional active agents, preferably one or more antifungal agents.
  • compositions may in addition to one or more compounds according to formula (I) also comprise one or more antifungal agents capable of at least one of: a) inhibiting ergosterol biosynthesis; b) binding to ergosterol; c) inhibiting 1,3-P-glucan synthase; d) inhibiting epoxidase; e) inhibiting leucyl-tRNA synthetase; and/or f) inhibition of elongation factor 2.
  • antifungal agents capable of at least one of: a) inhibiting ergosterol biosynthesis; b) binding to ergosterol; c) inhibiting 1,3-P-glucan synthase; d) inhibiting epoxidase; e) inhibiting leucyl-tRNA synthetase; and/or f) inhibition of elongation factor 2.
  • the additional antifungal agent may be for example be selected from the group consisting of polyene antifungal agents (such as any of the polyene antifungal agents described herein above in the section), azole antifungal agents (such as any of the azole antifungal agents described herein above in the section), allylamine antifungal agents (such as any of the allylamine antifungal agents described herein above in the section) and echinocandins (such as any of the echinocandins described herein above in the section).
  • polyene antifungal agents such as any of the polyene antifungal agents described herein above in the section
  • azole antifungal agents such as any of the azole antifungal agents described herein above in the section
  • allylamine antifungal agents such as any of the allylamine antifungal agents described herein above in the section
  • echinocandins such as any of the echinocandins described herein above in the section.
  • the compounds of the present invention can be synthesized by one of the general methods shown in the following scheme. These methods are only given for illustrative purposes and should not to be construed as limiting.
  • Brominated alkyne precursors bearing the R 3 substituent are either sourced commercially or prepared according to the methods known to a skilled person, as exemplified below.
  • Compounds bearing a diyne moiety can be prepared by a Pd-catalyzed and Cu-catalyzed coupling reaction or a Cu-catalyzed coupling reaction, according to one of two general synthetic routes according to Scheme 1, depending on the position of the bromine atom on one of the two possible alkyne components.
  • R 3 being -COOH or its methyl ester is then derivatized so that compounds according to formula (I) wherein other R 3 moieties are present are obtained.
  • the Cu-catalyzed coupling reaction was typically performed as follows: a brominated alkyne derivative (pure or solution in THF or solution in ether) was added to a mixture of alkyne derivative, copper chloride, hydroxylamine hydrochloride and ethyl amine (70%-w/w in water) in methanol at 0-30°C and the reaction mixture was stirred at 0-30°C until complete conversion was reached. Work-up and purification of the respective isolated product bearing a diyne moiety were done using appropriate methods.
  • the filtrate was concentrated under reduced pressure and passed through a pad of silica gel using diethyl ether as eluent.
  • the filtrate was evaporated under reduced pressure and the residue was resuspended in n-pentane (10 mL), filtrated and concentrated under reduced pressure, 1b was obtained with 44 % yield as a colorless condensed n-pentane solution.
  • the mixture was diluted with 10 mL water and extracted with diethyl ether (3 x 10 ml). The combined organic layers were washed with brine (10 mL), dried over Na 2 SO 4 and concentrated under reduced pressure.
  • the intermediate was purified by flash chromatography (SiO 2 , gradient cyclohexane-ethyl acetate). LiOH ⁇ H 2 O (0.39 mmol, 3.0 equiv.) was added to a solution of the purified intermediate in THF/MeOH/water (3 mL, 6:1:1) at 0°C. The solution was allowed to warm to rt and stirred for 3h before water (10 mL) was added.
  • the mixture was acidified with 3M HC1 and extracted with diethyl ether (3 x 10 mL). The combined organic layers were washed with brine (10 mL), dried over Na 2 SO 4 and concentrated under reduced pressure.
  • the crude product was purified by flash chromatography (SiO 2 , gradient cyclohexane-ethyl acetate; reversed phase C 18, gradient 10-100 % methanol in water). 1 was obtained with 20 % yield as a colorless oil.
  • ED AC (0.18 mmol, 1.2 equiv.) was added to a solution of EV-086 (0.15 mmol, 1.0 equiv.), hydroxylamine » HCl (0.15 mmol, 1.0 equiv.), HOBt monohydrate (0.18 mmol, 1.2 equiv.) and N-methylmorpholine (0.33 mmol, 2.2 equiv.) in DCM (2 mL) and the mixture was stirred for 3h at rt. The reaction mixture was diluted with water (10 mL) and extracted with DCM (3 x 10 mL). The combined organic layers were dried over Na 2 SO 4 and concentrated under reduced pressure. The crude product was purified by flash chromatography (SiO 2 , gradient cyclohexane- ethyl acetate). 4 was obtained with 18 % yield as an off-white solid.
  • ED AC (0.165 mmol, 1.1 equiv.) was added to a solution of EV-086 (0.15 mmol, 1.0 equiv.), methoxyamine*HCl (0.11 mmol, 1.1 equiv.), 1-hydroxybenzotriazole (HOBt) monohydrate (0.165 mmol, 1.1 equiv.) and triethylamine (TEA) (0.33 mmol, 2.2 equiv.) in DCM (2 mL) and the mixture was stirred for 3h at rt. The reaction mixture was diluted with water (10 mL) and extracted with DCM (3 x 10 mL). The combined organic layers were dried over Na 2 SO 4 and concentrated under reduced pressure. The crude product was purified by flash chromatography (SiO 2 , gradient cyclohexane-ethyl acetate). 5 was obtained with 75 % yield as a brown solid.
  • TEA 0.182 mmol, 1.3 equiv.
  • 2- chloro[l,3,2]dioxaphospholane (0.14 mmol, 1.0 equiv.) and 7a (0.14 mmol, 1.0 equiv.) in anhydrous toluene (1 mL) at 0°C under argon atmosphere.
  • the mixture was stirred for 4 h at rt.
  • additional chloro[l,3,2]dioxaphospholane (1.96 mmol, 1.4 equiv.) and TEA (1.96, 1.4 equiv.) was added and the mixture was stirred for 2 h at rt.
  • the mixture was diluted with 10 mL water, acidified with 2M HC1 and extracted with diethyl ether (3 x 10 mL). The combined organic layers were washed with brine (10 mL), dried over Na 2 SO 4 and concentrated under reduced pressure.
  • the crude product was purified by flash chromatography (SiO 2 , gradient cyclohexane-ethyl acetate; reversed phase C 18, gradient 10-100 % methanol in water). 7 was obtained with 19 % yield as a brown solid.
  • Triton B 50% in methanol, 50 ⁇ L was placed in a dried flask and the solvent was removed under reduced pressure. 7-Octyne-l-ol (2.75 mmol, 1.1 equiv.) was added, followed by methyl acrylate (2.5 mmol, 1.0 equiv.) and the mixture was stirred at 50°C for 3 h. The mixture was filtered over a Celite / silica gel mixture with diethyl ether. The filtrate was concentrated under reduced pressure and the crude intermediate was dissolved in THF/water (10 mL, 1:1).
  • XtalFluor-E (0.127 mmol, 2.02 equiv.) was added to a stirred solution of TEA ⁇ 3 HF (0.139 mmol, 2.2 equiv.) and TEA (0.064 mmol, 1.02 equiv.) in DCM (lmL) at -72°C under argon atmosphere. After stirring for 10 min, a solution of 9c (0.063 mmol, 1.0 equiv.) in DCM (0.3 mL) was added dropwise to the above solution. The reaction mixture was stirred for lh at -72°C and was then allowed to warm to rt and stirred for another 3h.
  • the reaction was quenched with 5 % NaHCO 3 solution (10 mL) and the aqueous layer was extracted with diethyl ether (3 x 10 mL). The combined organic layers were dried over Na 2 SO 4 and concentrated under reduced pressure.
  • the crude product was purified by flash chromatography (SiO 2 , gradient cyclohexane-ethyl acetate). 9d was obtained with 74 % yield as a colorless oil.
  • Step E LiOH ⁇ H 2 O (0.14 mmol, 3.0 equiv.) was added to a solution of 9d (0.047 mmol, 1.0 equiv.) in THF/MeOH/water (3 mL, 6: 1 : 1) at 0°C. The solution was allowed to warm to rt and stirred for 3.5h before water (10 mL) was added. The pH was adjusted to pH 2 with 2M HC1. 10 mL diethyl ether was added and the layers were separated. The aqueous layer was extracted with diethyl ether (2 x 10 mL) and the combined organic layers were washed with brine (10 mL), dried over Na 2 SO 4 and concentrated under reduced pressure. The crude product was purified by flash chromatography (SiO 2 , gradient cyclohexane-ethyl acetate). 9 was obtained with 84 % yield as a white solid.
  • the combined organic layers were washed with brine (10 mL), dried over Na 2 SO 4 and concentrated under reduced pressure.
  • the intermediate was purified by flash chromatography (SiO 2 , gradient cyclohexane- ethyl acetate). LiOH ⁇ H 2 O (0.6 mmol, 3.0 equiv.) was added to a solution of the purified intermediate in THF/MeOH/water (5 mL, 6:1:1) at 0°C. The solution was allowed to warm to rt and stirred for 3.5h before water (10 mL) was added. The mixture was acidified with 3M HC1 and extracted with diethyl ether (3 x 10 mL).
  • the combined organic layers were washed with brine (10 mL), dried over Na 2 SO 4 and concentrated under reduced pressure.
  • the intermediate was purified by flash chromatography (SiO 2 , gradient cyclohexane- ethyl acetate). LiOH ⁇ H 2 O (0.6 mmol, 3.0 equiv.) was added to a solution of the purified intermediate in THF/MeOH/water (5 mL, 6:1:1) at 0°C. The solution was allowed to warm to rt and stirred for 3.5h before water (10 mL) was added. The mixture was acidified with 3M HC1 and extracted with diethyl ether (3 x 10 mL).
  • MIC values measured for the exemplary compounds are given in Table 1. All MIC-values reported herein are measured using the SOP of the The European Committee on Antimicrobial Susceptibility Testing (EUCAST - https://www.eucast.org/astoffungi, EUCAST E.DEF 7.3.1 for Candida spp. and EUCAST E.DEF 9.3.1 for Asperigillus spp).
  • Flat-bottom well microdilution plates are used to add 100 ⁇ L of antifungal test solution in double strength RPMI 1640 (with L-glutamine and a pH indicator but without bicarbonate) supplemented with glucose to a final concentration of 2% (RPMI 2% G medium) plus 100 ⁇ L fungal species inoculum obtained from an 18-24 h culture on nutritive agar medium, in sterile distilled water with a final inoculum between 0.5 x 105 and 2.5 x 105 CFU/mL.
  • a stock solution from powder test compound is prepared with RPMI 2% G medium to a concentration of 20.4 mg/L. 2-fold dilutions are prepared to 0.005mg/L and distributed to the microtiter plates.
  • Microdilution plates are incubated without agitation at 35 ⁇ 2 °C in ambient air for 48 h.
  • Cell growth after 24 h and / or 48 h is determined by a microplate reader by measuring the UV- absorbance at 530 nm.
  • EV-086 in concentrations of 10.2 mg/L, 0.04 mg/L and 0.005 mg/L are used as positive control and RPMI 2% G medium as negative control. Relative growth rates are calculated from one to three independent experiments and the compound concentration leading to a ⁇ 10% reduction of growth is reported as MIC-value.
  • Aspergillus flavus ATCC MYA-3631
  • Aspergillus fumigatus Fresenius ATCC MYA-3626
  • Trichophyton rubrum ATCC MYA-4438
  • Trichophyton interdigitale Priestley ATCC MYA-4439
  • PBS phosphate buffered saline

Abstract

The present invention refers to a compound having the formula (I): R1–Z–C≡C–C≡C–R3 (I) and pharmaceutical compositions comprising the same that can be used in prevention, treatment and alleviation of a fungal infection. The instant compounds and pharmaceutical compositions are particularly useful in treating, preventing and alleviating fungal infections caused by Candida fungi and Aspergillus fungi.

Description

Novel antifungal compounds
Field of the invention
The present invention relates to novel compounds and pharmaceutical compositions comprising the same that can be used in the prevention, treatment and alleviation of a fungal infection. The instant compounds and pharmaceutical compositions are particularly useful in treating, preventing and alleviating fungal infections caused by Candida fungi (including, but not limited to C. albicans, C. parapsilosis and C. krusei) and Aspergillus fungi (including, but not limited to A. fumigatus, A. flavus and A. niger).
Background of the invention
Fungal infections (mycoses) can contribute to and complicate many diseases, including pulmonary diseases and respiratory tract diseases. The incidence of fungal infections is steadily rising as a consequence of antibiotic treatments, an immune-suppressed or immune- compromised population (mainly caused by cancer treatment, HIV, allergy-treatments, transplantations and general surgery) and an aging population.
An estimated 15,000 allogenic and 25,000 autologous stem cell transplants are performed worldwide every year. In addition, from 1998 to 2002, 113,682 solid organ transplants were performed in the United States, which is a 20% increase over the previous 5-year period. Unfortunately, patients undergoing these life-saving procedures are at increased risk for fungal infections, for example, by Aspergillus fumigatus and other Aspergillus spp. due to their immuno-compromised condition. Additionally, the populations of immuno-compromised patients due to HIV, cancer therapy, surgical non-transplants and general ageing also continue to the increase and with it the number of cases of severe fungal infections, for example, of systemic candidiasis. Candida species account for 80% of infections in general medicine, 40% in HIV populations and 90% in both cancer therapy and surgical-non transplant cases. Candida is now the 4th largest cause of nosocomial blood stream infections. Mortality from systemic fungal infections remains high despite the development of new antifungal agents, and Candida bloodstream infections in the United States are associated with a 40% crude mortality rate. Since 1980, the mortality due to Aspergillus fumigatus is continually increasing.
Furthermore, there have been recent changes in the epidemiology of these systemic infections, with five species (C. albicans, C. glabrata, C. parapsilosis, C. tropicalis, and C. krusei) being responsible for more than 90% of invasive infections due to Candida spp. Candida fungi are the fourth most common cause of nosocomial bloodstream infections. While C. albicans was the predominant cause of Candida bloodstream infections in the early 80s, C. glabrata has emerged as the second most common cause of said infections in various parts of the world, including the United States. In addition to Aspergillus and Candida infections, other fungal pathogens such as Scedosporium spp. are becoming increasingly important. Their susceptibility to existing antifungals is limited and their mortality rate is >70% in patients with hematological malignancies. In other patients the mortality rates vary between 30 and 80%.
Onychomycosis is a fungal infection of the nails which is estimated to affect up to 25% of the geriatric and diabetic populations and 2 to 13% of the general US population. Common risk factors include age, male gender, diabetes, nail trauma, and chronic Tinea pedis (fungal infection of the foot). Onychomycosis has significant cosmetic, psychological and social implications. In some patient subsets it has serious medical consequences (e.g., foot amputations in diabetics).
The basic structure of fungal cells and human cells is similar, making it difficult to find a target for an anti-fungal drug that does not also exist in human. Consequently, currently available antifungal drugs have side-effects, some of which can be life- threatening. Currently available treatments are based on four major classes of drugs: polyenes (e.g., Amphotericin B) that bind to sterols in a fungal cell membrane, principally to ergosterol; imidazoles and triazoles (e.g., Fluconazole or Itraconazole) which inhibit cytochrome P450 14a-demethylase, which is required for ergosterol synthesis and homeostasis of fungal cell membrane; allylamines (e.g., Terbinafine) which inhibit the enzyme squalene epoxidase, another enzyme required for ergosterol synthesis; and echinocandins (e.g., Caspofungin) that inhibit the synthesis of glucan in the cell wall. These drugs exhibit a poor efficacy rate and are associated with serious side effects. Amphotericin B is nephrotoxic. Terbinafine is associated with liver damage. Furthermore, there is generalized intolerance to azoles, for example, up to 20% of females with vaginal candidiosis cannot tolerate Fluconazole. Moreover, many pathogenic strains are insensitive or resistant to currently available antifungal drugs and resistance development is a concern. Thus, mortality due to fungal infections remains high.
Caspofungin resistance is relatively rare with ca. 8% of C. tropicalis and ca. 2% of C. glabrata isolates having been classified as resistant (corresponding MIC values of > 2mg/L). Given the observation that 2001 to 2004 surveillance studies identified > 99.5% of patients as Caspofungin sensitive, it is disconcerting how rapidly echinocandin resistance is spreading. Furthermore, there have been reported cases of reduced C. glabrata susceptibility developing during Caspofungin therapy. The target of Caspofungin is the enzyme 1,3-P-D-glucan synthase, encoded by one of several FKS genes, depending on the species. In clinical isolates, mutations in the FKS 1 gene resulting in amino acid changes in the protein were shown to be necessary and sufficient to confer reduced susceptibility to Caspofungin. Candida spp. with reduced susceptibility to the newer members of the echinocandin family have also been reported.
In terms of resistance, for the azoles alone, three different resistance mechanisms have been identified: a) alternative pathways for the synthesis of cell membrane sterols, b) mutations in the target demethylase site and c) increased efflux of drug from the fungal cell.
WO 01/25197 refers to certain substantially pure ene-diyne compounds bearing a carboxylic group or a moiety that can be hydrolyzed to a carboxylic group, which are useful as antifungal agents.
WO 2011/006061 refers to certain substantially pure cis-isomer compounds bearing an ene- diyne moiety that are useful as Olel protein inhibitors and can be applied as antifungal agents.
WO 2011/134538 discloses specific diyne compounds substituted with an optionally substituted carbon chain bearing a -COO- group and with an optionally substituted heteroaromatic ring that can be used in fungicidal formulations.
EV-086, the natural diyne-furan fatty acid compound of formula:
Figure imgf000005_0001
is described as an inhibitor of fungal Δ-9 fatty acid desaturation, which shows efficacy in a model of skin dermatophytosis (Antimicrobial Agents and Chemotherapy, 2014, 58, 455-456).
Summary of the invention
It was an object of the present invention to provide novel compounds that can be employed in therapy, amelioration and prevention of fungal infections. In particular, the compounds should exhibit broad antifungal activity, in other words be suitable for treating fungal infections caused by diverse fungi.
The present inventors have surprisingly found that this object can be solved by the compounds having the formula (I). Among others, the compounds of the present invention are useful in treating infections caused by Candida fungi such as C. albicans, C. parapsilosis, C. tropicalis and C. glabrata. The compounds of the present invention are also useful in treating the infections caused by Aspergillus fungi such as A. fumigatus, A. flavus, A. niger and A. terreus.
Furthermore, there exists a need in the art for compounds which can be used against fungi that have developed resistance against existing antifungal therapies. One example of such fungus is Candida parapsilosis, responsible for 30% cases of Candida infections in the south of Europe. Candida parapsilosis is also the main Candida species, apart from Candida albicans, isolated from patients in South America. Due to mutations in Ergl 1 gene and overexpression of transporter proteins, Candida parapsilosis is resistant against treatment with fluconazole, an oral antifungal medication considered to be a standard of care. As demonstrated in the examples, compounds of formula (I) are active against Candida parapsilosis, and have surprisingly shown improved efficacy against Candida parapsilosis as compared with the state of the art compound EV-086. Furthermore, the compounds of formula (I) have surprisingly shown improved efficacy against Aspergillus fungi, for example, against A. flavus and A. fumigatus, as compared to the state-of-the-art compound EV-086.
It is also desirable to provide novel antifungal compounds with improved pharmaceutical properties. As demonstrated in the examples, compounds of formula (I) show surprisingly improved stability in plasma compared to the state-of-the-art compound EV-086.
The present invention can be summarized in the following aspects.
In one aspect, the present invention relates to a compound having the formula (I):
R1-Z-C≡C-C≡C-R3
(I) and all stereoisomers, racemic mixtures, tautomers, pharmaceutically acceptable salts, prodrugs, hydrates, solvates and polymorphs thereof, wherein
R1 is selected from HO-NH(O=)C-, C1-4 alkyl-O-NH(O=)C-, C 1 -4 alkyl-SO2-NH-(O=)C- ,C1-4 alkyl-O-C(=O)-O- C1-4 alkyl-C(=O)-O-CH2-O-(O=)C-,
C1-4 alkyl-NH-(O=)C-, C1-4 alkyl-C(=O)-NH-CH2-NH-
C1-4 alkyl-C(=O)NH-C(=O)-NH-, C1-4 alkyl-SO2-NH-C(=O)-NH-,
C1-4 alkyl-C(=O)N(OH)-, C1-4 alkyl-O-C(=O)-NH-, C1-4 alkyl-C(=O)-NH-O- C1-4 alkyl-NH-C(=O)-O-, CF3CH(OH)-, C1-4 alkyl-SO2NH-
Figure imgf000006_0001
Figure imgf000007_0001
Z represents an alkylene moiety having 6 to 12 carbon atoms and optionally containing 1 or 2 double bonds, wherein
(i) one or more non-adjacent -CH2- groups can be independently replaced by -O-, -S- -S(O)-, -C(O)-N(H)-, or-C(O)-N(C1-4 alkyl)- and/or
(ii) one or more hydrogen atoms can be independently replaced by -F, -OH or C1-4 alkyl, and/or
(iii) one or more -CH2-CH2- moieties can be independently replaced by a catenary 3- to 6- membered heterocyclic ring, and/or
(iv) one or more -CH2-CH2-CH2- moieties can be independently replaced by a catenary 3- to 6-membered heterocyclic ring; and
R3 is selected from
(i) an optionally substituted heterocyclic or carbocyclic group having 5 to 10 ring atoms, wherein the optional substituent is independently selected from -C1-4 alkyl, -OH, -OC1-4 alkyl, -Hal, -NH2, -NH(C1-4 alkyl), -N(C1-4 alkyl)2, and -NO2, or
(ii) an alkyl group containing 1 to 4 carbon atoms, wherein one divalent carbon atom can be replaced by oxygen or sulfur and wherein the alkyl group R3 can be optionally substituted by one or more Hal. In a further aspect, the present invention relates to the compound of formula (I), wherein R1 is selected from HO-NH(O=)C-, C1-4 alkyl-O-NH(O=)C-, C1-4 alkyl-SO2-NH-(O=)C-,and
Figure imgf000008_0001
In yet a further aspect, the present invention relates to the compound of formula (I), wherein Z is -(CH2)7-CH=CH- or -(CH2)7-CH2-CH2- preferably -(CH2)7-CH=CH-.
In yet a further aspect, the present invention relates to the compound of formula (I), wherein R3 is optionally substituted as defined in claim 1 and is selected from furane, thiophene, norbomane, cyclopropyl, pyrrole, oxazole, isoxazole, thiazole, isothiazole, pyrazole, and imidazole, preferably R3 is furane.
In yet a further aspect, the present invention relates to the compound of formula (I), wherein the compound is selected from:
Figure imgf000008_0002
Another aspect of the present invention relates to a compound of formula (I): R1-Z-C≡C-C≡C-R3
(I) and all stereoisomers, racemic mixtures, tautomers, pharmaceutically acceptable salts, prodrugs, hydrates, solvates and polymorphs thereof, wherein
R1 is selected from -COOH, a moiety that can be hydrolyzed to -COOH, HO-NH(O=)C-, C1-4 alkyl-O-NH(O=)C-, C1-4 alkyl-SO2-NH-(O=)C-, C1-4 alkyl-O-C(=O)-O-, C1-4 alkyl-C(=O)-O-CH2-O-(O=)C-, C1-4 alkyl-NH-(O=)C- C1-4 alkyl-C(=O)-NH-CH2-NH-, C1-4 alkyl-C(=O)NH-C(=O)-NH-
C1-4 alkyl-SO2-NH-C(=O)-NH-, C1-4 alkyl-C(=O)N(OH)-, C1-4 alkyl-O-C(=O)-NH- C1-4 alkyl-C(=O)-NH-O-, C1-4 alkyl-NH-C(=O)0-, CF3CH(OH)-, C1-4 alkyl-SO2NH-
Z represents an alkylene moiety having 6 to 12 carbon atoms and optionally containing 1 or 2 double bonds, wherein (i) one or more non-adjacent -CH2- groups can be independently replaced by -O-, -S-, - S(O)-, -C(O)-N(H)-, or -C(O)-N(C1-4 alkyl)- and/or
(ii) one or more hydrogen atoms can be independently replaced by -F, -OH or C1-4 alkyl, and/or
(iii) one or more -CH2-CH2- moieties can be independently replaced by a catenary 3- to 6- membered heterocyclic ring, and/or
(iv) one or more -CH2-CH2-CH2- moieties can be independently replaced by a catenary 3- to 6-membered heterocyclic ring; and
R3 represents an aliphatic group containing 1 to 4 carbon atoms, wherein one divalent carbon atom can be replaced by oxygen or sulfur and wherein the alkyl group R3 can be optionally substituted by one or more Hal.
In a further aspect, the present invention relates to the compound of formula (I), wherein R1 is selected from HO-NH(O=)C-, C1-4 alkyl-O-NH(O=)C-, C1-4 alkyl-SO2-NH-(O=)C-,and
Figure imgf000010_0001
In another further aspect, the present invention relates to the compound of formula (I), wherein R1 is selected from -COOH, -COONa and -COOK.
In yet a further aspect, the present invention relates to the compound of formula (I), wherein Z is -(CH2)7-CH=CH-or -(CH2)7-CH2-CH2-, preferably -(CH2)7-CH=CH-.
In yet a further aspect, the present invention relates to the compound of formula (I), wherein R3 is selected from -CH2CH2CH3, -CH2OCH3, -CH2SCH3, -CHFCH2CH3, -CF2CH2CH3, preferably R3 is -CH2CH2CH3.
In yet a further aspect, the present invention relates to the compound of formula (I), wherein R3 is cyclopropyl or cyclobutyl. In another further aspect, the present invention relates to the compound of the present invention, wherein the compound has a minimum inhibitory concentration of at most 1.0 mg/L against Candida albicans.
Another aspect of the present invention relates to the compound of the present invention for use as a medicament.
A yet another aspect of the present invention relates to a pharmaceutical composition comprising the compound of the present invention and optionally a pharmaceutically acceptable carrier.
Another aspect of the present inventions relates to the compound of the present invention for use in the treatment, alleviation or prevention of a fungal infection.
A yet another aspect of the present invention relates to a use of the compound of the present invention for the preparation of a medicament for treating, alleviating or preventing a fungal infection.
Another aspect of the present invention relates to a method of treating, alleviating or preventing a fungal infection, the method comprising administering an effective amount of the compound of the invention to a subject in need thereof.
The fungal infection can be caused by a fungus selected from Candida spp. (for example C. albicans, C. auri, C. krusei, C. glabrata, C. tropicalis, C. parapsilosis, C. guilliermondii, C. haemulonii, C. lusitaniae, C. lipolytica, C. norvegensis, C. viswanathii, C. kefyr or C. dubliniensis), Aspergillus spp. (for example A.fumigatus, A.flavus, A. niger or A. terreus) Histoplasma capsulatum, Coccidioides immitis, Coccidioides posadasii, Cryptococcus spp. (for example C. neoformans (for example var. neoformans or var. gattii), C. bidus, C. laurentii, or C.fusarium), Zygomycetes (such as Saksenaea vasiformis), Malassezia spp. (for example M. furfur or M. globosa), Hyalohyphomycetes (for example Scedosporium spp., such as S. prolificans or S. apiospermum), Dermatophytes (for example Trichophyton spp. (for example T. mentagrophytes, T. rubrum or T. tonsurans), Epidermophyton floccosum, Microsporum spp (for example M. cookei, M. canis, M. vanbreuseghemii, M. gallinae or M. gypseum) or Trichosporon terrestre), Blastomyces dermatitidis, Sporothrix schenkii, Chromomycotic fungi (for example Fonsecaea pedrosoi, F. compacta, Cladophylophora carrionii or Phial ophora verrucosa) and Madurella spp. (for example M. mycetomatis or M. griseum), Pneumocystis jirovecii, and Pneumocystis carinii. In a preferred embodiment, the fungal infection can be caused by a fungus selected from Candida fungi (including, but not limited to C. albicans, C. parapsylosis and C. krusei) and Aspergillus fungi (including, but not limited to A. fumigatus, A. flavus and A. niger).
The compounds, which are disclosed in the examples or in claims as well as stereoisomers, racemic mixtures, pharmaceutically acceptable salts, hydrates, solvates, prodrugs and polymorphs thereof, are considered to be particularly suitable compounds of the present invention.
Definitions
Within the meaning of the present application the following definitions apply:
The term "alkyl" refers to a saturated straight or branched organic moiety consisting of carbon and hydrogen atoms. Examples of suitable alkyl groups have 1 to 6 carbon atoms, preferably 1 to 4 carbon atoms, and include methyl, ethyl, propyl, isopropyl, n-butyl, t-butyl and isobutyl. An alkyl group referred to as C1-4 alkyl is an alkyl group having 1 to 4 carbon atoms.
The term "heterocyclic group" covers any mono-, bi- or polycyclic ring system which includes one or more heteroatoms in the ring system, whereby the heteroatoms are the same or different and are selected from O, N and S. Preferably the ring system includes 3 to 15 ring atoms. More preferably the ring system is mono- or bicyclic and has 5 to 10 ring atoms, even more preferably the ring system is monocyclic and has 5 or 6 ring atoms. Typically, the ring system can include 1 to 4, more typically 1 or 2 heteroatoms at available positions. The term "heterocyclic group" also covers heteroaryl rings. The monocyclic heterocyclic group may also be referred to as a heterocyclic ring. Examples include azetidine, pyrrole, pyrrolidine, oxolane, furan, imidazolidine, imidazole, pyrazole, oxazolidine, oxazole, thiazole, piperidine, pyridine, morpholine, piperazine, and dioxolane. The term "heteroaryl" preferably refers to a five or six-membered aromatic ring wherein one or more of the carbon atoms in the ring have been replaced by 1, 2, 3, or 4 (for the five membered ring) or 1, 2, 3, 4, or 5 (for the six membered ring) of the same or different heteroatoms, whereby the heteroatoms are selected from O, N and S. Examples of the heteroaryl group include furane, thiophene, pyrrole, oxazole, isoxazole, isothiazole, thiazole, pyrazole, imidazole, 1,2,4-triazole, 1,2, 3 -triazole, 1,2,5-oxadiazole, 1,2,3-oxadiazole, 1,3,4-thiadiazole, 1,2,5-thiadiazole, tetrazole, pyridine, pyridazine, pyrimidine, pyrazine.
The term "carbocyclic group" covers any mono-, bi- or polycyclic ring system which does not include heteroatoms in the ring. Preferably the ring system includes 3 to 15 ring atoms. More preferably the ring system is mono- or bicyclic and has 5 to 10 ring atoms, even more preferably the ring system is monocyclic and has 5 or 6 ring atoms. The monocyclic carbocyclic group may also be referred to as a carbocyclic ring. A further example of a "carbocyclic group" or a “carbocyclic ring” is a C3 -6 cycloalkyl ring. The term "carbocyclic group" also covers aryl rings.
The term "aryl" preferably refers to an aromatic monocyclic ring containing 5 or 6 carbon atoms, an aromatic bicyclic ring system containing 10 carbon atoms or an aromatic tricyclic ring system containing 14 carbon atoms. Examples are phenyl, naphthyl or anthracenyl, preferably phenyl.
The term "halogen" or "Hal" covers F, Cl, Br and I, preferably F or Cl.
The term “moiety that can be hydrolyzed to -COOH” is any moiety that upon reaction with water yields -COOH moiety. Preferred examples of such moiety include ester -C(=O)-O-alkyl (for example methyl ester), amide -C(=O)-NH2 , N-substituted amide - C(=O)-NH-alkyl, N, N-substituted amide -C(=O)-N(alkyl)2, and nitryle -C≡N.
Compounds of the present invention having one or more optically active carbons can exist as racemates and racemic mixtures, stereoisomers (including diastereomeric mixtures and individual diastereomers, enantiomeric mixtures and single enantiomers, mixtures of conformers and single conformers), tautomers, atropisomers, and rotamers. All isomeric forms are included in the present invention. Compounds described in this invention containing olefmic double bonds include E and Z geometric isomers, unless stated otherwise. Also included in this invention are all salt forms, polymorphs, hydrates and solvates. If a compound or moiety is referred to as being "optionally substituted" it can in each instance include one or more of the indicated substituents, whereby the substituents can be the same or different.
The term "polymorphs" refers to the various crystalline structures of the compounds of the present invention. This may include, but is not limited to, crystal morphologies (and amorphous materials) and all crystal lattice forms. Salts of the present invention can be crystalline and may exist as more than one polymorph.
Solvates, hydrates as well as anhydrous forms of the salt are also encompassed by the invention. The solvent included in the solvates is not particularly limited and can be any pharmaceutically acceptable solvent. Examples include water and C1-4 alcohols (such as methanol or ethanol).
"Pharmaceutically acceptable salts" are defined as derivatives of the disclosed compounds wherein the parent compound is modified by making acid or base salts thereof. 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 carboxylic acids; and the like. The pharmaceutically acceptable salts include the conventional non-toxic salts or the quaternary ammonium salts of the parent compound formed, for example, from nontoxic inorganic or organic acids. For example, such conventional non-toxic salts include those derived from inorganic acids such as, but not limited to, hydrochloric, hydrobromic, sulfuric, sulfamic, phosphoric, nitric and the like; and the salts prepared from organic acids such as, but not limited to, acetic, propionic, succinic, glycolic, stearic, lactic, malic, tartaric, citric, ascorbic, pamoic, maleic, hydroxymaleic, phenylacetic, glutamic, benzoic, salicylic, sulfanilic, 2- acetoxybenzoic, fumaric, toluenesulfonic, methanesulfonic, ethane disulfonic, oxalic, isethionic, and the like. The pharmaceutically acceptable salts of the present invention can be synthesized from the parent compound which contains a basic or acidic moiety by conventional chemical methods. Generally, such salts can be prepared by reacting the free acid or base forms of these compounds with a stoichiometric amount of the appropriate base or acid in water or in an organic solvent, or in a mixture of the two. Organic solvents include, but are not limited to, nonaqueous media like ethers, ethyl acetate, ethanol, isopropanol, or acetonitrile. Lists of suitable salts can be found in Remington’s Pharmaceutical Sciences, 18th ed., Mack Publishing Company, Easton, PA, 1990, p. 1445, the disclosure of which is hereby incorporated by reference.
"Pharmaceutically acceptable" is defined as those compounds, materials, compositions, and/or dosage forms which are, within the scope of sound medical judgment, suitable for use in contact with the tissues of human beings and animals without excessive toxicity, irritation, allergic response, or other problem or complication commensurate with a reasonable benefit/risk ratio.
The compounds of the present invention can also be provided in the form of a prodrug, namely a compound which is metabolized in vivo to the active metabolite. The reference by Goodman and Gilman (The Pharmacological Basis of Therapeutics, 8 ed, McGraw-Hill, Int. Ed. 1992, "Biotransformation of Drugs", p 13-15) describing prodrugs generally is hereby incorporated herein by reference.
The patients or subjects in the present invention are typically animals, particularly mammals, more particularly humans.
The preferred definitions given in the "Definition"-section apply to all of the embodiments described below unless stated otherwise.
Description of the Figures and Tables
Figure 1: Table 1: Summary of the studies of antifungal properties of the exemplary compounds 1 to 14 of the present invention against selected fungi. It is noted that compound 3 is shown as a reference example.
Figure 2: The results of the measurement of the plasma stability of the antifungal compounds 5, 8, 10, 13 and EV-086. The ID of the compound is shown on the x-axis and the recovery rate on the y-axis. The error bars indicate the standard errors of the measured triplicates. For each compound there are four bars which show the recovery rates of the measurements of the four timepoints 0, 60, 120 and 180 minutes (T0 to T3, respectively).
Detailed description of the invention The compounds of the present invention will be described in the following. It is to be understood that all possible combinations of the following definitions are also envisaged.
In one embodiment, the present invention relates to a compound of formula (I):
R1-Z-C≡C-C≡C-R3:
(I) and all stereoisomers, racemic mixtures, tautomers, pharmaceutically acceptable salts, prodrugs, hydrates, solvates and polymorphs thereof.
R1 is selected from HO-NH(O=)C-, C1-4 alkyl-O-NH(O=)C-, C1-4 alkyl-SO2-NH-(O=)C-
,C1-4 alkyl-O-C(=O)-O- C1-4 alkyl-C(=O)-O-CH2-O-(O=)C-
C1-4 alkyl-NH-(O=)C-, C1-4 alkyl-C(=O)-NH-CH2-NH-
C1-4 alkyl-C(=O)NH-C(=O)-NH- C1-4 alkyl-SO2-NH-C(=O)-NH-
C1-4 alkyl-C(=O)N(OH)-, C1-4 alkyl-O-C(=O)-NH-, C1-4 alkyl-C(=O)-NH-O-,
C1-4 alkyl-NH-C(=O)-O-, CF3CH(OH)-, C1-4 alkyl-SO2NH-,
Figure imgf000016_0001
Figure imgf000016_0002
selected from HO-NH(O=)C-, C1-4 alkyl-O-NH(O=)C-, C1-4 alkyl-SO2-NH-(O=)C- and
Figure imgf000017_0001
Z represents an alkylene moiety having 6 to 12 carbon atoms and optionally containing 1 or 2 double bonds, wherein
(i) one or more non-adjacent -CH2- groups can be independently replaced by -O-, -S-, -S(O)-, -C(O)-N(H)-, or -C(O)-N(C1-4 alkyl)-, and/or
(ii) one or more hydrogen atoms can be independently replaced by -F, -OH or C 1-4 alkyl, and/or one or more -CH2-CH2- moieties can be independently replaced by a catenary 3- to 6-membered heterocyclic ring, and/or
(iii) one or more -CH2-CH2-CH2- moieties can be independently replaced by a catenary 3- to 6-membered heterocyclic ring.
Preferably, Z is -(CH2)7-CH=CH- or -(CH2)7-CH2-CH2- More preferably, Z is -(CH2)7-CH=CH-, Preferably, the double bond of-(CH2)7-CH=CH- has a (Z) -configuration.
R3 is selected from
(i) an optionally substituted heterocyclic or carbocyclic group having 5 to 10 ring atoms, wherein the optional substituent is independently selected from — C 1-4 alkyl, -OH, -OC1-4 alkyl, -Hal, -NH2, -NH(C1-4 alkyl), -N(C1-4 alkyl)2, and -NO2, or
(ii) an alkyl group containing 1 to 4 carbon atoms, wherein one divalent carbon atom can be replaced by oxygen or sulfur and wherein the alkyl group R3 can be optionally substituted by one or more Hal.
Preferably, R3 is selected from an optionally substituted furane, thiophene, norbornane, cyclopropyl, pyrrole, oxazole, isoxazole, thiazole, isothiazole, pyrazole, and imidazole, wherein the optional substituent is independently selected from — C 1-4 alkyl, -OH, -OC1-4 alkyl, -Hal, -NH2, -NH(C1-4 alkyl), -N(C 1-4 alkyl)2, and -NO2. More preferably R3 is furane.
It is understood that all combinations of the above definitions and preferred definitions are also envisaged. The compound according to formula (I) can be, for instance, a compound according to formula (la)
R1-(CH2)7-CH=CH-C≡C-C≡C-R3
(la) and all stereoisomers, racemic mixtures, tautomers, pharmaceutically acceptable salts, prodrugs, hydrates, solvates and polymorphs thereof, wherein R1 and R3 are as defined herein. In a preferred embodiment, R1 is selected from HO-NH(O=)C-, C1-4 alkyl-O-NH(O=)C-,
C1-4 alkyl-SO2-NH-(O=)C-,and
Figure imgf000018_0001
Preferably, R3 is selected from an optionally substituted furane, thiophene, norbornane, cyclopropyl, pyrrole, oxazole, isoxazole, thiazole, isothiazole, pyrazole, and imidazole, wherein the optional substituent is independently selected from —C1-4 alkyl, -OH, -OC1-4 alkyl, -Hal, -NH2, -NH(C1-4 alkyl), -N(C1-4 alkyl)2, and -NO2. More preferably R3is furane.
Preferably, the double bond of -(CH2)7-CH=CH- has a (Z) -configuration.
Thus, preferably, the compound according to formula (la) can be a compound according to formula (lb)
Figure imgf000018_0002
and all stereoisomers, racemic mixtures, tautomers, pharmaceutically acceptable salts, prodrugs, hydrates, solvates and polymorphs thereof, wherein R1 is as defined herein. In a preferred embodiment, R1 is selected from HO-NH(O=)C-, C1-4 alkyl-O-NH(O=)C-
C1-4 alkyl-SO2-NH-(O=)C-,and
Figure imgf000018_0003
Preferably, the double bond of -(CH2)7-CH=CH- has a (Z) -configuration. The compounds according to formula (lb) are preferably selected from:
Figure imgf000019_0001
In another embodiment, the present invention relates to a compound of formula (I) and all stereoisomers, racemic mixtures, tautomers, pharmaceutically acceptable salts, prodrugs, hydrates, solvates and polymorphs thereof, wherein R1 is selected from -COOH, a moiety that can be hydrolyzed to -COOH, HO-NH(O=)C-, C1-4 alkyl-O-NH(O=)C-
C1-4 alkyl-SO2-NH-(O=)C-,C1-4 alkyl-O-C(=O)-O-
C1-4 alkyl-C(=O)-O-CH2-O-(O=)C-, C1-4 alkyl-NH-(O=)C-
C1-4 alkyl-C(=O)-NH-CH2-NH-, C1-4 alkyl-C(=O)NH-C(=O)-NH-
C1-4 alkyl-SO2-NH-C(=O)-NH-, C1-4 alkyl-C(=O)N(OH)-, C1-4 alkyl-O-C(=O)-NH- C1-4 alkyl-C(=O)-NH-O-, C1-4 alkyl-NH-C(=O)O-, CF3CH(OH)-, C1-4 alkyl-SO2NH-,
Figure imgf000019_0002
Figure imgf000020_0001
preferred embodiment, R1 is selected from HO-NH(O=)C-, C1-4 alkyl-O-NH(O=)C-,
C1-4 alkyl-SO2-NH-(O=)C-,and In another preferred embodiment, R
Figure imgf000020_0002
1 is selected from -COOH, -COONa and -COOK.
Z represents an alkylene moiety having 6 to 12 carbon atoms and optionally containing 1 or 2 double bonds, wherein
(i) one or more non-adjacent -CH2- groups can be independently replaced by -O-, -S-, -S(O)-, -C(O)-N(H)-, or-C(O)-N(C1-4 alkyl)-, and/or
(ii) one or more hydrogen atoms can be independently replaced by -F, -OH or C1-4 alkyl, and/or one or more -CH2-CH2- moieties can be independently replaced by a catenary 3- to 6-membered heterocyclic ring, and/or
(iii) one or more -CH2-CH2-CH2- moieties can be independently replaced by a catenary 3- to 6-membered heterocyclic ring.
Preferably, Z is -(CH2)7-CH=CH- or -(CH2)7-CH2-CH2- More preferably, Z is -(CH2)7-CH=CH- Preferably, the double bond of-(CH2)7-CH=CH- has a (Z) -configuration.
R3 represents an aliphatic group containing 1 to 4 carbon atoms, wherein one divalent carbon atom can be replaced by oxygen or sulfur and wherein the alkyl group R3 can be optionally substituted by one or more Hal. Preferably, R3 is selected from -CH2CH2CH3. -CH2OCH3, -CH2SCH3, -CHFCH2CH3, -CF2CH2CH3. More preferably, R3 is -CH2CH2CH3. In another preferred embodiment, R3 is cyclopropyl or cyclobutyl.
The compound according to formula (I) as described herein can be, for instance, a compound according to formula (la) R1-(CH2)7-CH=CH-C≡C-C≡C-R3
(la) and all stereoisomers, racemic mixtures, tautomers, pharmaceutically acceptable salts, prodrugs, hydrates, solvates and polymorphs thereof, wherein R1 and R3 are as defined herein. In one embodiment, R3 represents an aliphatic group containing 1 to 4 carbon atoms, wherein one divalent carbon atom can be replaced by oxygen or sulfur and wherein the alkyl group R3 can be optionally substituted by one or more Hal. Preferably, R3 is selected from -CH2CH2CH3, -CH2OCH3, -CH2SCH3, -CHFCH2CH3, -CF2CH2CH3. More preferably, R3 is -CH2CH2CH3.
Preferably, the double bond of -(CH2)7-CH=CH- has a fZ) -configuration.
The compound according to formula (la) as described herein, is preferably a compound according to formula (Ic)
HOOC-(CH2)7-CH=CH-C≡C-C≡C-R3
(Ic) and all stereoisomers, racemic mixtures, tautomers, pharmaceutically acceptable salts, prodrugs, hydrates, solvates and polymorphs thereof, wherein R3 is as defined herein. In one embodiment, R3 represents an aliphatic group containing 1 to 4 carbon atoms, wherein one divalent carbon atom can be replaced by oxygen or sulfur and wherein the alkyl group R3 can be optionally substituted by one or more Hal. Preferably, R3 is selected from -CH2CH2CH3, -CH2OCH3, -CH2SCH3, -CHFCH2CH3, -CF2CH2CH3. More preferably, R3 is -CH2CH2CH3.
The compound according to formula (I) as described herein can be, for instance, a compound according to formula (Id)
R1-(CH2)9-C≡C-C≡C-R3
(Id) and all stereoisomers, racemic mixtures, tautomers, pharmaceutically acceptable salts, prodrugs, hydrates, solvates and polymorphs thereof, wherein R1 and R3 are as defined herein. In certain embodiment, R3 represents an aliphatic group containing 1 to 4 carbon atoms, wherein one divalent carbon atom can be replaced by oxygen or sulfur and wherein the alkyl group R3 can be optionally substituted by one or more Hal. Preferably, R3 is selected from -CH2CH2CH3. -CH2OCH3, -CH2SCH3, -CHFCH2CH3, -CF2CH2CH3. More preferably, R3 is -CH2CH2CH3.
The compound according to formula (Id) as described herein, is preferably a compound according to formula (Ie)
HOOC-(CH2)9-C≡C-C≡C-R3
(Ie) and all stereoisomers, racemic mixtures, tautomers, pharmaceutically acceptable salts, prodrugs, hydrates, solvates and polymorphs thereof, wherein R3 is as defined herein. In certain embodiment, R3 represents an aliphatic group containing 1 to 4 carbon atoms, wherein one divalent carbon atom can be replaced by oxygen or sulfur and wherein the alkyl group R3 can be optionally substituted by one or more Hal. Preferably, R3 is selected from -CH2CH2CH3, -CH2OCH3, -CH2SCH3, -CHFCH2CH3, -CF2CH2CH3. More preferably, R3 is -CH2CH2CH3.
Preferred compounds are illustrated in the examples.
Pharmaceutical compositions
The compounds of the present invention can be administered to a patient in the form of a pharmaceutical composition which can optionally comprise one or more pharmaceutically acceptable excipient(s) and/or carrier(s).
The pharmaceutical compositions according to the present invention are useful for treating fungal infections in an individual in need thereof. The pharmaceutical compositions may be in any suitable form depending on the fungal infection to be treated. Thus, the pharmaceutical composition may be formulated for topical administration or for systemic administration. If the fungal infection is a local infection on a body surface, then typically the pharmaceutical composition is formulated for topical administration. If the fungal infection is a disseminated infection and/or an infection of one or more inner organs, tissues or cells, then typically the pharmaceutical composition is formulated for systemic administration, including oral, rectal, intragastrical, and parenteral administration, e.g., intravenous, intramuscular, intranasal, intradermal, subcutaneous, and other administration routes.
Depending on the route of administration the compound of the invention can be provided in various pharmaceutical formulations and some of those may require that protective coatings are applied to the drug formulation to prevent degradation of the compound of the invention in, for example, the digestive tract. Thus, a compound of the invention can be formulated as a syrup, an infusion solution, injection solution, a spray, a tablet, a capsule, a capslet, a lozenge, a liposome, a suppository, a plaster, a band-aid, a retard capsule, a powder, or a slow release formulation.
With respect to certain fungal infections, for example infections of the urinary tract, a compound of the present invention is preferably administered orally. Therefore, particular preferred pharmaceutical forms for the administration of a compound of the invention are forms suitable for oral administration. Formulations for oral administration are usually supplied in dosage units and may contain conventional excipients, such as binders, fillers, diluents, tableting agents, lubricants, detergents, disintegrants, colorants, flavors and wetting agents. Tablets may be coated in accordance to methods well known in the art. Suitable fillers include or are preferably cellulose, mannitol, lactose and similar agents. Suitable disintegrants include or are preferably starch, polyvinyl pyrrolidone and starch derivatives such as sodium starch glycolate. Suitable lubricants include or are preferably, for example, magnesium stearate. Suitable wetting agents include or are preferably sodium lauryl sulfate. These solid oral compositions can be prepared with conventional mixing, filling or tableting methods. The mixing operations can be repeated to disperse the active agent in compositions containing large quantities of fillers. These operations are known to the skilled person.
Pharmaceutical compositions comprising the compound of the invention as liquid compositions for oral administration can be provided in the form of, for example, aqueous solutions, emulsions, syrups or elixirs or in the form of a dry product to be reconstituted with water or with a suitable liquid carrier at the time of use. The liquid compositions can contain conventional additives, such as suspending agents, for example sorbitol, syrup, methylcellulose, gelatin, hydroxyethyl cellulose, carboxymethyl cellulose, aluminum stearate gel or hydrogenated edible fats; emulsifying agents, for example lecithin, sorbitan monooleate, or acacia; non aqueous carriers (which can include edible oil), for example almond oil, fractionated coconut oil, oily esters, such as glycerin esters, propylene glycol or ethyl alcohol; preservatives, for example methyl or propyl p-hydroxybenzoate or sorbic acid; penetration enhancer, for example dimethylsulfoxide (DMSO); pH buffer systems, for example phosphate buffer, carbonate buffer, citrate buffer, citrate-phosphate buffer and other pharmaceutically acceptable buffer systems; solubilizers, for example beta-cyclodextrin, and if desired, conventional flavors or colorants.
Oral formulations may optionally further include taste-masking components to optimize the taste) perception of the oral formulation. Examples of such taste-masking components may be citrus-, licorice-, mint-, grape-, black currant- or eucalyptus-based flavorants known to those well-skilled in the art.
Further preferred forms of administration of compounds of the present invention are forms suitable for injectionable use and include sterile aqueous solutions or dispersions and sterile powders for the extemporaneous preparation of sterile injectable solutions or dispersion. In all cases the final solution or dispersion form must be sterile and fluid. Typically, such a solution or dispersion will include a solvent or dispersion medium, containing, for example, water- buffered aqueous solutions, e.g., biocompatible buffers (e.g., citrate buffer), ethanol, polyol, such as glycerol, propylene glycol, polyethylene glycol, suitable mixtures thereof, surfactants or vegetable oils. A compound of the invention can also be formulated into liposomes, in particular for parenteral administration. Liposomes provide the advantage of increased half-life in the circulation, if compared to the free drug and a prolonged even more release of the enclosed drug.
Sterilization of infusion or injection solutions can be accomplished by any number of art recognized techniques including but not limited to addition of preservatives like anti -bacterial or anti -fungal agents, e.g., parabene, chlorobutanol, phenol, sorbic acid or thimersal. Further, isotonic agents, such as sugars or salts, in particular sodium chloride may be incorporated in infusion or injection solutions.
Production of sterile injectable solutions containing one or several of the compounds of the invention is accomplished by incorporating the respective compound in the required amount in the appropriate solvent with various ingredients enumerated above as required followed by sterilization. To obtain a sterile powder the above solutions are vacuum-dried or freeze-dried as necessary. Preferred diluents of the present invention are water, physiologically acceptable buffers, physiologically acceptable buffer salt solutions or salt solutions. Preferred carriers are cocoa butter and vitebesole.
If a compound of the invention is administered intranasally, it may be administered in the form of a dry powder inhaler or an aerosol spray from a pressurized container, pump, spray or nebulizer with the use of a suitable propellant, e.g., dichlorodifluoromethane, trichlorofluorom ethane, dichlorotetrafluoroethane, a hydrofluoroalkane such as 1, 1,1,2- tetrafluoroethane (HFA 134A™) or 1,1,1,2,3,3,3-heptafluoropropane (HFA 227EA™), carbon dioxide, or another suitable gas. The pressurized container, pump, spray or nebulizer may contain a solution or suspension of the compound of the invention, e.g., using a mixture of ethanol and the propellant as the solvent, which may additionally contain a lubricant, e.g., sorbitan trioleate.
If the fungal infection is a local infection on a body surface, then the pharmaceutical composition can be formulated as a topical formulation. Accordingly, one or more compound of the present invention can be formulated in combination with a solid or a liquid dermatologically acceptable carrier. Useful solid carriers include finely divided solids such as talc, clay, micro crystalline cellulose, silica, alumina, and the like. Useful liquid carriers include water, alcohols, or glycols (or water-alcohol/glycol blends), in which the present compounds can be dissolved or dispersed at effective levels. Adjuvants (such as flavourings and/or fragrances), surfactants, and additional antimicrobial agents can be added to optimize the properties for a given use. The compositions can be applied from absorbent pads, used to impregnate bandages and other dressings, or sprayed onto the affected area using pump-type or aerosol sprayers. The liquid compositions can also be employed as eye drops, mouth washes, douches, etc.
Thickeners such as synthetic polymers, fatty acids, fatty acid salts and esters, fatty alcohols, modified celluloses, or modified mineral materials can also be employed with liquid carriers to form spreadable pastes, gels, ointments, soaps, and the like, for application directly to the skin of the user, which is mainly relevant for treating fungal infections of the skin.
Further excipients which can be used with the various pharmaceutical forms of a compound of the invention can be chosen from the following non-limiting list: a) binders such as lactose, mannitol, crystalline sorbitol, dibasic phosphates, calcium phosphates, sugars, microcrystalline cellulose, carboxymethyl cellulose, hydroxyethyl cellulose, polyvinyl pyrrolidone and the like; b) lubricants such as magnesium stearate, talc, calcium stearate, zinc stearate, stearic acid, hydrogenated vegetable oil, leucine, glycerids and sodium stearyl fumarates, c) disintegrants such as starches, croscaramellose, sodium methyl cellulose, agar, bentonite, alginic acid, carboxymethyl cellulose, polyvinyl pyrrolidone and the like.
Other suitable excipients can be found in the Handbook of Pharmaceutical Excipients, published by the American Pharmaceutical Association, which is herein incorporated by reference.
It is to be understood that depending on the severity of the infection and the particular type which is treatable with one of the compounds of the invention, as well as on the respective patient to be treated, e.g., the general health status of the patient, etc., different doses of the respective compound can be required to elicit a therapeutic or prophylactic effect. The determination of the appropriate dose lies within the discretion of the attending physician. It is contemplated that the dosage of a compound of the invention in the therapeutic or prophylactic use of the invention should be in the range of about 0.1 mg to about 1 g of the active ingredient (i.e. compound of the invention) per kg body weight. However, in a preferred use of the present invention a compound of the invention can be administered to a subject in need thereof in an amount ranging from 1.0 to 500 mg/kg body weight, preferably ranging from 1 to 200 mg/kg body weight. The duration of therapy with a compound of the invention will vary, depending on the severity of the disease being treated and the condition and idiosyncratic response of each individual patient. In one preferred embodiment of a prophylactic or therapeutic use, between 100 mg to 200 mg of the compound is orally administered to an adult per day, depending on the severity of the disease.
The compounds of the present invention are particularly useful for treating, ameliorating, or preventing fungal infections. Preferably, the compounds of the present invention are employed to treat fungal infections. The subject to be treated is not particularly restricted and can be any vertebrate, such as birds and mammals (including humans). The fungal infection that can be treated, ameliorated or prevented with the compounds of the present invention is not particularly limited and can be caused by a fungus selected from Candida spp. (for example C. albicans, C. auris, C. krusei, C. glabrata, C. tropicalis, C. parapsilosis, C. guilliermondii, C. haemulonii, C. lusitaniae, C. lipolytica, C. norvegensis, C. viswanathii, C. kefyr or C. dubliniensis), Aspergillus spp. (for example A. fumigatus, A. flavus, A. niger or A. terreus) Histoplasma capsulatum, Coccidioides immitis, Coccidioides posadasii, Cryptococcus spp. (for example C. neoformans (for example var. neoformans or var. gattii), C. bidus, C. laurentii, or C. fusarium), Zygomycetes (such as Saksenaea vasiformis), Malassezia spp. (for example M. furfur or M. globosa), Hyalohyphomycetes (for example Scedosporium spp., such as S. prolificans or S. apiospermum), Dermatophytes (for example Trichophyton spp. (for example T. mentagrophytes, T. rubrum or T. tonsurans), Epidermophyton floccosum, Microsporum spp (for example M. cookei, M. canis, M. vanbreuseghemii, M. gallinae or M. gypseum or Trichosporon terrestre), Blastomyces dermatitidis, Sporothrix schenkii, Chromomycotic fungi (for example Fonsecaea pedrosoi, F. compacta, Cladophylophora carrionii or Phialophora verrucosa) and Madurella spp. (for example M. mycetomatis or M. griseum), Pneumocystis jirovecii, and Pneumocystis carinii. Preferably, the fungal infection that can be treated, ameliorated or prevented with the compounds of the present invention is caused by a fungus selected from Candida spp. (for example C. albicans, C. auris, C. krusei, C. glabrata, C. tropicalis, C. parapsilosis, C. guilliermondii, C. haemulonii, C. lusitaniae, C. lipolytica, C. norvegensis, C. viswanathii, C. kefyr or C. dubliniensis), and Aspergillus spp. (for example A. fumigatus, A. flavus, A. niger or A. terreus).
The compounds having the formula (I) can be used in combination with one or more other medicaments. The type of the other medicaments is not particularly limited and will depend on the disorder to be treated. Preferably the other medicament will be a further medicament which is useful in treating, ameloriating or preventing a fungal infection, more preferably a further medicament which is useful in treating, ameloriating or preventing a fungal infection that is caused by a fungus selected from Candida spp. (for example C. albicans, C. auris, C. krusei, C. glabrata, C. tropicalis, C. parapsilosis, C. guilliermondii, C. haemulonii, C. lusitaniae, C. lipolytica, C. norvegensis, C. viswanathii, C. kefyr or C. dubliniensis), Aspergillus spp. (for example A. fumigatus, A. flavus, A. niger or A. terreus) Histoplasma capsulatum, Coccidioides immitis, Coccidioides posadasii, Cryptococcus spp. (for example C. neoformans (for example var. neoformans or var. gattii), C. bidus, C. laurentii, or C. fusarium), Zygomycetes (such as Saksenaea vasiformi), Malassezia spp. (for example M. furfur or M. globosa), Hyalohyphomycetes (for example Scedosporium spp., such as S. prolificans or S. apiospermum), Dermatophytes (for example Trichophyton spp. (for example T. mentagrophytes, T. rubrum or T. tonsurans), Epidermophyton floccosum, Microsporum spp (for example M. cookei, M. canis, M. vanbreuseghemii, M. gallinae or M. gypseum or Trichosporon terrestre), Blastomyces dermatitidis, Sporothrix schenkii, Chromomycotic fungi (for example Fonsecaea pedrosoi, F. compacta, Cladophylophora carrionii or Phial ophora verrucosa) and Madurella spp. (for example M. mycetomatis or M. griseum), Pneumocystis jirovecii, and Pneumocystis carinii. Preferably, the fungal infection that can be treated, ameliorated or prevented with the compounds of the present invention is caused by a fungus selected from Candida spp. (for example C. albicans, C. auris, C. krusei, C. glabrata, C. tropicalis, C. parapsilosis, C. guilliermondii, C. haemulonii, C. lusitaniae, C. lipolytica, C. norvegensis, C. viswanathii, C. kefyr or C. dubliniensis), and Aspergillus spp. (for example A. fumigatus, A. flavus, A. niger or A. terreus).
The pharmaceutical compositions of the invention may be for treatment of a disseminated infection or a local infection.
Accordingly, the infection by a fungus may preferably be an infection involving at least partly infection of tissue, organs or cells with hypoxic conditions, preferably the infection may be infection of tissues, organs or cells with hypoxic conditions. Thus, the infection may at least partly involve infection of one or more inner organs, tissues or cells of a mammal, preferably of a human being. More preferably, the infection may be infection of one or more inner organs, tissues or cells of a mammal, preferably of a human being. The hypoxic condition is preferably an oxygen partial pressure (Po2) of at the most 140 mmHg, preferably at the most 110 mmHg, such as at the most 80 mmHg. Such conditions may in general be found in inner organs, for example in the liver, pancreas, gut, duodenum, skeletal muscles, brain, kidney or peritoneal cavity.
The infection by the fungus may also involve, at least partly, infection of a body surface. Infection of a body surface may, for example, be infection of skin, nails or mucosal membranes of body surfaces. Thus, the infection may be infection of a body surface, for example infection of skin, nails or mucosal membranes of body surfaces. As meant herein, body surfaces may include the oral cavity, the genital organs, nose or eyes. Accordingly, the fungal infection may be one or more selected from oropharyngeal fungal infections (such as thrush, glossitis, stomatitis or angular cheilitis), cutaneous fungal infections (such as intertrigo, diaper candidiasis, paronychia or onychomycosis), paronychia, onychomycosis, vulvovaginal fungal infection, balanitis, mucocutaneous fungal infection, neonatal fungal infection, congenital fungal infection, oesophageal fungal infection, gastrointestinal fungal infection, pulmonary fungal infection, peritonitis, urinary tract fungal infections, renal fungal infection, meningitis associated with fungi, hepatic fungal infection, hepatosplenic fungal infection, endocarditis, myocarditis, pericarditis, ocular fungal infection, endophthalmitis and osteo articular fungal infection.
The compounds according to the present invention can be useful for treating onychomycosis, i.e., a fungal infection of the nails.
The infection by a fungus may be an infection by one species of fungus or an infection by more than one fungal species, such as 2, for example 3, such as 4, for example 5, such as more than 5 different fungal species. The fungus may be any fungus, but usually it is a pathogenic fungus, such as a fungus pathogenic in the individual to be treated. In one preferred embodiment of the invention, the individual to be treated is a human being, and then the fungus is a fungus pathogenic in human beings.
The fungus may preferably be selected from Candida spp., Aspergillus spp., Histoplasma capsulatum, Coccidioides immitis, Coccidioides posadasii, Cryptococcus spp., Zygomycetes, Malassezia spp., Hyalohyphomycetes, Dermatophytes, Epi derm ophy ton floccosum, Microsporum spp, Blastomyces dermatitidis, Sporothrix schenkii, Chromomycotic fungi and Madurella spp.
Thus the fungus may be selected from Candida spp., preferably from C. albicans, C. krusei, C. glabrata, C. tropicalis, C. parapsilosis, C. guilliermondii, C. haemulonii, C. lusitaniae, C. lipolytica, C. norvegensis, C. viswanathii, C. kefyr and C. dubliniensis.
The fungus may also be selected from the Aspergillus spp., preferably from A. fumigatus, A. flavus, A. niger and A. terreus. The fungus may also be selected from Cryptococcus spp., preferably from C. neoformans, C. bidus, C. laurentii, and C. fusarium. The C. neoformans is preferably selected from var. neoformans and var. gattii.
The fungus may also be selected from zygomycetes, preferably the fungus is Saksenaea vasiformis.
The fungus may also be selected from Malassezia spp., preferably from M. furfur and globosa.
The fungus may also be selected from Hyalohyphomycetes, preferably from Scedosporium spp., wherein the Scedosporium spp. preferably is selected from S. prolificans and S. apiospermum.
The fungus may also be selected from Dermatophytes. In particular this is the case when the infection is partly or entirely an infection of the skin. The Dermatophyte may preferably be selected from Trichophyton spp., Epidermophyton floccosum, Microsporum spp and Trichosporon terrestre. The Trichophyton spp. may preferably be selected from T. mentagrophytes, T. rubrum and T. tonsurans. The Microsporum spp may preferably be selected from M. cookei, M. canis, M. vanbreuseghemii, M. gallinae and M. gypseum.
The fungus may also be selected from Chromomycotic fungi, preferably from Fonsecaea pedrosoi, F. compacta, Cladophylophora carrionii and Phialophora verrucosa.
The fungus may also be selected from Madurella spp., preferably from mycetomatis and M. griseum.
Combinations of more than one fungus can also be treated.
Without wishing to be bound by theory it is assumed that the compounds of the present invention are capable of inhibiting transcriptional activation of Olel protein. The Olel protein is a A9-fatty acid desaturase, which converts stearic acid to oleic acid. Deletion of the Olel gene induces an oleic acid auxotrophy, which is lethal to the fungal cell. Such activity has been previously demonstrated for the compound EV-086 that belongs to the prior art. However, delivery of a compound into a fungal cell may represent a problem depending on, e.g., the solubility of the compound or its capabilities to cross the fungal cell membrane. The present invention not only shows that the claimed compounds according to formula (I) have in vivo antifungal activity against common Candida albicans fungus, but that the claimed compounds show an improved in vivo antifungal activity against fungi that are resistant to fluconazole, including Candida parapsilosis, when compared to EV-086.
Therefore, another aspect of the present invention is that the compounds according to the formula (I) are capable of treating infections by fungi which are resistant to one or more conventional antifungal agents, in particular antifungal agents, which are not capable of inhibiting conversion of a saturated fatty acid to a A9-monounsaturated fatty acid in a fungus.
The fungi may be resistant for any reason. Thus, for example, particular species of fungus may be resistant to treatment with that particular antifungal agent. It is also possible that the fungus may have acquired resistance, i.e. in general the fungal species is not resistant to treatment with the particular antifungal agent, but this particular fungus has become resistant. In a preferred embodiment of the invention, the fungus has acquired resistance to one or more conventional antifungal agents.
Thus, the compounds according to formula (I) according to the invention and pharmaceutical compositions comprising the same are useful for treating infection by a fungus, which is resistant to one or more other antifungal agents. In particular, the compounds according to formula (I) according to the invention and pharmaceutical compositions comprising the same are useful for treating an infection by a fungus, which is resistant to one or more antifungal agents capable of at least one of: a) inhibiting ergosterol biosynthesis; b) binding to ergosterol; c) inhibiting 1,3-P-glucan synthase; d) inhibiting epoxidase; e) inhibiting leucyl-tRNA synthetase; and/or f) inhibition of elongation factor 2.
In particular, the compounds according to formula (I) according to the invention and pharmaceutical compositions comprising the same are useful for treating infection by a fungus, which is resistant to one or more antifungal agents selected from the group consisting of polyene antifungal agents, azole antifungal agents, allylamine antifungal agents and echinocandins. Non-limiting examples of polyenes include Natamycin, Rimocidin, Filipin, Nystatin, Amphotericin B or Candicin.
Azole antifungal agents may for example be imidazole or triazole or thiazole antifungal agents. Non- limiting examples of imidazole antifungal agents include miconazole, ketoconazole, clotromazole, econazole, bifonazole, butoconazole, fenticonazole, isoconazole, oxiconazole, seraconazole, sulconazole or tioconazole. Non-limiting examples of triazole antifungal agents include fluconazole, itraconazole, isavuconazole, ravuconazole, posaconazole, voriconazole or terconazole. A non-limiting example of a thiazole antifungal is abafungin.
Non-limiting examples of allylamine antifungals include Terbinafme, Amorolfme, Naftifme or Butenafme.
Non-limiting examples of echinocandins include Anidulafungin, Caspofungin or Micafungin.
The pharmaceutical compositions comprising compounds according to formula (I) of the invention may also be useful for treating infection by a fungus, which is resistant to one or more antifungal agents selected from the group consisting of benzoic acid, ciclopirox, tolnaftate, undecylenic acid, flucytosine, griseofulvin, haloprogin and sodium bicarbonate.
By the term "resistant to an antifungal agent", it is meant that the infection by fungus in the individual cannot be treated in a curable manner with the antifungal agent.
Compounds of formula (I) may demonstrate activity against more than one type of organism and are therefore particularly suitable and effective for administration to patients with more than one type of infection (e.g., a patient may have two types of fungal infections). The pharmaceutical compositions for treating fungal infections according to the invention may in addition to one or more compounds according to formula (I) also comprise additional active agents, preferably one or more antifungal agents.
The pharmaceutical compositions may in addition to one or more compounds according to formula (I) also comprise one or more antifungal agents capable of at least one of: a) inhibiting ergosterol biosynthesis; b) binding to ergosterol; c) inhibiting 1,3-P-glucan synthase; d) inhibiting epoxidase; e) inhibiting leucyl-tRNA synthetase; and/or f) inhibition of elongation factor 2.
Thus, the additional antifungal agent may be for example be selected from the group consisting of polyene antifungal agents (such as any of the polyene antifungal agents described herein above in the section), azole antifungal agents (such as any of the azole antifungal agents described herein above in the section), allylamine antifungal agents (such as any of the allylamine antifungal agents described herein above in the section) and echinocandins (such as any of the echinocandins described herein above in the section).
Synthesis of the compounds of the present invention
The compounds of the present invention can be synthesized by one of the general methods shown in the following scheme. These methods are only given for illustrative purposes and should not to be construed as limiting.
Scheme 1
Figure imgf000034_0001
Brominated alkyne precursors bearing the R3 substituent are either sourced commercially or prepared according to the methods known to a skilled person, as exemplified below. Compounds bearing a diyne moiety can be prepared by a Pd-catalyzed and Cu-catalyzed coupling reaction or a Cu-catalyzed coupling reaction, according to one of two general synthetic routes according to Scheme 1, depending on the position of the bromine atom on one of the two possible alkyne components. In a subsequent step, R3 being -COOH or its methyl ester is then derivatized so that compounds according to formula (I) wherein other R3 moieties are present are obtained.
The Cu- and Pd-catalyzed coupling reaction was typically performed as follows: a brominated alkyne derivative (pure or solution in THF or solution in ether) was added to a mixture of alkyne derivative, copper iodide and bis(triphenylphosphine)palladium(II) dichloride in pyrrolidine at 0-30°C and the reaction mixture was stirred at 0-30°C until complete conversion was reached. Work-up and purification of the respective isolated product bearing a diyne moiety were done using appropriate methods.
The Cu-catalyzed coupling reaction was typically performed as follows: a brominated alkyne derivative (pure or solution in THF or solution in ether) was added to a mixture of alkyne derivative, copper chloride, hydroxylamine hydrochloride and ethyl amine (70%-w/w in water) in methanol at 0-30°C and the reaction mixture was stirred at 0-30°C until complete conversion was reached. Work-up and purification of the respective isolated product bearing a diyne moiety were done using appropriate methods.
The invention is illustrated by the following examples which, however, should not be construed as limiting.
EXAMPLES All reagents and solvents were purchased from Sigma-Aldrich, Alfa Aesar, Enamine, or Fluorochem and were used as received. All NMR spectra were recorded on a Bruker AVANCE III HD 500 One Bay spectrometer with a magnetic field of 11.75 T and a 5 mm SmartProbe BB(F)-H-D. For 1H NMR spectra, a frequency of 500 MHz resulted. Chemical shifts are reported in ppm from tetramethylsilane as an internal standard in CDCl3, or from MeOD-d4 and DMSO-d6 as an internal standard (δ = 3.31 and 2.50). Data are reported as follows: chemical shift, multiplicity (s = singlet, d = doublet, t = triplet, q = quartet, quint = quintet, br = broad, m = multiplet), coupling constants (Hz), integration. For 13C NMR spectra, a frequency of 125 MHz resulted. Chemical shifts are reported in ppm from tetramethylsilane as an internal standard in CDCl3, or from MeOD-d4 and DMSO-d6 as an internal standard (d = 49.00 and 39.52). High-resolution mass spectrometry was performed on an Agilent Technologies 6530 Q- TOF. Purity was assayed by HPLC (Agilent Technologies 1200 Series, Interchim Strategy C- 18 column, 4.6 mm x 250 mm, particle size 2.5 μm) with a gradient of 5 to 100 % methanol in water + 0.2 % acetic acid with UV detection at λ = 254 nm. Microwave synthesis was performed in a Biotage initiator. Solid phase peptide synthesis was performed using a CEM Liberty Blue microwave peptide synthesizer. MS analysis was done by UHPLC-MS (Ultimate 3000, Thermo Scientific C-18 column, 2.1 mm x 100 mm, particle size 2.6 μm, Thermo Scientific MSQ Plus) with a gradient of 3 to 97 % acetonitrile in water + 0.2 % formic acid with UV detection at λ = 220, 254, 280, 300 nm.
Preparative Example 1
Compound 1 was prepared according to the following scheme:
Scheme 2
Figure imgf000035_0001
AgNO3 (0.25 mmol, 0.1 equiv.) was added to a solution of 1-pentyne (2.5 mmol, 1.0 equiv.) in acetone (5 mL) and stirred for 5 min. at room temperature (rt.) N-bromosuccinimide (NBS) (2.75 mmol, 1.1 equiv.) was added and the mixture was stirred for 3h in the dark at rt. The mixture was diluted with n-pentane (10 mL) and the solids were filtered off on a pad of Celite, which was washed several times with diethyl ether. The filtrate was concentrated under reduced pressure and passed through a pad of silica gel using diethyl ether as eluent. The filtrate was evaporated under reduced pressure and the residue was resuspended in n-pentane (10 mL), filtrated and concentrated under reduced pressure, 1b was obtained with 44 % yield as a colorless condensed n-pentane solution.
1H-NMR (500 MHz, CDCl3-d): δ 2.21 (t, J= 7.1 Hz, 2H), 1.62-1.51 (m, 2H), 1.00 (t, J= 7.4 Hz, 3H) ppm; Remark: contains n-pentane.
13C-NMR (125 MHz, CDCl3-d): δ 80.3, 37.6, 29.2, 21.8, 21.7 ppm; Remark: contains n- pentane.
MS (ESI-Quad.): was not measured.
Purity: ~80 % (NMR), condensed n-pentane solution.
Step B
CuCl (0.029 mmol, 0.13 equiv.), NH2OH·HCl (0.209 mmol, 0.95 equiv.) and ethylamine (0.814 mmol, 3.7 equiv.) were added in MeOH (1 mL) and cooled to 0°C. Compound 1a, obtained as reported previously (WO 2011/006061 Al) (0.22 mmol, 1.0 equiv.) in MeOH ( 0.5 mL) was added dropwise to the above solution followed by dropwise addition of 1b (0.286 mmol, 1.3 equiv., dissolved in 0.4 mL DCM) over lh. The resulting solution was stirred at 0°C for 3h. The mixture was diluted with 10 mL water and extracted with diethyl ether (3 x 10 ml). The combined organic layers were washed with brine (10 mL), dried over Na2SO4 and concentrated under reduced pressure. The intermediate was purified by flash chromatography (SiO2, gradient cyclohexane-ethyl acetate). LiOH·H2O (0.39 mmol, 3.0 equiv.) was added to a solution of the purified intermediate in THF/MeOH/water (3 mL, 6:1:1) at 0°C. The solution was allowed to warm to rt and stirred for 3h before water (10 mL) was added. The mixture was acidified with 3M HC1 and extracted with diethyl ether (3 x 10 mL). The combined organic layers were washed with brine (10 mL), dried over Na2SO4 and concentrated under reduced pressure. The crude product was purified by flash chromatography (SiO2, gradient cyclohexane-ethyl acetate; reversed phase C 18, gradient 10-100 % methanol in water). 1 was obtained with 20 % yield as a colorless oil. 1H-NMR (500 MHz, CDCl3-d):δ 6.04 (dt, J = 10.8, 7.5 Hz, 1H), 5.49 (dt, J = 10.9, 1.6 Hz, 1H), 2.42-2.30(m, 6H), 1.69-1.58 (m, 4H), 1.43-1.32 (m, 8H), 1.02 (t, J= 7.4 Hz, 3H) ppm.
13C-NMR (125 MHz, CDCl3-d): δ 179.7, 147.6, 108.2, 84.8, 78.2, 72.1, 65.3, 34.0, 30.6, 29.0, 29.0, 28.9, 28.7, 24.7, 21.8, 21.6, 13.5 ppm.
MS (ESI-Quad.): calc. = 260.37 g/mol, found [M+H]+ = 261.2 m/z, [2M+H]+ = 521.3 m/z, [M- H]- = 259.1 m/z, [2M-H]- = 519.2 m/z, [M-H+formate adduct(FA)]- = 305.0 m/z.
Purity: 93.8 % (HPLC, UV254nm).
Preparative example 2
Compound 2 was prepared according to the following scheme starting from the known compound EV-086:
Figure imgf000037_0001
4-Dimethylaminopyridine (DMAP) (0.96 mmol, 2.4 equiv.) was added to a suspension of 1- ethyl-3-(3-dimethylaminopropyl)carbodiimide (ED AC) (0.52 mmol, 1.3 equiv.) in DCM (2 mL). The mixture was stirred at rt until all solids were dissolved. EV-086 (0.4 mmol, 1.0 equiv.) and methanesulfonamide (0.4 mmol, 1.0 equiv.) was added to the reaction mixture at 0°C. The mixture was stirred at rt for 24h. Additional 1 equiv. EDAC and 2 equiv. DMAP was added and the mixture was stirred again for 24h at rt. The reaction mixture was diluted with water (10 mL) and acidified to pH 1 with 3M HC1 and the aqueous layer was extracted with DCM (2 x 10 mL). The combined organic layers were washed with saturated NH4Cl solution (10 mL), brine (10 mL), dried over Na2SO4 and concentrated under reduced pressure. The crude product was purified by flash chromatography (SiO2, gradient cyclohexane-ethyl acetate; reversed phase C 18, gradient 10-100 % methanol in water). 2 was obtained with 52 % yield as a white solid.
1H-NMR (500 MHz, CDCl3-d): δ 8.29 (br. S, 1H), 7.44 (d, J= 1.8 Hz, 1H), 6.74 (d, J= 3.4 Hz, 1H), 6.43 (dd, J = 1.9, 3.5 Hz, 1H), 6.16 (dt, J= 10.8, 7.6 Hz, 1H), 5.61 (dt, J= 10.8, 1.4 Hz, 1H), 3.32 (s, 3H), 2.42-2.31 (m, 4H), 1.74-1.63 (m, 2H), 1.49-1.42 (m, 2H), 1.41-1.33 (m, 6H) ppm.
13C-NMR (125 MHz, CDCl3-d): δ 171.9, 149.2, 144.6, 136.5, 117.9, 111.2, 107.9, 81.8, 78.7, 71.1, 41.6, 36.5, 30.7, 28.9, 28.8, 28.7, 28.5, 24.4 ppm; Remark: 1 C quart, signal is not visible.
MS (ESI-Quad.): calc. = 361.46 g/mol, found [M+H]+ = 362.2 m/z, [M+NH4]+ = 379.2 m/z, [M-H]- = 360.1 m/z.
Purity: 100 % (HPLC, UV254nm).; ~91% (NMR; ~9% E product).
Preparative example 3 (reference example)
Compound 3 was prepared according to the following scheme:
Figure imgf000038_0001
EV-086 (0.2 mmol, 1.0 equiv.) was dissolved in MeOH (1 mL) under an argon atmosphere and cooled to 0°C. BF3·Et2O (0.2 mmol, 1.0 equiv.) was added and the mixture was stirred for 3h at rt. The reaction mixture was diluted with water (15 mL) and the aqueous layer was extracted with diethyl ether (3 x 10 mL). The combined organic layers were dried over Na2SO4 and concentrated under reduced pressure. The crude product was purified by flash chromatography (SiO2, gradient cyclohexane-ethyl acetate). 3 was obtained with 87 % yield as a yellow oil. 1H-NMR (500 MHz, CDCl3-d): δ 7.46-7.39 (m, 1H), 6.77-6.68 (m, 1H), 6.41 (dd, J= 3.5, 1.8 Hz, 1H), 6.14 (dt, J= 10.9, 7.6 Hz, 1H), 5.59 (dt, J= 10.9, 1.4 Hz, 1H), 3.67 (s, 3H), 2.40- 2.29 (m, 4H), 1.69-1.58 (m, 2H), 1.47-1.40 (m, 2H), 1.37-1.30 (m, 6H) ppm.
13C-NMR (125 MHz, CDCl3-d): δ 174.3, 149.2, 144.5, 136.6, 117.77, 111.15, 107.8, 81.6,
78.7, 77.0, 71.0, 51.4, 34.1, 30.9, 29.0, 29.0, 28.9, 28.6, 24.9 ppm.
MS (ESI-Quad.): calc. = 298.38 g/mol, found [M+H]+ = 299.0 m/z, [M-H]- = did not ionize.
Purity: 89.3 % (HPLC, UV254nm).; ~90% (NMR; -10% E product).
Preparative example 4
Compound 4 was prepared according to the following reaction scheme:
Figure imgf000039_0001
ED AC (0.18 mmol, 1.2 equiv.) was added to a solution of EV-086 (0.15 mmol, 1.0 equiv.), hydroxylamine»HCl (0.15 mmol, 1.0 equiv.), HOBt monohydrate (0.18 mmol, 1.2 equiv.) and N-methylmorpholine (0.33 mmol, 2.2 equiv.) in DCM (2 mL) and the mixture was stirred for 3h at rt. The reaction mixture was diluted with water (10 mL) and extracted with DCM (3 x 10 mL). The combined organic layers were dried over Na2SO4 and concentrated under reduced pressure. The crude product was purified by flash chromatography (SiO2, gradient cyclohexane- ethyl acetate). 4 was obtained with 18 % yield as an off-white solid.
1H-NMR (500 MHZ, CDCl3-d): δ 8.31 (br. s, 1H), 7.43 (dd, J= 1.9, 0.7 Hz, 1H), 6.74 (dd, J = 3.5, 0.8 Hz, 1H), 6.43 (dd, J= 3.4, 1.9 Hz, 1H), 6.15 (dt, J= 10.9, 7.6 Hz, 1H), 5.60 (dt, J = 11.0, 1.5 Hz, 1H), 2.43-2.32 (m, 2H), 2.22-2.13 (m, 2H), 1.72-1.62 (m, 2H), 1.48-1.39 (m, 2H), 1.37-1.30 (m, 6H) ppm. 13C-NMR (125 MHz, CDCl3-d): δ 171.4, 149.2, 144.5, 136.6, 117.8, 111.2, 107.9, 81.7, 78.7, 71.1, 69.9, 32.9, 30.7, 29.0, 28.9, 28.8, 28.5, 25.6 ppm.
MS (ESI-Quad.): calc. = 299.36 g/mol, found [M+H]+ = 300.2 m/z, [M+Na]+ = 322.2 m/z, [M+Na+ACN]+ = 363.2 m/z , [M-H]- = 298.0 m/z, [M-H+Cl]- = 334.0 m/z.
Purity: 96.8 % (HPLC, UV254nm).; -70% (NMR; -30% E product).
Preparative example 5
Compound 5 was prepared according to the following reaction scheme:
Figure imgf000040_0001
ED AC (0.165 mmol, 1.1 equiv.) was added to a solution of EV-086 (0.15 mmol, 1.0 equiv.), methoxyamine*HCl (0.11 mmol, 1.1 equiv.), 1-hydroxybenzotriazole (HOBt) monohydrate (0.165 mmol, 1.1 equiv.) and triethylamine (TEA) (0.33 mmol, 2.2 equiv.) in DCM (2 mL) and the mixture was stirred for 3h at rt. The reaction mixture was diluted with water (10 mL) and extracted with DCM (3 x 10 mL). The combined organic layers were dried over Na2SO4 and concentrated under reduced pressure. The crude product was purified by flash chromatography (SiO2, gradient cyclohexane-ethyl acetate). 5 was obtained with 75 % yield as a brown solid.
1H-NMR (500 MHz, CDCl3-d): δ 8.70 (br. s, 1H), 7.41 (dd, J = 1.8, 0.7 Hz, 1H), 6.72 (dd, J = 3.5, 0.6 Hz, 1H), 6.41 (dd, J=3.5, 1.9 Hz, 1H), 6.14 (dt, J= 10.8, 7.6 Hz, 1H), 5.59 (dt, J=10.8, 1.4 Hz, 1H), 3.76 (s, 3H), 2.43-2.30 (m, 2H), 2.20-2.04 (m, 2H), 1.72-1.62 (m, 2H), 1.47-1.39 (m, 2H), 1.38-1.30 (m, 6H) ppm.
13C-NMR (125 MHz, CDCl3-d): δ 171.1, 149.9, 144.5, 136.5, 117.8, 111.2, 107.8, 81.7, 78.7, 77.0, 71.1, 64.4, 33.2, 30.8, 29.1, 28.9, 28.8, 28.6, 25.3 ppm. MS (ESI-Quad.): calc. = 313.39 g/mol, found [M+H]+ = 314.2 m/z, [M+Na]+ = 336.2 m/z, [M+Na+ACN]+ = 377.2 m/z , [M-H]- = did not ionize, [M-H+FA]- = 348.0 m/z.
Purity: 99.5 % (HPLC, UV254nm).; -80% (NMR; -20% E product).
Preparative example 6
Compound 6 was prepared according to the following reaction scheme:
Figure imgf000041_0001
Step A
To a suspension of LiAlH4 (1M in THF, 1.48 mmol, 0.74 equiv.) in dry diethyl ether (8 mL) was added dropwise a solution of compound 3 (2.0 mmol, 1.0 equiv.) at 0°C under argon atmosphere. The mixture was stirred for 5 min at 0°C. The reaction mixture was quenched with care by adding water (0.1 mL), 0.1M NaOH (0.1 mL) and additional water (0.3 mL). After stirring for 1 h, the resulting suspension was filtered through a pad of Celite and the filtrate was concentrated under reduced pressure. The crude product was purified by flash chromatography (SiO2, gradient cyclohexane-ethyl acetate). 7a was obtained with 60 % yield as yellow oil.
1H-NMR (500 MHz, CDCl3-d): δ 7.45-7.39 (m, 1H), 6.76-6.71 (m, 1H), 6.46-6.39 (m, 1H), 6.16 (dt, J= 10.9, 7.6 Hz, 1H), 5.60 (dt, J= 10.9, 1.3 Hz, 1H), 3.66 (t, J= 6.6 Hz, 2H), 2.43- 2.33 (m, 2H), 1.66-1.54 (m, 2H), 1.49-1.42 (m, 2H), 1.41-1.32 (m, 8H) ppm. 13C-NMR (125 MHz, CDCl3-d): δ 149.3, 144.5, 136.6, 117.8, 111.2, 107.8, 100.0, 81.7, 78.7, 71.0, 63.1, 32.8, 30.9, 29.3 (2C), 29.0, 28.7, 25.7 ppm.
MS (ESI-Quad.): calc. = 270.37 g/mol, found [M+H]+ = 271.1 m/z, [M-H]- = did not ionize. Purity: 98 % (NMR).
Step B
TEA (0.182 mmol, 1.3 equiv.) was added slowly under stirring to a solution of 2- chloro[l,3,2]dioxaphospholane (0.14 mmol, 1.0 equiv.) and 7a (0.14 mmol, 1.0 equiv.) in anhydrous toluene (1 mL) at 0°C under argon atmosphere. The mixture was stirred for 4 h at rt. Then additional chloro[l,3,2]dioxaphospholane (1.96 mmol, 1.4 equiv.) and TEA (1.96, 1.4 equiv.) was added and the mixture was stirred for 2 h at rt. The solution was then filtered through a pad of Celite and the filtrate was concentrated under reduced pressure. The crude product was purified by flash chromatography (SiO2, gradient cyclohexane-ethyl acetate). 7 was obtained with 65 % yield as a brown oil.
1H-NMR (500 MHz, CDCl3-d): δ 7.44-7.41 (m, 1H), 6.75-6.71 (m, 1H), 6.42 (ddd, J= 3.5, 1.9, 0.5 Hz, 1H), 6.15 (dt, J= 10.9, 7.6 Hz, 1H), 5.59 (dt, J= 10.8, 1.5 Hz, 1H), 4.49-4.41 (m, 2H), 4.41-4.33 (m, 2H), 4.20-4.12 (m, 2H), 2.41-2.31 (m, 2H), 1.76-1.66 (m, 2H), 1.44-1.30 (m, 10H) ppm.
13C-NMR (125 MHz, CDCl3-d): δ 149.3, 144.5, 136.6, 117.8, 111.2, 107.8, 81.7, 78.7, 77.0, 71.0, 69.2 (d, JCP = 6.3 Hz), 65.9 (d, JCP = 2.7 Hz, 2C), 30.9, 30.3 (d, JCP = 5.9 Hz), 29.2, 29.0, 28.9, 28.6, 25.3 ppm.
MS (ESI-Quad.): calc. = 376.38 g/mol, found [M+H]+ = 377.2 m/z, [M+NH4]+ = 394.1 m/z, [M-H]- = did not ionize, [M-H]2- = 137.0 m/z.
Purity: 85 % (NMR; ~11% E product; HPLC: product not stable).
Preparative example 7
Compound 7 was prepared according to the following reaction scheme:
Figure imgf000043_0001
Step A
Nal (80.0 mmol, 8.0 equiv.) was added to a solution of 10-bromodecanoic acid (10.0 mmol, 1.0 equiv.) in acetone (50 mL) at rt under argon atmosphere. The mixture was stirred for 18h at rt. The mixture was filtered through a pad of Celite, concentrated to one-fifth of volume and diluted with brine (50 mL). The aqueous layer was extracted with n-hexane (4x 15 mL). The combined organic layers were washed with freshly prepared 10% Na2S2O3 solution (2 x 15 mL), dried over Na2SO4 and concentrated under reduced pressure. 7a was obtained with 96 % yield as a white solid.
1H-NMR (500 MHz, CDCl3-d): δ 3.20 (t, J= 7.0 Hz, 2H), 2.37 (t, J= 7.5 Hz, 2H), 1.87-1.80 (m, 2H), 1.69-1.60 (m, 2H), 1.43-1.28 (m, 10H) ppm.
13C-NMR (125 MHz, CDCl3-d): δ 180.1, 34.1, 33.5, 30.5, 29.2, 29.1, 29.0, 28.5, 24.6, 7.3 ppm. MS (ESI-Quad.): was not measured.
Purity: 95 % (NMR)
Step B
A suspension of lithium acetylide ethylenediamine complex (57.0 mmol, 3.0 equiv.) in hexamethylphosphoramide (HMPA) (35 mL) was stirred at rt for 45 min under argon atmosphere before it was cooled to -5°C. A solution of 7a (19.0 mmol, 1.0 equiv.) in HMPA (15 mL) was then added to the cooled suspension at such a rate so to maintain the temperature between 0°C and 5°C. The reaction mixture was allowed to stir for an additional 30 min at - 3°C and then was carefully quenched in portions into ice (250 mL). The mixture was acidified to pH 2.5 with 5M H2SO4 and extracted with diethyl ether (3 x 50 mL). The combined organic layers were washed with water (4 x 20 mL), dried over Na2SO4 and concentrated under reduced pressure. 7b was obtained with 67 % yield as a yellow solid.
1H-NMR (500 MHz, CDCl3-d): δ 11.6, (br. s, 1H), 2.36 (t, J= 7.5 Hz, 2H), 2.19 (td, J= 7.1, 2.7 Hz, 2H), 1.95 (t, J= 2.7 Hz, 1H), 1.68-1.62 (m, 2H), 1.57-1.50 (m, 2H), 1.42-1.30 (m, 10H) ppm.
13C-NMR (125 MHz, CDCl3-d): δ 180.4, 84.7, 68.1, 34.1, 29.3, 29.2, 29.0 (2C), 28.7, 28.5, 24.6, 18.4 ppm.
MS (ESI-Quad.): was not measured.
Purity: 95 % (NMR).
Step C
7b (0.2 mmol, 1.0 equiv.), PdCl2(PPh3)2 (0.006 mmol, 0.03 equiv.), Cul (0.006 mmol, 0.03 equiv.) and a small amount of NH2OH·HCl were dissolved in THF (5 mL) at rt under argon atmosphere. Diisopropylamine (DIP A) (0.6 mmol, 3.0 equiv.) was added followed by dropwise addition of 1b (dissolved in 3 mL THF, 0.24 mmol, 1.2 equiv.) over lh. The solution was stirred at rt for an additional lh. The mixture was diluted with 10 mL water, acidified with 2M HC1 and extracted with diethyl ether (3 x 10 mL). The combined organic layers were washed with brine (10 mL), dried over Na2SO4 and concentrated under reduced pressure. The crude product was purified by flash chromatography (SiO2, gradient cyclohexane-ethyl acetate; reversed phase C 18, gradient 10-100 % methanol in water). 7 was obtained with 19 % yield as a brown solid.
1H-NMR (500 MHz, CDCl3-d): δ 2.36 (t, J = 7.5 Hz, 2H), 2.29-2.18 (m, 4H), 1.69-1.61 (m, 2H), 1.60-1.48 (m, 4H), 1.42-1.28 (m, 10H), 1.01 (t, J= 7.4 Hz, 3H) ppm.
13C-NMR (125 MHz, CDCl3-d): δ 179.5, 77.5, 77.4, 65.4, 65.3, 34.1, 29.3, 29.2, 29.0, 29.0, 28.8, 28.3, 24.7, 21.9, 21.2, 19.2, 13.5 ppm. MS (ESI-Quad.): calc. = 262.39 g/mol, found [M+H]+ = did not ionize, [M-H]- = 261.1 m/z, [M-H+FA]- 307.1 m/z.
Purity: 98 % (NMR).
Preparative example 8
Compound 8 was prepared according to the following synthetic scheme:
Figure imgf000045_0001
Step A
A catalytic amount of Triton B (40% in methanol, 50 μL) was placed in a dried flask and the solvent was removed under reduced pressure. 7-Octyne-l-ol (2.75 mmol, 1.1 equiv.) was added, followed by methyl acrylate (2.5 mmol, 1.0 equiv.) and the mixture was stirred at 50°C for 3 h. The mixture was filtered over a Celite / silica gel mixture with diethyl ether. The filtrate was concentrated under reduced pressure and the crude intermediate was dissolved in THF/water (10 mL, 1:1). LiOH·H2O (20.0 mmol, 8.0 equiv.) was then added at rt and the mixture was stirred for 12 h. THF was evaporated, the aqueous layer was washed with diethyl ether (3 x 10 mL). The aqueous layer was then acidified with 3M HC1 to pH 1 and extracted with diethyl ether (3 x 20 mL). The combined organic layers were washed with brine (30 mL), dried over Na2SO4 and concentrated under reduced pressure. The crude product was purified by flash chromatography (SiO2, gradient cyclohexane-ethyl acetate). 8a was obtained with 14 % yield as a colorless oil.
1H-NMR (500 MHz, CDCl3-d): δ 11.31 (br. s, 1H), 3.70 (t, J= 6.4 Hz, 2H), 3.46 (t, J= 6.6 Hz, 2H), 2.63 (t, J= 6.4 Hz, 2H), 2.18 (td, J= 7.1, 2.7 Hz, 2H), 1.94 (t, J= 2.7 Hz, 1H), 1.62-1.48 (m, 4H), 1.45-1.29 (m, 4H) ppm. 13C-NMR (125 MHz, CDCl3-d): δ 177.6, 84.6, 71.1, 68.2, 65.7, 34.9, 29.3, 28.5, 28.4, 25.5,
18.3 ppm.
MS (ESI-Quad.): calc. = 198.26 g/mol, found [M+H]+ = did not ionize, [M-H]- = 197.2 m/z, [M-H+FA]- = 243.0 m/z.
Purity: 90% (NMR).
Step B
8a (0.15 mmol, 1.0 equiv.) and 1b (0.3 mmol, 2.0 equiv.) were dissolved in pyrrolidine (1 mL) and cooled to 0°C under argon atmosphere. Cul (0.015 mmol, 0.1 equiv.), PdCl2(PPh3)2 (0.008 mmol, 0.05 equiv.) and a small amount of NH2OH·HCl were added to the above solution. The solution was stirred at rt overnight. The mixture was diluted with 10 mL water, acidified with 2M HC1 and extracted with diethyl ether (3 x 10 ml). The combined organic layers were washed with brine (10 mL), dried over Na2SO4 and concentrated under reduced pressure. The crude product was purified by flash chromatography (SiO2, gradient cyclohexane-ethyl acetate; reversed phase C 18, gradient 10 to 100 % methanol in water). 8 was obtained with 28 % yield as a white solid.
1H-NMR (500 MHz, CDCl3-d): δ 3.72 (t, J= 6.3 Hz, 2H), 3.47 (t, J= 6.6 Hz, 2H), 2.65 (t, J =
6.3 Hz, 2H), 2.31-2.17 (m, 4H), 1.65-1.47 (m, 6H), 1.46-1.29 (m, 4H), 1.00 (t, J= 7.4 Hz, 3H) ppm.
13C-NMR (125 MHz, CDCl3-d): δ 176.9, 77.4, 77.4, 71.2, 65.7, 65.4, 65.4, 34.9, 29.3, 28.6, 28.3, 25.6, 21.9, 21.2, 19.1, 13.5 ppm.
MS (ESI-Quad.): calc. = 264.36 g/mol, found [M+H]+ = 265.1 m/z, [M+Na]+ = 287.2 m/z, [M- H]- = 263.1 m/z, [M-H+FA]- = 309.2 m/z.
Purity: 98 % (NMR).
Preparatory example 9
Compound 9 was prepared according to the following reaction scheme:
Figure imgf000047_0001
Step A
Concentrated sulfuric acid (100 uL) was added to a solution of 7b (7.0 mmol, 1.0 equiv.) in MeOH (40 mL) and the mixture was stirred under reflux for 16h. The mixture was concentrated to a third of its volume, diluted with n-hexane/n-pentane (50 mL, 2:1) and washed with saturated NaHCO3 solution (2 x 10 mL). The aqueous layer was then extracted with diethyl ether (2 x 20 mL). The combined organic layers were washed with brine (300 mL), dried over Na2SO4 and concentrated under reduced pressure. The biphasic residue was partitioned between n-hexane (50 mL) and brine (10 mL) and the layers were separated. The aqueous layer was extracted with n-hexane (2 x 10 mL) and the combined organic layers were dried over Na2SO4 and concentrated under reduced pressure. 9a was obtained with 93 % yield as a brown oil.
1H-NMR (500 MHz, CDCl3-d): δ 3.68 (s, 3H), 2.31 (t, J= 7.5 Hz, 2H), 2.19 (td, J= 7.1, 2.7 Hz, 2H), 1.95 (t, J = 2.7 Hz, 1H), 1.67-1.59 (m, 2H), 1.57-1.48 (m, 2H), 1.44-1.36 (m, 2H), 1.34-1.27 (m, 8H) ppm.
13C-NMR (125 MHz, CDCl3-d): δ 174.3, 84.7, 68.1, 51.4, 34.1, 29.3, 29.2, 29.1, 29.0, 28.7, 28.5, 24.9, 18.4 ppm.
MS (ESI-Quad.): was not measured.
Purity: 90 % (NMR). Step B
NBS (7.15 mmol, 1.1 equiv.) was added to a solution of 9a (6.5 mmol, 1.0 equiv.) in acetone (30 mL) under argon atmosphere. AgNO3 (0.65 mmol, 0.1 equiv.) was added and the mixture was stirred for 21h at rt in the dark. The mixture was filtrated and the filtrate was concentrated to a third of its volume and diluted with water (50 mL). The mixture was extracted with n- hexane (2 x 20 mL) and the combined organic layers were washed with brine (20 mL), dried over Na2SO4 and concentrated under reduced pressure. 9b was obtained with 92 % yield as a yellow liquid.
1H-NMR (500 MHz, CDCl3-d): δ 3.68 (s, 3H), 2.31 (t, J= 7.5 Hz, 2H), 2.21 (t, J= 7.1 Hz, 2H), 1.67-1.59 (m, 2H), 1.56-1.46 (m, 2H), 1.40-1.27 (m, 10H) ppm.
13C-NMR (125 MHz, CDCl3-d): δ 174.3, 80.4, 51.4, 37.5, 34.1, 29.3, 29.2, 29.1, 29.0, 28.7, 28.3, 24.9, 19.7 ppm.
MS (ESI-Quad.): was not measured.
Purity: 90 % (NMR).
Step C
9b (0.35 mmol, 1.0 equiv.) and pent- 1-yn-3-ol (0.525 mmol, 1.5 equiv.) were dissolved in pyrrolidine (1 mL) and cooled to 0°C under argon atmosphere. Cul (0.035 mmol, 0.1 equiv.), PdCl2(PPh3)2 (0.018 mmol, 0.05 equiv.) and a small amount of NH2OH·HCl was added to the above solution. The solution was stirred at 0°C for lh. The mixture was diluted with 20 mL water, acidified with 2M HC1 and extracted with diethyl ether (3 x 10 ml). The combined organic layers were washed with brine (10 mL), dried over Na2SO4 and concentrated under reduced pressure. The crude product was purified by flash chromatography (SiO2, gradient cyclohexane-ethyl acetate). 9c was obtained with 60 % yield as a yellow oil.
1H-NMR (500 MHz, CDCl3-d): δ 4.35 (t, J= 6.3 Hz, 1H), 3.67 (s, 3H), 2.34-2.23 (m, 5H), 1.78- 1.69 (m, 2H), 1.66-1.57 (m, 2H), 1.56-1.48 (m, 2H), 1.41-1.34 (m, 2H), 1.34-1.25 (m, 8H), 1.01 (t, J= 7.4 Hz, 3H) ppm. 13C-NMR (125 MHz, CDCl3-d): δ 174.5, 81.5, 76.4, 69.9, 64.5, 64.0, 51.5, 34.1, 30.8, 29.2, 29.1, 29.1, 28.9, 28.7, 28.1, 24.9, 19.2, 9.4 ppm.
MS (ESI-Quad.): calc. = 292.41 g/mol, found [M+Na]+ = 315.2 m/z, [M+Na+ACN]+ = 356.2 m/z [M-H]-= did not ionize.
Purity: 98 % (NMR).
Step D
XtalFluor-E (0.127 mmol, 2.02 equiv.) was added to a stirred solution of TEA· 3 HF (0.139 mmol, 2.2 equiv.) and TEA (0.064 mmol, 1.02 equiv.) in DCM (lmL) at -72°C under argon atmosphere. After stirring for 10 min, a solution of 9c (0.063 mmol, 1.0 equiv.) in DCM (0.3 mL) was added dropwise to the above solution. The reaction mixture was stirred for lh at -72°C and was then allowed to warm to rt and stirred for another 3h. The reaction was quenched with 5 % NaHCO3 solution (10 mL) and the aqueous layer was extracted with diethyl ether (3 x 10 mL). The combined organic layers were dried over Na2SO4 and concentrated under reduced pressure. The crude product was purified by flash chromatography (SiO2, gradient cyclohexane-ethyl acetate). 9d was obtained with 74 % yield as a colorless oil.
1H-NMR (500 MHz, CDCl3-d): δ 5.10 (dtt, J= 48.3, 6.2, 0.9 Hz, 1H), 3.69 (s, 3H), 2.37-2.28 (m, 4H), 1.95-1.84 (m, 2H), 1.69-1.60 (m, 2H), 1.58-1.51 (m, 2H), 1.43-1.36 (m, 2H), 1.34- 1.28 (m, 8H), 1.06 (t, J= 7.4 Hz, 3H) ppm.
13C-NMR (125 MHz, CDCl3-d): δ 174.3, 84.0 (d, JCF = 169.1 Hz), 83.4 (d, JCF = 4.5 Hz), 73.1 (d , JCF= 10.7 Hz), 71.5 (d, JCF = 25.5 Hz), 64.1 (d , JCF= 6.4 Hz), 51.4, 34.1, 29.2, 29.2 (d, JCF = 23.1 Hz), 29.2, 29.1, 29.0, 28.8, 28.0, 24.9, 19.3, 8.7 (d, JCF = 5.5 Hz) ppm.
MS (ESI-Quad.): was not measured.
Purity: 98 % (NMR).
Step E LiOH·H2O (0.14 mmol, 3.0 equiv.) was added to a solution of 9d (0.047 mmol, 1.0 equiv.) in THF/MeOH/water (3 mL, 6: 1 : 1) at 0°C. The solution was allowed to warm to rt and stirred for 3.5h before water (10 mL) was added. The pH was adjusted to pH 2 with 2M HC1. 10 mL diethyl ether was added and the layers were separated. The aqueous layer was extracted with diethyl ether (2 x 10 mL) and the combined organic layers were washed with brine (10 mL), dried over Na2SO4 and concentrated under reduced pressure. The crude product was purified by flash chromatography (SiO2, gradient cyclohexane-ethyl acetate). 9 was obtained with 84 % yield as a white solid.
1H-NMR (500 MHz, CDCl3-d): δ 5.10 (dt, J= 48.2, 6.1 Hz, 1H), 2.37 (t, J= 7.5 Hz, 2H), 2.34- 2.27 (m, 2H), 1.97-1.82 (m, 2H), 1.73-1.60 (m, 2H), 1.60-1.51 (m, 2H), 1.42-1.28 (m, 10H), 1.06 (t, J= 7.4 Hz, 3H) ppm.
13C-NMR (125 MHz, CDCl3-d): δ 179.4, 84.0 (d, JCF = 169.2 Hz), 83.4 (d, JCF = 4.7 Hz), 73.1 (d, JCF = 10.8 Hz), 71.5 (d, JCF = 25.4 Hz), 64.1 (d, JCF = 6.8 Hz), 33.9, 29.2, 29.2 (d, JCF = 23.2 Hz), 29.2, 29.0, 29.0, 28.8, 28.0, 24.7, 19.3, 8.7 (d, JCF = 5.4 Hz) ppm.
MS (ESI-Quad.): calc. = 280.38 g/mol, found [M+H]+ = did not ionize, [M-H]- = 279.1 m/z, [M-H+FA]- = 325.1 m/z.
Purity: 98 % (NMR).
Preparative example 10
Compound 10 was obtained according to the following synthetic scheme:
Figure imgf000050_0001
Step A
Pyridinium chlorochromate (PCC) (0.098 mmol, 1.25 equiv.) was added in DCM (1 mL) and the mixture was cooled to 0°C. 9c (0.078 mmol, 1.0 equiv.) dissolved in DCM (0.3 ml) was added dropwise over 10 min and the mixture was stirred overnight at rt. Diethyl ether (10 mL) was added and the mixture was filtered through a pad of Celite. The filtrate was concentrated under reduced pressure. The crude product was purified by flash chromatography (SiO2, gradient cyclohexane-ethyl acetate). 10a was obtained with 48 % yield as a brown oil.
1H-NMR (500 MHz, CDCl3-d): δ 3.69 (s, 3H), 2.61 (q, J= 7.4 Hz, 2H), 2.38 (t, J = 7.1 Hz, 2H), 2.33 (t, J= 7.5 Hz, 2H), 1.67-1.56 (m, 4H), 1.44-1.37 (m, 2H), 1.34-1.29 (m, 8H), 1.17 (t, J= 7.4 Hz, 3H) ppm.
13C-NMR (125 MHz, CDCl3-d): δ 187.7, 174.3, 90.6, 76.2, 72.1, 63.8, 51.5, 38.8, 34.1, 29.2, 29.2, 29.1, 28.9, 28.8, 27.7, 24.9, 19.6, 8.0 ppm.
MS (ESI-Quad.): calc. = 290.40 g/mol, found [M+H]+ = 291.1 m/z, [M+Na]+ = 313.2 m/z, [M+Na+ACN]+ = 354.2 m/z, [M-H]-= did not ionize.
Purity: 98 % (NMR).
Step B
10a (0.2 mmol, 1.0 equiv.) was added to a mixture of DAST (1.0 mmol, 5.0 equiv.) and ethanol (5 uL) under argon atmosphere. The reaction mixture was stirred at 50°C for 24h before it was quenched with saturated NH4CI solution (10 mL) and extracted with EtOAc (2 x 10 mL). The combined organic layers were washed with brine (10 mL), dried over Na2SO4 and concentrated under reduced pressure. The intermediate was purified by flash chromatography (SiO2, gradient cyclohexane-ethyl acetate). LiOH·H2O (0.6 mmol, 3.0 equiv.) was added to a solution of the purified intermediate in THF/MeOH/water (3 mL, 6:1:1) at 0°C. The solution was allowed to warm to rt and stirred for 3.5h. The mixture was diluted with 10 mL water, acidified with 3M HC1 and extracted with diethyl ether (3 x 10 mL). The combined organic layers were washed with brine (10 mL), dried over Na2SO4 and concentrated under reduced pressure. The crude product was purified by flash chromatography (SiO2, gradient cyclohexane-ethyl acetate). 10 was obtained with 45 % yield as a colorless oil.
1H-NMR (500 MHz, CDCL): δ 2.40-2.31 (m, 4H), 2.08 (tq, J= 14.8, 7.5 Hz, 2H), 1.70-1.61 (m, 2H), 1.60- 1.52 (m, 2H), 1.42-1.30 (m, 10H), 1.11 (t, J= 7.5 Hz, 3H) ppm; Remark: contains ethyl acetate. 13C-NMR (125 MHz, CDCL): δ 179.9, 115.3 (t, JCF = 233.2 Hz), 85.3, 71.9 (t, JCF = 7.2 Hz), 66.9 (t, JCF = 41.6 Hz), 63.3 (t, JCF = 4.8 Hz), 34.0, 32.9 (t, JCF = 27.1 Hz), 29.2, 29.1, 29.0, 28.9, 28.8, 27.8, 24.6, 19.2, 7.0 (t, JCF = 4.2 Hz) ppm; Remark: contains ethyl acetate.
MS (ESI-Quad.): calc. = 298.37 g/mol, found [M+Na+ACN]+ = 362.1 m/z, [M-H]- = 297.1 m/z.
Purity: 98 % (NMR).
Preparative example 11
Compound 11 was obtained according to the following synthetic scheme:
Figure imgf000052_0001
9b (0.4 mmol, 1.0 equiv.) and methyl propargylether (1.0 mmol, 2.5 equiv.) were dissolved in pyrrolidine (1 mL) and cooled to 0°C under argon atmosphere. Cul (0.04 mmol, 0.1 equiv.), PdCl2(PPh3)2 (0.02 mmol, 0.05 equiv.) and a small amount of NH2OH·HCl were added to the above solution. The solution was stirred at rt for 2h. The mixture was diluted with 10 mL water, acidified with 2M HC1 and extracted with diethyl ether (3 x 10 ml). The combined organic layers were washed with brine (10 mL), dried over Na2SO4 and concentrated under reduced pressure. The crude product was purified by flash chromatography (SiO2, gradient cyclohexane- ethyl acetate; reversed phase C 18, gradient 10-100 % methanol in water). 11 was obtained with 67 % yield as a colorless oil.
1H-NMR (500 MHz, CDCl3-d): δ 4.14 (t, J= 1.0 Hz, 2H), 3.65 (s, 3H), 3.37 (s, 3H), 2.32-2.21 (m, 4H), 1.65-1.56 (m, 2H), 1.56-1.47 (m, 2H), 1.41-1.33 (m, 2H), 1.31-1.23 (m, 8H) ppm.
13C-NMR (125 MHz, CDCl3-d): δ 174.2, 81.2, 71.6, 71.5, 64.5, 60.2, 57.7, 51.4, 34.1, 29.2, 29.2, 29.1, 29.0, 28.7, 28.1, 24.9, 19.2 ppm. MS (ESI-Quad.): calc. = 278.39 g/mol, found [M+H] = 279.2 m/z, [M-H]- = did not ionize.
Purity: 98 % (NMR).
Preparative example 12
Compound 12 was obtained according to the following synthetic scheme:
Figure imgf000053_0001
9b (0.2 mmol, 1.0 equiv.) and methyl propargylether (0.5 mmol, 2.5 equiv.) were dissolved in pyrrolidine (1 mL) and cooled to 0°C under argon atmosphere. Cul (0.02 mmol, 0.1 equiv.), PdCl2(PPh3)2 (0.01 mmol, 0.05 equiv.) and a small amount of NH2OH·HCl were added to the above solution. The solution was stirred at rt for 2h. The mixture was diluted with 10 mL water, acidified with 2M HC1 and extracted with diethyl ether (3 x 10 ml). The combined organic layers were washed with brine (10 mL), dried over Na2SO4 and concentrated under reduced pressure. The intermediate was purified by flash chromatography (SiO2, gradient cyclohexane- ethyl acetate). LiOH·H2O (0.6 mmol, 3.0 equiv.) was added to a solution of the purified intermediate in THF/MeOH/water (5 mL, 6:1:1) at 0°C. The solution was allowed to warm to rt and stirred for 3.5h before water (10 mL) was added. The mixture was acidified with 3M HC1 and extracted with diethyl ether (3 x 10 mL). The combined organic layers were washed with brine (10 mL), dried over Na2SO4 and concentrated under reduced pressure. The crude product was purified by flash chromatography (SiO2, gradient cyclohexane-ethyl acetate). 12 was obtained with 48 % yield as a white solid.
1H-NMR (500 MHz, CDCl3-d): δ 4.17 (t, J= 1.1 Hz, 2H), 3.41 (s, 3H), 2.37 (t, J= 7.5 Hz, 2H), 2.29 (tt, J= 7.0, 1.1 Hz, 2H), 1.69-1.61 (m, 2H), 1.58-1.51 (m, 2H), 1.41-1.28 (m, 10H) ppm.
13C-NMR (125 MHz, CDCl3-d): δ 179.9, 81.3, 71.5, 71.5, 64.5, 60.2, 57.7, 34.0, 29.2, 29.2, 29.0, 29.0, 28.8, 28.1, 24.7, 19.2 ppm. MS (ESI-Quad.): calc. = 264.36 g/mol, found [M+H]+ = 265.1 m/z, [M-H]- = 263.1 m/z, [M- H+FA]- 309.1 m/z.
Purity: 98 % (NMR).
Preparative example 13
Compound 13 was prepared according to the following synthetic scheme:
Figure imgf000054_0001
9b (0.2 mmol, 1.0 equiv.) and ethynylcyclopropane (0.5 mmol, 2.5 equiv.) were dissolved in pyrrolidine (1 mL) and cooled to 0°C under argon atmosphere. Cul (0.02 mmol, 0.1 equiv.), PdCl2(PPh3)2 (0.01 mmol, 0.05 equiv.) and a small amount of NH2OH·HCl were added to the above solution. The solution was stirred at rt for lh. The mixture was diluted with 10 mL water, acidified with 2M HC1 and extracted with diethyl ether (3 x 10 ml). The combined organic layers were washed with brine (10 mL), dried over Na2SO4 and concentrated under reduced pressure. The intermediate was purified by flash chromatography (SiO2, gradient cyclohexane- ethyl acetate). LiOH·H2O (0.6 mmol, 3.0 equiv.) was added to a solution of the purified intermediate in THF/MeOH/water (5 mL, 6:1:1) at 0°C. The solution was allowed to warm to rt and stirred for 3.5h before water (10 mL) was added. The mixture was acidified with 3M HC1 and extracted with diethyl ether (3 x 10 mL). The combined organic layers were washed with brine (10 mL), dried over Na2SO4 and concentrated under reduced pressure. The crude product was purified by flash chromatography (SiO2, gradient cyclohexane-ethyl acetate; reversed phase C18, gradient 10-100 % methanol in water). Compound 13 was obtained with 24 % yield as a white solid.
1H-NMR (500 MHz, CDCl3-d): δ 2.37 (t, J= 7.5 Hz, 2H), 2.25 (t, J= 7.0 Hz, 2H), 1.69-1.60 (m, 2H), 1.55-1.47 (m, 2H), 1.43-1.22 (m, 11H), 0.90-0.68 (m, 4H) ppm. 13C-NMR (125 MHz, CDCl3-d): δ 80.4, 77.1, 65.4, 60.7, 34.0, 29.2, 29.2, 29.0, 29.0, 28.8, 28.3, 24.7, 19.2, 8.7 (2C), 0.0 ppm; Remark: 1 C quart, signal is not visible
MS (ESI-Quad.): calc. = 260.37 g/mol, found [M+H]+ = 261.1 m/z, [M-H]- = 259.1 m/z, [M- H+FA]- = 305.1m/z.
Purity: 98 % (NMR)
Preparative example 14
Compound 14 was prepared according to the following reaction scheme:
Figure imgf000055_0001
DMAP (0.216 mmol, 2.4 equiv.) was added to a suspension of EDAC (0.117 mmol, 1.3 equiv.) in DCM (2 mL). The mixture was stirred at rt until all solids were dissolved. Compound 13 (0.09 mmol, 1.0 equiv.) and methanesulfonamide (0.09 mmol, 1.0 equiv.) were added to the reaction mixture at 0°C. The mixture was stirred at rt for 4h. The reaction mixture was diluted with water (10 mL) and acidified to pH 1 with 3M HC1 and the aqueous layer was extracted with EtOAc (2 x 10 mL). The combined organic layers were washed with saturated NH4CI solution (10 mL) and brine (10 mL), dried over Na2SO4 and concentrated under reduced pressure. The crude product was purified by flash chromatography (SiO2, gradient cyclohexane- ethyl acetate). 14 was obtained with 75 % yield as a white solid.
1H-NMR (500 MHz, CDCl3-d): δ 3.33 (s, 3H), 2.34 (t, J= 7.5 Hz, 2H), 2.25 (td, J= 7.0, 0.9 Hz, 2H), 1.73-1.62 (m, 2H), 1.57-1.45 (m, 2H), 1.41-1.24 (m, 11H), 0.90-0.68 (m, 4H) ppm.
13C-NMR (125 MHz, CDCl3-d): δ 172.1, 80.5, 77.1, 65.5, 60.7, 41.6, 36.5, 29.2, 29.1, 29.0, 28.9, 28.7, 28.3, 24.4, 19.2 6.7 (2C), 0.0 ppm. MS (ESI-Quad.): calc. = 337.48 g/mol, found [M+H]+ = 338.2 m/z, [M+Na]+ = 360.1 m/z, [M+Na+ACN]+= 401.1 m/z, [M-H]- = 336.1 m/z.
Purity: 95 % (NMR).
Antifungal activity data
The MIC values measured for the exemplary compounds are given in Table 1. All MIC-values reported herein are measured using the SOP of the The European Committee on Antimicrobial Susceptibility Testing (EUCAST - https://www.eucast.org/astoffungi, EUCAST E.DEF 7.3.1 for Candida spp. and EUCAST E.DEF 9.3.1 for Asperigillus spp).
Flat-bottom well microdilution plates are used to add 100 μL of antifungal test solution in double strength RPMI 1640 (with L-glutamine and a pH indicator but without bicarbonate) supplemented with glucose to a final concentration of 2% (RPMI 2% G medium) plus 100 μL fungal species inoculum obtained from an 18-24 h culture on nutritive agar medium, in sterile distilled water with a final inoculum between 0.5 x 105 and 2.5 x 105 CFU/mL. A stock solution from powder test compound is prepared with RPMI 2% G medium to a concentration of 20.4 mg/L. 2-fold dilutions are prepared to 0.005mg/L and distributed to the microtiter plates. Microdilution plates are incubated without agitation at 35 ± 2 °C in ambient air for 48 h. Cell growth after 24 h and / or 48 h is determined by a microplate reader by measuring the UV- absorbance at 530 nm. EV-086 in concentrations of 10.2 mg/L, 0.04 mg/L and 0.005 mg/L are used as positive control and RPMI 2% G medium as negative control. Relative growth rates are calculated from one to three independent experiments and the compound concentration leading to a <10% reduction of growth is reported as MIC-value.
Fungal strains used are: C. albicans (Candida albicans (DSMZ 11225), Candida krusei (DSMZ 6128), Candida parapsilosis (DSMZ 5784) and Aspergillus niger (DSMZ 1957) were obtained from Deutsche Stammsammlung für Mikrobiologie und Zellkulturen (DSMZ) in DE-38124 Braunschweig. Aspergillus flavus (ATCC MYA-3631), Aspergillus fumigatus Fresenius (ATCC MYA-3626), Trichophyton rubrum (ATCC MYA-4438) and Trichophyton interdigitale Priestley (ATCC MYA-4439) were obtained from the American Type Culture Collection (ATCC) F-67123 Molsheim. Plasma stability
The determination of the plasma stability of the antifungal compounds took place in human serum. The compounds were incubated, and the recovery thereof was measured at four timepoints as described below. The described procedure bases on a protocol described before (Di, L., Kerns, E.H., 2016. Drug-Like Properties: Concepts, Structure, Design, and Methods from ADME to Toxicity Optimization. Elsevier, UK. ISBN: 978-0-12-801076-1).
The human serum was 50:50 diluted with phosphate buffered saline (PBS, pH 7.4, v:v) and preincubated for 30 min at 37.5 °C. Afterwards, 12 μL of the compound stock solution (cStock = 1 mmol/L in DMSO) was added to 500 μL plasma in a 1.5 mL centrifuge tube (cAssay = 23.43 μmol/L). This was done for each compound and each timepoint in triplicates (12 tubes per compound). The tubes were then incubated for 0, 60, 120 and 180 minutes at 37.5 °C under continuous shaking at 50 rpm. The reaction was then quenched using 500 μL cold (0 °C) acetonitrile. After vortexing, the solution was kept in the fridge (4 °C) for 20 to 30 min, before the tubes were centrifuged at 20’ 000 ref for 10 min. After the centrifugation, 400 μL of the clear supernatant was transferred into a 1.1 mL UHPLC glass vial and then measured. As controls the compounds benfluorex and propranolol were used. The first is reported to have a poor plasma stability and the latter a good one. Also, as a control (blank), serum without compounds was incubated to gain information about the background and possible changes in the background.
In Figure 2, the results of the measurement of the plasma stability of the antifungal compounds are shown. The ID of the compound is shown on the x-axis and the recovery rate on the y-axis. The error bars indicate the standard errors of the measured triplicates. For each compound there are four bars which show the recovery rates of the measurements of the four timepoints 0, 60, 120 and 180 minutes (T0 to T3, respectively).
The further data in Figure 2 shows that, for each compound except 8, there was a decrease of the compound recovery rates over the timepoints Tl, T2 and T3 (60 min, 120 min and 180 min incubation). Especially, the compounds 13, 10 and EV-086 showed a high decrease from T 1 to T3. Compounds 5 and 8 showed a significantly lower decrease in the recovery rate from T 1 to T3, which is indicative of their increased plasma stability.

Claims

1. A compound having the formula (I):
R1-Z-C≡C-C≡C-R3
(I) and all stereoisomers, racemic mixtures, tautomers, pharmaceutically acceptable salts, prodrugs, hydrates, solvates and polymorphs thereof, wherein
R1 is selected from HO-NH(O=)C-, C1-4 alkyl-O-NH(O=)C-
C1-4 alkyl-SO2-NH-(O=)C-,CiM alkyl-O-C(=O)-O-
C1-4 alkyl-C(=O)-O-CH2-O-(O=)C-, C1-4 alkyl-NH-(O=)C-
C1-4 alkyl-C(=O)-NH-CH2-NH-, C1-4 alkyl-C(=O)NH-C(=O)-NH-
C1-4 alkyl-SO2-NH-C(=O)-NH-, C1-4 alkyl-C(=O)N(OH)-, C1-4 alkyl-O-C(=O)-NH-
, C1-4 alkyl-C(=O)-NH-O- C1-4 alkyl-NH-C(=O)-O- CF3CH(OH)-, C1-4 alkyl-SO2NH-
Figure imgf000058_0001
Figure imgf000059_0001
Z represents an alkylene moiety having 6 to 12 carbon atoms and optionally containing 1 or 2 double bonds, wherein
(i) one or more non-adjacent -CH2- groups can be independently replaced by -O-, -S-, -S(O)-, -C(O)-N(H)-, or-C(O)-N(C 1-4 alkyl)-, and/or
(ii) one or more hydrogen atoms can be independently replaced by -F, -OH or C1-4 alkyl, and/or
(iii) one or more -CH2-CH2- moieties can be independently replaced by a catenary 3- to 6-membered heterocyclic ring, and/or
(iv) one or more -CH2-CH2-CH2- moieties can be independently replaced by a catenary 3- to 6-membered heterocyclic ring; and
R3 is selected from
(i) an optionally substituted heterocyclic or carbocyclic group having 5 to 10 ring atoms, wherein the optional substituent is independently selected from -C 1-4 alkyl, -OH, -OC 1-4 alkyl, -Hal, -NH2, -NH(C 1-4 alkyl), -N(C 1-4 alkyl)2, and -NO2, or
(ii) an alkyl group containing 1 to 4 carbon atoms, wherein one divalent carbon atom can be replaced by oxygen or sulfur and wherein the alkyl group R3 can be optionally substituted by one or more Hal.
2. The compound according to claim 1, wherein R1 is selected from HO-NH(O=)C-
C 1-4 alkyl-O-NH(O=)C-, C 1-4 alkyl-SO2-NH-(O=)C-,and
Figure imgf000059_0002
3. The compound according to claim 1 or 2, wherein Z is -(CH2)7-CH=CH- or -(CH2)7-CH2-CH2-, preferably -(CH2)7-CH=CH-.
4. The compound according to any one of claims 1 to 3, wherein R3 is optionally substituted as defined in claim 1 and is selected from furane, thiophene, norbornane, cyclopropyl, pyrrole, oxazole, isoxazole, thiazole, isothiazole, pyrazole, and imidazole, preferably R3 is furane.
5. The compound according to any one of claims 1 to 4, wherein the compound is selected from:
Figure imgf000060_0001
6. A compound of formula (I):
R1-Z-C≡C-C≡C-R3
(I) and all stereoisomers, racemic mixtures, tautomers, pharmaceutically acceptable salts, prodrugs, hydrates, solvates and polymorphs thereof, wherein
R1 is selected from -COOH, a moiety that can be hydrolyzed to -COOH, HO-NH(O=)C-, C1-4 alkyl-O-NH(O=)C-, C1-4 alkyl-SO2-NH-(O=)C-
C1-4 alkyl-O-C(=O)-O- C1-4 alkyl-C(=O)-O-CH2-O-(O=)C- C1-4 alkyl-NH-(O=)C- C 1-4 alkyl-C(=O)-NH-CH2-NH-,
C1-4 alkyl-C(=O)NH-C(=O)-NH-, C1-4 alkyl-SO2-NH-C(=O)-NH-, C1-4 alkyl-C(=O)N(OH)-, C1-4 alkyl-O-C(=O)-NH- C 1-4 alkyl-C(=O)-NH-O-, C1-4 alkyl-NH-C(=O)0-, CF3CH(OH)-, C1-4 alkyl-SO2NH-
Figure imgf000061_0001
Z represents an alkylene moiety having 6 to 12 carbon atoms and optionally containing 1 or 2 double bonds, wherein
(i) one or more non-adjacent -CH2- groups can be independently replaced by -O-, - S-, -S(O)-, -C(O)-N(H)-, or-C(O)-N(C1-4 alkyl)-, and/or
(ii) one or more hydrogen atoms can be independently replaced by -F, -OH or C1-4 alkyl, and/or
(iii) one or more -CH2-CH2- moieties can be independently replaced by a catenary 3- to 6-membered heterocyclic ring, and/or
(iv) one or more -CH2-CH2-CH2- moieties can be independently replaced by a catenary 3- to 6-membered heterocyclic ring; and
R3 represents an aliphatic group containing 1 to 4 carbon atoms, wherein one divalent carbon atom can be replaced by oxygen or sulfur and wherein the alkyl group R3 can be optionally substituted by one or more Hal.
7. The compound according to claim 6, wherein R1 is selected from HO-NH(O=)C- C1-4 alkyl-O-NH(O=)C- C1-4 alkyl-SO2-NH-(O=)C- and
Figure imgf000062_0001
8 The compound according to any one of claims 6 or 7, wherein R1 is selected from -COOH, -COONa and -COOK.
9. The compound according to any one of claims 6 to 8, wherein Z is -(CH2)7-CH=CH-or -(CH2)7-CH2-CH2- preferably -(CH2)7-CH=CH-.
10 The compound according to any one of claims 6 to 9, wherein R3 is selected from -CH2CH2CFh -CH2OCH3, -CH2SCH3, -CHFCH2CH3, -CF2CH2CH3, preferably R3 is -CH2CH2CH3.
11 The compound according to any one of claims 6 to 9, wherein R3 is cyclopropyl or cyclobutyl.
12 The compound according to any one of claims 1 to 11, wherein the compound has a minimum inhibitory concentration of at most 1.0 mg/L against Candida albicans.
13. The compound according to any one of claims 1 to 12 for use as a medicament.
14. A pharmaceutical composition comprising the compound according to any one of claims 1 to 12 and optionally a pharmaceutically acceptable carrier.
15. The compound according to any one of claims 1 to 12 for use in the treatment, alleviation or prevention of a fungal infection.
16 The compound according to claim 15, wherein the fungal infection is caused by a fungus selected from Candida spp. (for example C. albicans, C. auris, C. krusei, C. glabrata, C. tropicalis, C. parapsilosis , C. guilliermondii , C. haemulonii , C. lusitaniae , C. lipolytica , C. norvegensis, C. viswanathii, C. kejyr or C. dubliniensis ), Aspergillus spp. (for example A. fumigatus, A. flavus, A. niger or A. terreus ) Histoplasma capsulatum , Coccidioides immitis , Coccidioides posadasii, Cryptococcus spp. (for example C. neoformans (for example var. neoformans or var. gattii ), C. bidus, C. laurentii , or C. fusarium ), Zygomycetes (such as Saksenaea vasiformis ), Malassezia spp. (for example M. furfur or M. globosa ), Hyalohyphomycetes (for example Scedosporium spp., such as S. prolificans or S. apiospermum ), Dermatophytes (for example Trichophyton spp. (for example T. mentagrophytes, T. rubrum or T. tonsurans ), Epidermophyton floccosum , Microsporum spp (for example M cookei , M cams, M. vanbreuseghemii , M gallinae or M gypseum or Trichosporon terrestre), Blastomyces dermatitidis , Sporothrix schenkii , Chromomycotic fungi (for example Fonsecaea pedrosoi, F. compacta , Cladophylophora carrionii or Phialophora verrucosa) and Madurella spp. (for example M mycetomatis or M griseum), Pneumocystis jirovecii , Pneumocystis carinii.
PCT/EP2022/070389 2021-07-20 2022-07-20 Novel antifungal compounds WO2023001905A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
EP21186586.0 2021-07-20
EP21186586 2021-07-20

Publications (1)

Publication Number Publication Date
WO2023001905A1 true WO2023001905A1 (en) 2023-01-26

Family

ID=76999666

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/EP2022/070389 WO2023001905A1 (en) 2021-07-20 2022-07-20 Novel antifungal compounds

Country Status (1)

Country Link
WO (1) WO2023001905A1 (en)

Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2001025197A1 (en) 1999-10-07 2001-04-12 Phytera, Inc. Antifungal enediynes
WO2011006061A1 (en) 2009-07-10 2011-01-13 Evolva Sa Diyne compositions
WO2011134538A1 (en) 2010-04-30 2011-11-03 Evolva Ltd Diyne compositions
US20130260054A1 (en) * 2012-03-27 2013-10-03 Massachusetts Institute Of Technology Three-Dimensional Photoresists via Functionalization of Polymer Thin Films Fabricated by iCVD

Patent Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2001025197A1 (en) 1999-10-07 2001-04-12 Phytera, Inc. Antifungal enediynes
WO2011006061A1 (en) 2009-07-10 2011-01-13 Evolva Sa Diyne compositions
WO2011134538A1 (en) 2010-04-30 2011-11-03 Evolva Ltd Diyne compositions
US20130260054A1 (en) * 2012-03-27 2013-10-03 Massachusetts Institute Of Technology Three-Dimensional Photoresists via Functionalization of Polymer Thin Films Fabricated by iCVD

Non-Patent Citations (5)

* Cited by examiner, † Cited by third party
Title
ANTIMICROBIAL AGENTS AND CHEMOTHERAPY, vol. 58, 2014, pages 455 - 456
DI, L.KERNS, E.H.: "Drug-Like Properties: Concepts, Structure, Design, and Methods from ADME to Toxicity Optimization", 2016, AMERICAN PHARMACEUTICAL ASSOCIATION
GOODMANGILMAN: "The Pharmacological Basis of Therapeutics", 1992, MCGRAW-HILL, INT. ED., article "Biotransformation of Drugs", pages: 13 - 15
KNECHTLE PHILIPP ET AL: "The Natural Diyne-Furan Fatty Acid EV-086 Is an Inhibitor of Fungal Delta-9 Fatty Acid Desaturation with Efficacy in a Model of Skin Dermatophytosis", ANTIMICROBIAL AGENTS AND CHEMOTHERAPY, vol. 58, no. 1, January 2014 (2014-01-01), US, pages 455 - 466, XP055963648, ISSN: 0066-4804, Retrieved from the Internet <URL:https://journals.asm.org/doi/pdf/10.1128/AAC.01443-13> DOI: 10.1128/AAC.01443-13 *
RADCHENKO O. A. ET AL: "ChemInform Abstract: Diacetylenic Hydroxamic Acids.", CHEMINFORM, vol. 20, no. 5, 31 January 1989 (1989-01-31), XP055964064, ISSN: 0931-7597, Retrieved from the Internet <URL:https://api.wiley.com/onlinelibrary/tdm/v1/articles/10.1002%2Fchin.198905154> DOI: 10.1002/chin.198905154 *

Similar Documents

Publication Publication Date Title
US6407129B1 (en) Azole compounds, their production and their use
RU2727520C2 (en) Antifungal agents
EP1143959B1 (en) Antifungal activity of dicationic molecules
EP2776402A1 (en) Meldrum &#39;s acid, barbituric acid and pyrazolone derivatives substituted with hydroxylamine as hno donors
JP3108997B2 (en) Azole compounds, their production and use
JP5132569B2 (en) Pharmaceutical composition containing phenylamidine derivative and method of using antifungal agent in combination
EP3010896B1 (en) New macrocyclic amidinourea derivatives, methods of preparation and uses thereof as chitinase inhibitors
US20080213398A1 (en) Modified Azole Compounds as Antifungal and Antibacterial Agents
WO2023001905A1 (en) Novel antifungal compounds
JP2006500373A (en) Azole derivatives as antifungal agents
KR20190087547A (en) Compositions and methods for the treatment of fungal infections
RU2345769C2 (en) Combined voriconasol and antimycotic inhibitor cyp2c19
RU2690161C1 (en) 3,5-substituted thiazolidine-2,4-dione derivatives, having antimicrobial activity
EP3060549B1 (en) Novel antifungal oxodihydropyridinecarbohydrazide derivative
JP6811727B2 (en) Antifungal 4-(4-(4-(((3R, 5R) -5-((1H-1,2,4-triazole-1-yl) methyl) -5- (2,4-difluorophenyl) methyl) ) Tetrahydrofuran-3-yl) Methoxy) -3-methylphenyl) Piperazin-1-yl) -N- (2-Hydroxycyclohexyl) benzamide, or a pharmaceutically acceptable salt thereof
RU2662153C1 (en) Hybrid ethers based on derivatives of thiazolidine-2,4-diona and azoles (1h-1,3-imidazole and 1h-1,3,4-triazole) and application thereof
US6960600B2 (en) Azole or triazole derivatives, method for preparing the same and use thereof as antifungal medicaments
RU2814730C1 (en) New potential antifungal agents based on thiazolidine-2,4-dione and triazole
EP3906969A1 (en) Antihypertensive polyol compound and derivative thereof
US6583164B1 (en) Antimycotic drug composition
JP2023513196A (en) Triazole derivatives with antifungal activity
WO2002051408A1 (en) Azole compounds as anti-fungals agents
KR102653821B1 (en) Antifungal 4-(4-(4-(((3R,5R)-5-((1H-1,2,4-triazol-1-yl)methyl)-5-(2,4-difluoro Phenyl) tetrahydrofuran-3-yl) methoxy) -3-methylphenyl) piperazin-1-yl) -N- (2-hydroxycyclohexyl) benzamide or a pharmaceutically acceptable salt thereof.
CN114920697A (en) Heterocyclic group substituted indan propionamide compound and application thereof
WO2023187607A1 (en) Nickel n-heterocyclic carbene complexes based on xanthines and use therof

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: 22753686

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE