AU773405B2 - Hydroxyitraconazole enantiomer mixtures - Google Patents

Hydroxyitraconazole enantiomer mixtures Download PDF

Info

Publication number
AU773405B2
AU773405B2 AU14775/02A AU1477502A AU773405B2 AU 773405 B2 AU773405 B2 AU 773405B2 AU 14775/02 A AU14775/02 A AU 14775/02A AU 1477502 A AU1477502 A AU 1477502A AU 773405 B2 AU773405 B2 AU 773405B2
Authority
AU
Australia
Prior art keywords
mixture
isomers
hydroxyitraconazole
itraconazole
water
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Ceased
Application number
AU14775/02A
Other versions
AU1477502A (en
Inventor
Yaping Hong
Patrick Koch
John R. Mccullough
Chris Hugh Senanayake
Gerald J. Tanoury
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Sunovion Pharmaceuticals Inc
Original Assignee
Sepracor Inc
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
Priority claimed from AU48027/00A external-priority patent/AU4802700A/en
Application filed by Sepracor Inc filed Critical Sepracor Inc
Publication of AU1477502A publication Critical patent/AU1477502A/en
Application granted granted Critical
Publication of AU773405B2 publication Critical patent/AU773405B2/en
Anticipated expiration legal-status Critical
Ceased legal-status Critical Current

Links

Landscapes

  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Plural Heterocyclic Compounds (AREA)

Description

2R,4S-HYDROXYITRACQNAZOLE
ISOMERS
Field of the Invention The present invention relates to a method of treating fungal infections employing any combination of two or more of the four isomers of 2 R,,4S-hydroxyitraconazole, and phosphate and sulfate derivatives thereof.
Background of the Invention Itraconazole, a well-known antifungal agent is defined in the USAN and USP Dictionary of Drug Names as 4 4 4 4 -[[2-(2,4-dichlorophenyl)-2-( IH-i 2 ,4-triazol-lylmethyl)- 1 ,3-dioxolan-4-yijmetnoxyjpnenyij- I-piperazinyl]phenyl ]-2,4-dihydro--. 1methylpropyl)-3H-l1,2,4-triazol-3-one or alternatively as &_)-lI-sec-butyl-4-fp.4..p- ,S)2-24dihorp~yl -I H- I ,2,4-triazol- I -ylmethyl)- 1 ,3-diloxolan-4yl]methoxy~phenyl]. I-piperazinyllphenyl]-A 2 l, 2 ,4-triazolin-5-one. The commercially :available material is the cis isomer in the dioxolane ring and is represented by the structural formula I: C I C H
I.
0
N.
oH NH
N
It will be noted that there are three asymmetric carbons in formula I (denoted by asterisks): two in the dioxolane ring and one in the sec-butyl side chain on the triazolone. There are eight possible isomers of a structure having three asymmetric carbons: (.RARS), and (SSR). Because the commercially available itraconazole is a cis isomer, it comprises a mixture of only those isomers that have a cis relationship in the dioxolane ring. Adopting the convention that the first denoted chiral center is at C-2 of the dioxolane ring, the second is at C-4 of the dioxolane and the third is in the sec-butyl group, commercial itracona-zole is a mixture of and isomers. Compounds of this invention have the (2R,4S) configuration in the dioxolane ring and are substantially free of the SR isomers.
The hydroxylation of the methylene carbon of the sec-butyl side chain creates an additional chiral center and gives rise to eight additional possible stereoisomers. The methods of the present invention employ a mixture of at least two of the four isomers of the two asymmetric centers in the butyl chain, i.e. a mixture ofRSRR, RSRS, RSSR and RSSS; or a mixture ofRSRR, RSRS and RSSR; or a mixture of RSRR and RSSR; etc.
CH3 C
N
N 'c OH 10 The graphic representations ofracemic, ambiscalemic and scalemic or enantiomerically pure compounds used herein are taken from Maehr J. Chem. Ed. 62, 114-120 (1985): solid and broken wedges are used to denote the absolute configuration of a chiral element; wavy lines indicate disavowal of any stereochemical implication which the bond it represents could generate; solid and broken bold lines are geometric 15 descriptors indicating the relative configuration shown but denoting racemic character; and wedge outlines and dotted or broken lines denote enantiomerically pure compounds of indeterminate absolute configuration. Thus, among the structures below, those having open wedges are intended to encompass both of the pure enantiomers of that pair, those having solid wedges are intended to encompass the single, pure enantiomer having the absolute stereochemistry shown.
Itraconazole is an orally active, broad-spectrum anti-fungal agent and is structurally related to miconazole and clotrimazole. It impairs the synthesis of ergosterol, which is the principal sterol of fungal cell membranes. This presumably results in an increased permeability and leakage ofintracellular content. At high concentration, cellular internal organelles involute, peroxisomes increase, and necrotic changes occur.
Following oral administration, itraconazole is slowly absorbed. Peak plasma levels are attained after 15 days of daily administration, and the pharmacokinetic behavior of itraconazole is nonlinear. The compound is eventually metabolized through the biologically active hydroxyitraconazole to several inactive metabolites. Metabolism is apparently through hepatic mechanisms, and in most subjects no metabolites are excreted in the urine [see, Hardin et al., Antimicro. Agents and Chemotherapy 32, 1310- 10 1313 (1988)].
The racemic mixture of itraconazole has been approved for use as an antifungal agent for blastomycosis and histoplasmosis. The compound is also being investigated for use in aspergillosis, coccidioidomycosis, cryptococcosis, onychomycosis, dermatophyte and candidiasis infections.
15 Systemic fungal diseases (systemic mycoses) are usually chronic, very slowly S developing conditions induced by opportunistic causative fungi which may not normally be pathogenic. However when they enter a host compromised by HIV, ionizing irradiation, corticosteroids, immunosuppressives, etc. or by such conditions as emphysema, bronchiectasis, diabetes mellitus, leukemia, burs and the like, they may become pathogenic. Symptoms in such fungal diseases may include fever, chills, anorexia and weight loss, malaise, and depression. Fungal diseases are often confined to typical anatomic distributions, and many involve a primary focus in the lung, with more characteristic manifestations of specific fungal infections when the fungus disseminates from a primary focus. For example, coccidioidomycosis occurs in a primary form as an acute, benign, self-limiting respiratory disease, with progressive disease developing from the primary form as a chronic, often fatal infection of the skin, lymph glands, spleen and liver. Similarly, blastomycosis primarily involves the lungs, and occasionally spreads to the skin. Other infectious diseases such as candidiasis and paracoccidioidomycosis offer a different course, and depending on the etiology may exhibit several forms involving the skin, mucous membranes, lymph nodes, and internal organs.
Superficial fungal infections are caused by dermatophytes or fungi that involve the outer layers of the skin, hair or nails. The infections may result in a mild inflammation, and cause intermittent remissions and exacerbations of a gradually extending, scaling, raised lesion. Yeast infections including candidiasis, and oral candidiasis (thrush) are usually restricted to the skin, and mucous membranes, and the symptoms vary with the site of infection.
Adverse effects associated with the administration of itraconazole include 10 hepatotoxicity and inhibition of drug metabolism in the liver, leading to numerous, clinically significant, adverse drug interactions. [See, Gascon and Dayer Eur. J. Clin.
Pharmacol. 41, 573-578 (1991) (interaction with midazolam); Honig et al. J. Clin.
:Pharmacol. 33, 1201-1206 (1993) (interaction with terfenadine); and Neuvonen et al.
Clin. Pharmacol. Therap. 60, 54-61 (1996) (lovastatin).] Hypersensitivity reactions including urticaria and elevations in serum liver enzymes are also associated with the administration of the drug. Hepatoxicity is a less common but more serious adverse S effect. Indeed, the use of oral conazoles as first line antifungals is usually discouraged because of the potentially serious consequences of the low incidence of hepatotoxicity [See, Lavrijsen et al., Lancet 340, 251-252 (1992)].
We have found evidence in our own studies in isolated guinea pig or rabbit hearts that the administration of racemic conazoles may be associated with an increased risk of cardiac arrhythmia. Arrhythmia has not been heretofore reported as a side effect of systemic itraconazole, although a particular subtype of arrhythmia, Torsades de Pointes, has been reported when racemic itraconazole was administered concurrently with terfenadine [Pohjola et al. Eur. J. Clin. Pharmacol. 45, 191-193 (1993)]. The lack of clinical reports of arrhythmia or QT anomalies may simply be a reflection of the fact that there is to date a relatively small subject population.
The relative non-polarity and insolubility of itraconazole give rise to two other drawbacks: it cannot be readily formulated in parenteral solution and it does not penetrate the blood-brain barrier. As a result, numerous therapeutic indications which require rapid achievement of efficacious blood levels or access to the CNS are beyond treatment with itraconazole. In particular, central candidiasis, which may be responsible for AIDS related dementia, cannot be treated with itraconazole.
Thus it would be particularly desirable to find a compound with the advantages of itraconazole which would not have the aforementioned disadvantages.
Summary of the Invention The various butyl chain isomers of 2R,4S hydroxyitraconazole and mixtures thereof -possess potent activity in treating local and systemic fungal, yeast and dermatophyte infections while avoiding adverse effects associated with the administration of itraconazole. The butyl S 15 chain isomers of 2R,4S hydroxyitraconazole and mixtures thereof also enjoy the particular advantage of being more soluble in physiologically compatible solutions than is itraconazole.
Moreover, we have found, quite surprisingly, that the side chain isomers of 2R,4S hydroxyitraconazole and in particular mixtures of such isomers produce blood levels of hydroxyitraconazole that are significantly higher than the blood levels of hydroxyitraconazole and itraconazole that can be obtained from the administration ofracemic [mixture of(R,S,S), and isomers] itraconazole.
In the specification the term "comprising" shall be understood to have a broad meaning similar to the term "including" and will be understood to imply the inclusion of a stated integer or step or group of integers or steps but not the exclusion of any other integer or step or group of integers or steps. This definition also applies to variations on the term "comprising" such as "comprise" and "comprises".
In one respect the invention relates to a method for treating fungal infection comprising administering to a mammal suffering from said fungal infection a therapeutically effective amount of a mixture of isomers of formulae: N 4 1 (R,S,S,R)
CH
3 N2 OR 2 (R,S,R,S)
QH
3 CHN- OR 3 a a
N-N'
N
4 (R,S,S,S) wherein R is hydrogen, -P(O)(OH) 2
,-SO
3 H or a salt thereof.
In another aspect, the invention relates to the mixture of isomers above.
In another aspect, there is provided a 1:1 mixture of isomers of formulae:
CI
So
QH
3 N OHan and r r C1
CH
3 0_ MC HT-O Q NN Q NN
H
or a salt thereof.
a a
U,
In another aspect the invention relates to pharmaceutical compositions comprising the mixtures above and a pharmaceutically acceptable carrier.
Detailed Description of the Invention The four isomers cis-itraconazole are shown below: ci N. NOj-O 0ci S(RS,S) N l0- C I N*0I- 6 (RS,R) a a a a a 7 (SIRS) 0 8 (S,RR) 5 The eight isomers of cis-hydrox itraconazole are shown below: a.
a a Cl N I~~cI
N=
c I N OH 2
OH
cI N. 0- 3 (R,S,RR) NOH Ci N. 0 4 NOH
CI
N 0
OH
ci 9 (S,R,S,R)
OH
(S,R,R,S)
CI
N. CI 0- C 0 N'Q ,OaNN N N I-N
OH
11 (S,R,R,R) Ci N. Ci 0 SN-"1 0 N= r 12 (S,R,S,S) The isomers ofcis hydroxyitraconazole have been made according to Scheme 1 below in six forms: the pure R,S and S,R butyl chain isomers of 2R,4S and 2S,4R and the R,R/S,S mixture of butyl chain isomers of each of the 2R,4S and 2S,4R dioxolanes.
The synthetic approach is shown in detail for one enantiomer, 2 in Scheme 1; isomers 1, 9, and 10 may be made by the same synthesis employing other enantiomers of the starting materials. Similarly, the mixtures of RR/SS isomers in the butyl side chain, i.e.
mixtures of 3/4 and 11/12, will be produced from the prochiral cis cyclic sulfate corresponding to the chiral trans cyclic sulfate .o Scheme I 0 CHO -O 26 0 /'0 o 0
S
CI
<11 j 0 HO N 22~ HO CHrOTos TFAltoluenc 2 &c N
H
23 KB/I 8-C-6 0 qH 3 27 45% H~r 0 qH 3 HO NN-i -Ni H 28 S S
S.
S
S.
S
ci KOHJDIvfF N H CR N 6H 2 As shown in Scheme 1, the chiral dioxolane (23) is prepared by a stereospecific literature method from either R-3-tosyloxy-l ,2-propanediol or S-3-tosyloxy-1, 2-propanediol by acid-catalyzed ketalization to provide enantiomerically pure RS- or respectively.
The dioxothiolane 25 is prepared from a butanediol of appropriate configuration by treatment with thionyl chloride, followed by in situ oxidation of the resulting cyclic sulfite with sodium periodate in the presence of ruthenium trichloide. 2.4-Dihv dro-4-[4-[4-(4mehxphenyl)-piperazinyl]phenyll-3H-l1,2A-triazol-3 -one prepared by the method of example XVII in US patent 4;267.,179 (described below), is reacted with the dioxothiolane prepared by the procedure of Gao and Sharpless Am. Chem. Soc. 1 10. 75 38 (1988)].
using potassium hydride in DMF in the presence of crown ether. The resulting rnethoxysulfate salt is cleaved to the phenol-alcohol 28 by heating with 48% I/o Br at 100-110 'C.
The tosyl ester 23 and the phenol-alcohol 28 are reacted in the presence of potassium hydroxide in DMF to provide the substantially enantiomerically pure product 2. Isomers 1, 9, 10, 3+4, and 11+12 are prepared in analogous fashion. Their rotations are shown in Table 1:
S.
@59
S.
.5 S.
S
S
9'.
10 Table 1 Compound [a" 25 (c 0.1, MeOH) 1 +12.70 2 +22.3" 9 -22.00 10 -10.6° 3+4(mix) +19.70 11+12(mix) -18.5° 5 +19.2 6 +14.0 7 -13.4 8 -18.7 When individual isomers 3, 4, 11 and 12 are desired, they may be obtained by chromatography by methods well known in the art or by the methods described in PCT applications W098/21204 and W098/21205, the pertinent portions of which are laid out 20 below.
When it is desired to prepare the phosphate derivatives, 28 is treated first with t-butyldimethylsilyl chloride and diisopropylethylamine to protect the phenol, then with dibenzyl diisopropylphosphoramidite and t-butylhydroperooxide according to the procedure of PCT application W095/17407, which is incorporated herein by reference, to phosphorylate the alcohol. The silyl protecting group is removed with anhydrous tetrabutylammonium fluoride and the benzyl-protected phosphate is coupled with the doxolane tosylate as in Scheme 1. The benzyl protecting groups are cleaved by hydrogenolysis in the presence of a palladium catalyst to provide the phosphate product.
(2R,4S)-2-(2,4-dichlorophenyl)-2-(1 H- 1,2,4-triazol--ylmethyl)-4-tosyloxymethyl- 1,3dioxolane tosylate DTTT (23 tosylate salt) is prepared as follows: A suspension of tosyloxy-1,2-propanediol (10.0 g, 40 mmol) and 1-(2,4-dichlorophenyl)-2-(1H-1,2,4-triazol- 1,2,4-triazol-l-yl) ethanone (10.0 g, 39 mmol) in toluene (50mL) is cooled to 5" C.
Triflic acid (15 mL, 4 eq) is slowly added so that the temperature stays below 15° C.
After complete addition the reaction mixture (2 phases) is stirred at 250 C for 60 h. The mixture is diluted with ethyl acetate (EtOAc) (200 mL) and slowly dropped into a solution of K 2
CO
3 (50 g) in water (400 mL) at 5 C. The organic layer is separated and the aqueous layer washed with EtOAc (150 mL). The combined organic extracts are dried over Na 2
SO
4 (10 g) and filtered. A solution of toluenesulfonic acid (TsOH) (7.6 g monohydrate in EtOAc (50 mL) is slowly added at 250 C. The white solid product is o filtered after 30 min, washed and dried to give cis DTTT containing 5% trans. Two 10 crystallyzations from CH 3 CN (400 mL) gives 13.5 g pure (2R,4S)-DTTT (50% yield); [a]D 25 +16.6° MeOH); ee 99.6%. The 2S,4R isomer is prepared in analogous fashion from (S)-tosyloxy-I,2-propanediol (14.8 g, 60 mmol) and 1-(2,4dichlorophenyl)-2-(lH-1,2,4-triazol-l-yl) ethanone in (44% yield); [aD] 25 -16.6° (c=l, MeOH) ee 99.6% .g o° 15 (4R,5R)-4,5-dimethyl-1,2,3-dioxathiolane 2,2-dioxide (25) is prepared as follows: A three-necked 500 mL round-bottom flask fitted with a reflux condenser and a calcium chloride drying tube was charged with (2R,3R)-(+)-2,3-butanediol (10.0 g, 10.1 ml, 0.11 mol) and carbon tetrachloride (120 mL). Thionyl chloride (16.0 g, 9.8 mL, 0.13 mol) was added dropwise at room temperature. Rapid gas evolution began. The reaction 20 mixture was stirred at room temperature for 10 min, then warmed to reflux for 30'nin to insure complete removal of HCI gas. The reaction mixture was cooled to 0° C in an icewater bath and acetonitrile (120 mL), RuCl 3
-H
2 0 (14 mg, 0.07 mmol), NaIO 4 (35.6 g, 0.17 mol) and water (180mL) were added, respectively. The reaction mixture was allowed to warm to room temperature and stirred for 1.5 hr. The mixture was poured into methyl t-butyl ether (900 mL), and water was added to dissolve the remaining NaI04 (ca. 600 mL). The phases were separated and the aqueous phase was extracted with methyl t-butyl ether (2 x 100mL). The combined organic phases were washed with water (1 x 50mL), saturated aqueous sodium bicarbonate (2 x 50mL) and saturated aqueous bicarbonate (2 x 50mL) and saturated aqueous sodium chloride (1 x The organic solution was dried over anhydrous magnesium sulfate and filtered through a bed of silica gel to give a clear and colorless solution. The solvent was removed in vacuo to give 16.01 g (95% yield) of the title compound.
To a suspension of potassium hydride (530 mg, 4.6 mmol, 35 wt% dispersion in oil), prewashed with hexane (2 x 10mL), in dimethylformamide (37 mL) at room temperature was added 18-crown-6 (980 mg, 3.7 mmol) and 2,4-dihydro-4-[4-[4-(4methoxyphenyl)-1-piperazinyl]phenyl]-3H-1,2,4-triazol-3-one (1.09 Ig, 3.3 mmol).
The solution was warmed to 80-85° C for 2 hr. then cooled in an ice-water bath to 00 C.
To this solution was added (4R,5R)-4,5-dimethyl- 1,2,3-dioxathiolane 2,2-dioxide S"(500 mg, 3.3 mmol). The reaction mixture warmed to 4" C. After recooling to 0° C, the reaction mixture was warmed to room temperature and stirred for 21 hours. To the reaction mixture was added 150 mL of toluene and 400 mL of methyl t-butyl ether, and the white precipitated product was filtered from the mixture. The solid was dried in vacuo to give 1.70 g (quantitative yield) of a 87:13 mixture (by HPLC) of potassium (2R,3S)-3-[2,4-dihydro-4-[4-[4-(4-methoxyphenyl]- -piperazinyl]phenyl]-3H- 1,2,4- 15 triazol-3-on-2-yl]but-2-yl sulfate the title compound and triazolone starting material. A portion of this mixture was purified by flash chromatography (gradient from 95:5 chloroform:methanol to methanol) for characterization.
To potassium (2R,3S)-3-[2,4-Dihydro-4-[4-[4-(4-methoxyphenyl- 1piperazinyl]phenyl]-3H-1, 2 4 -triazol-3-on-2-yl]but-2-yl[sulfate(7) (1.50 g, 2.98 mmol) and sodium sulfite (84 mg, 0.67 mmol) was added 48% HBr (6.0 mL). The solution was heated to 110-115 C for 7 hr and cooled to room temperature. The reaction mixture was poured into a wide-mouthed beaker and the pH raised to 7 by slow addition of solid potassium carbonate. Water was added (100 mL), and the product was collected by filtration and dried in vacuo. Flash chromatography of the crude material eluting with a gradient of chloroform to 95:5 chloroform:methanol gave 1.03 g yield) of 2,4-dihydro-4-[4-[ 4 -(4-hydroxyphenyl)- 1-piperazinyl]phenyl]-2-[(1S,2R)-2hydroxy- -methylpropyl)]-3H-1,2,4-triazol-3-one (28) as an adduct with SO 3 -13- To 2,4-dihydro-4-[4- [4-(4-hydroxyphenyl)-l1-piperazinyl]phenyl]-2- [1S,2R)-(2hydroxy- 1-methylpropyl)]-3H- 1,2,4-triazol-3 one SO 3 adduct (28) (420 mg, 0.86 mmol) and (1 )-(2S,4S-2-(2,4-dichlorophenyl)-2-( 1H-i ,2,4-triazol-l-ylmethyl-4-tosyloxymethyl- 1,3 dioxolane tosylate (23 tosylate) (637 mg., 0.97 mmol) was added powdered potassium hydroxide (277 mg, 4.94 mmol) and NN-dimethylformamide (15 mL). The reaction mixture was warmed to 50-55' C for 8 hr and cooled to room temperature. Water was added (150 mL) and the crude product was collected by filtration and dried in vacuc. Purification by flash chromatography, eluting with ethyl acetate, followed by chloroform, 98:2 \chloroform:methanol, then 95:5 chloroform: methanol, gave 320 mg (52% yield) of (28, 4R)- 4- [[2,4-dichlorophenyl)-2-( 1H-i ,2,4-triazol-1-ylmethyl)- 1,3 -dioxolan-4yl]methoxy]phenyl] -1I -piperazinyl] -2,4-dihydro-2- lS,2,R)-(2-hydroxy- 1 -methylpropyl)] -3 H- 1,2,4,-triazol-3-one [MD 2 -22.0* MeOll).
A mixture of the 3 +4 isomers may be prepared according to Scheme Ia.
Scheme: Ia 150 0. 0 1 0 0 CH c 3 o 0 N N N I 26 IS 0 H CH 30 N N c N -CO 0 S0 3
K
27a kN~ 2 8a Preparation of meso-4.5-dimethyl-l,2,3-dioxathiolane 2,2-dioxide A three-necked 500 mL round-bottom flask fitted with a reflux condenser and a calcium chloride drying tube was charged with meso-2,3-butanediol (10.0 g, 10.1 mL, 0.11 mol) and carbon tetrachloride (20 mL). Thionyl chloride (15.98 g, 9.8 mL, 0.134 mol) was added dropwise at room temperature. Rapid gas evolution began. The reaction mixture was stirred at room temperature for 10 min, then warmed to reflux for 30 min to insure complete removal of HCI gas. The reaction mixture was cooled to 0° C in an ice-water bath and acetonitrile (120 mL), RuC1 3 H20 (14 mg, 0.07 mmol), NaI04 (35.6 0.166 mol) and water (180 mL) were added, respectively. The reaction mixture was allowed to warm to room 10 temperature and stirred for 1.5 hr. The mixture was poured into methyl t-butyl ether (900 mL) "•and water was added to dissolve the remaining NaI04 (ea. 600 mL). The phases were separated and the aqueous phase was extracted with methyl t-butyl ether (2 x 100 mL). The •combined organic phases were washed with water (1 x 50 mL), saturated aqueous sodium bicarbonate (2 x 650mL) and saturated aqueous sodium chloride (1 x 50 mL). The organic 15 solution was dried over anhydrous magnesium sulfate and filtered through a bed of silica gel to give a clear and colorless solution. The solvent was removed in vacuo to give 16.20 g (96% yield) of the title compound.
Preparation of Potassium (2R *,3R *)-3-[2,4[Dihydro-4-[4-[4-(4-methoxyphenyl]-1piperazinyl]phenyl]-3H-1,2,4-triazol-3-on-2-yl]but-2-yl Sulfate (27a).
20 To a suspension of potassium hydride (8.53 g, 74.4 mmol, 35% dispersion in oil), prewashed with hexane (2 x 50 mL) in N,N-dimethylformamide (120 mL) at room temperature was added 18-crown-6 (15.73 g, 59.5 mmol) and 2,4-dihydro-4-[4-[4-(4methoxyphenyl)-l-piperazinyl]phenyl]-3-h-1,2,4-triazol-3-one (26)(17.43 g, 49.6 mmol). The solution was warmed to 80-85° C for 1.5 hr, then cooled in an ice-water bath to 90 C. To this solution was added (4R*,5S*)-4,5-dimethyl-l,2,3-dioxathiolane 2,2-dioxide (8.00 g, 52.6 mmol). The reaction mixture exothermed to 130 C. After recooling to 00 C, the reaction mixture was warmed to room temperature and stirred for 18 hr. The mixture was poured into L of methyl t-butyl ether, and the solid product was filtered from the solution. Water (1 L) was added to the crude solid, and the mixture was warmed to 800 C and the solid by-product was removed by filtration. The solid was rinsed with water (400 mL), and the filtrate was allowed to cool to room temperature. Saturated aqueous potassium chloride (50 mL) was added and the product precipitated from the solution immediately. After filtration, the solid was dried in vacuo to give 13.46 g (47% yield) of the title compound.
Preparation of Racemic 2,4-Dihydro-4-[4-[4-(4-hydroxyphenyl)-l-piperazinyl]phenyl]-2- [(1S*,2R *)-2-hydroxy-l-methylpropyl)]-3H-1,2,4-triazol-3-one(28a).
To potassium (2R *,3R*)-3-[2,4-Dihydro-4-[4-[4-(4-methoxyphenyl)- 1piperazinyl]phenyl]-3H-1,2,4-triazol-3-on-2-yl]but-2-yl sulfate 97) (7.00 g, 12.9 mmol) and sodium sulfite (480 mg, 3.80 mmol) was added 48% HBr (40 mL). The solution was heated to reflux for 6 hr, then cooled to room temperature. The reaction mixture was poured into 10 water (100 mL) and the pH raised to 7-8 with saturated aqueous potassium carbonate solution.
The product was collected by filtration and dried in vacuo. The filtrate was extracted with chloroform (3 x 20 mL), and the combined organic fractions were dried over anhydrous magnesium sulfate, filtered and the solvent removed in vacuo to give a white solid. The solids were combined to give 3.89 g (62% yield) of the title compound as an adduct with SO 3 Preparation of (2R,4S)-4-[4-[4-[4-4-[[2-(2,4-dichlorophenyl)-2-(1 H-1,2,4-triazol-1 -ylmethyl)- 1,3-dioxolan-4-yl]methoxy]phenyl]-[-piperazinyl[phenyl]-2,4dihydro-2-[(1R*,2R hydroxy-1-methylpropyl)]-3H-1,2,4-triazol-3-one (3 4).
To 2,4-dihydro-4 [4-[4-(4-hydroxyphenyl)-1 -piperazinyl]phenyl]-2 [(1R *,2R 20 hydroxyl-l-methylpropyl)]-3H-1,2,4-triazol-3one S03 adduct (1.00 g, 2.04 mmol) and (2R, 4S)-2-(2,4-dichlorophenyl)-2-( 1 H-1,2,4-triazol-1 -ylmethyl)-4-tosyloxymethyl-1,3dioxolane tosylate (23) (1.52 g, 2.32 mmol) was added powdered potassium hydroxide (658 mg, 11.7 mmol) and N,N-dimethylformamide (30 mL). The reaction mixture was warmed to 50-55° C for 8 hr and cooled to room temperature. Water was added (300 mL), and the mixture was stirred for 30 min. Saturated aqueous sodium chloride (50 mL) was added to the mixture, the mixture was stirred for 1 hr and the crude product was collected by filtration and dried in vacuo. Purification by flash chromatography, eluting with ethyl acetate, followed by chloroform, 98:2 chloroform:methanol, then 95:5 chloroform:methanol gave 420 mg (29% yield) of the title compound; [a]D 2 5 19.70 MeOH).
14b When the single isomers 3 and 4 are desired, they can be produced by two different modifications of Scheme 1, shown in Schemes 2 and 3: o *1 14c Scheme 2 0 CH 3 CH3O-I NNN
NY
31 N Nci cl NIAb o CT-OTo 0IH 4 ci KOH-/DMF l0 H Q~C~ O Scheme 3
CH
3 O~ NNQ
H
S31 0
H
CH
3 O_ r\ N_ ON
OCH
CI
N J 0 ~36 0
CH,
NN c N N O Ci KOH-/DMF K'j ~>CH2-0-o N0N\, I
H
4 2,4-Dihydro-4-[4-[ 4 4 -methoxyphenyl)-piperazinyl]phenyl]-3H 1, 2 4 -triazol-3.one (26) is reacted with the dioxothiolane 25 as described for Scheme 1, but the sulfate ester of the resulting methoxy-sulfate salt is hydrolyzed without cleaving the methyl from the phenol by heating with 48% HBr at 45-50 0 C to produce alcohol 31. As shown in Scheme 2, alcohol 31 may be treated with diisopropylazodicarboxylate, triphenylphosphine and benzoic acid according to the method of Mitsunobu [Svnthesis 1981 1-27], hydrolyzed with potassium hydroxide in methanol, and cleaved with 48% HBr at reflux to provide 33, the single R,R enantiomer analogous to 28. When the S,S Ooo.. aenantiomer is desired, one begins with the opposite enantiomer of 25. Alternatively, as 10 shown in Scheme 3, the alcohol 31 may be treated with methanesulfonyl chloride in the presence of dimethylaminopyridine and diisopropylethylamine and the resulting mesylate ester of 34 inverted by treatment with cesium propionate according to the method of Senanayake et al. [Tet. Lett. 34, 2425 (1993)] to provide the propionate ester 36. The methoxy-ester 36 may be cleaved as before with potassium hydroxide and then HBr to 15 give 33.
Microbiological and pharmacologic studies can be used to determine the relative potency and the profile of specificity of the optically pure enantiomers, and the racemic mixture of itraconazole as antimycotic agents with a broad spectrum of activity against many fungi, yeast, and dermatophytes.
With respect to antimicrobial activity of the aforementioned compounds, selected experiments are illustrated to profile useful antimicrobial activity, and not to limit this invention in any way, including the scope of susceptible microorganisms. Antifungal conazoles may be evaluated in vitro at several concentrations (in pg/mL) against a number of fungi and bacteria. [See, Van Cutsem Chemotherapy 38 Suppl 1,3-11 (1992) and Van Cutsem et al. Rev. nfec. D.Su 1, S15-S32 (1987)]. The fungistatic assay is carried out in Sabouraud's liquid (1 g of neopeptone Difco and 2 g of glucose Difco per 100 mL of distilled water) in 16 X 160 mm test tubes, each containing mL of liquid medium which has been autoclaved at 1200 for 5 min. The compounds to be tested are dissolved in 50% alcohol at initial concentration of 20 mg/mL. The solutions are subsequently diluted with sterile distilled water to give a concentration of mg/mL. Successive decimal dilutions are made in distilled water. To tubes containing 4.5 mL of Sabouraud's liquid medium 0.5 mL of the solution of the drug is added, thereby obtaining concentrations of 1000, 500, 100, 10, and 1 Ig/mL of medium.
Control tubes are prepared by adding 0.5 mL of distilled water to 4.5 ofmL medium, alcohol being added to give concentrations identical with the tubes containing 1000 and 500 pg of the drug. The filamentous fungi are incubated in Sabouraud's agar at 25 for 2-3 weeks. A block of 2 X 2 X 2 mm is then inoculated into the medium. All cultures are made in duplicate and are incubated at 25 for 14 days. Itraconazole antifungal activity is enhanced in vitro in Sabouraud broth containing 10% inactivated bovine serum, and depends on the test medium used. Complete or marked inhibition of growth in Sabouraud broth after 14 days of incubation may be observed with Microsporum canis, Trichophyton mentagrophytes, Candida albicans, Sporothrix schenckii, Paracoccidioides brasiliensis, Blastomyces dermatitides, Histoplasma spp., Aspergillus spp. and other fungi and bacteria.
The hydroxyitraconazole isomers were tested for biological activity according three different assays. Table 2 summarizes the results of Kirby-Bauer Testing against three actively growing cultures: Candida albicans, Cryptococcus neoformens and Saccromyces cerevisiae. When compared to itraconazole, the zones of inhibition indicated that several hydroxyitraconazole isomers showed significantly greater potency.
C
Table 2.
Kirby-Bauer Test (zones of inhibition in mm) Compound Candida Cryptococcus Aspergillis Sacchromyces albicans neoformens fumigatus cerevisae 1 22 28 27 22 2 16 26 24 9 25 25 30 22 25 30 22 3+4 23 34 23 23 11+12 27 32 27 5+6+7+8 17 22 22 -18- Kirby-Bauer Testing A 1 OmL tube containing 4mL of Sabourauds dextrose broth was inoculated with 1 colony of Candida albicans picked from a plate of pure culture. The strain was ordered from the American Type Culture Collection (ATCC). The organism was grown for 4 hours at 300 C while shaking at 150 RPM. While the organism was growing, samples of hydroxy-itraconazoles were solubilized to a concentration of 1Omg/mL in DMSO. Each sample was then diluted 1:10 to make Img/mL samples or 1000lig/mL. These samples were then diluted by serial 2-fold dilutions to produce samples now containing 1000, 500, 250, 125, 62.5, 31.25, 15.6 gg/mL. A 96-well microtiter dish was set up with 10 98jL of liquid growth media in each test well, along with I tL of hydroxy-itraconazole solution. At 4 hours of growth time the Candida albicans was diluted to a McFarland standard representing about 10s-106 cells/mL and 1 JL of this inoculum placed into each test well of the microtiter dish. The dish was then covered and incubated at 30° C for 16 hours.
Actively growing cultures of Candida albicans, Cryptococcus neoformens and Saccharomyces cerevisiae were prepared as described above. The cultures were diluted to a 0.5 McFarland standard and swabbed onto a 150mm Sabouraud Dextrose agar plates. Paper disks (7mm) were placed onto the agar plates using a disk dispenser. Next 10pl of 10 mg/mL solutions of each sample hydroxy-itraconazole was pipetted onto separate paper disks. The plates were then incubated at 30° C for 16 hours. Zones of inhibition were then measured in mm.
Table 3 lists the MIC data for the hydroxyitraconazole isomers when tested against six strains of Candida albicans, and one strain each of Cr. neoformens, S. cerevisae and A. fumigatis. Nearly all the compounds showed good potency against Candida strains 28815 and 44203: 0.125 pig/mL (0.17 pM) or lower. Good potency is also observed for A.fumigatis: 0.137 p)g/ml (0.19 These numbers are compared to the MICs for itraconazole (Table 4) when tested against Candida albicans strain 44203. All the hydroxyitraconazole isomers were equipotent to the mixture of 2S,4R-isomers of itraconazole, (7 and 8) and more potent than the 2R,4S-isomers (5 and Moreover, the potencies of the itraconazole isomers were related to the stereochemistiy of the dioxolane., whereas the antifungal potency did not appear related to the stereochemnistry of the dioxolane ring in the hydroxyitraconazole isomers.
Table 3.
S. S *5 *5S* .5
*SS*
S
S
Antifuingal Mu[Cs (jig/mi) Organism 1 2 9 10 3+4 11+12 C albicans strain 10231 1.0 0.5 0.5 1.0 1.0 28516 0.25 0.25 0.25 0.25 0.25 0.25 28815 0.125 0.5 0.125 0.125 0.125 0.125 44203 0.05 0.05 0.05 0.025 0.10 0.05 44373 0.5 0.25 0.25 0.25 0.25 62342 0.5 1.0 0.5 0.5 1.0 Cr. neoformens 0.39 0.39 0.78 0.78 0.78 0.78 S. cerevisae 0.195 0.39 0.195 0.195 0.78 0.195 A. fumigatis 0.137 0.137 0.137 0.137 0.137 0.137 Table 4.
MiCs Against Candida albicans Strain 44203.
Isomer MIC jig/mil 1 0.05 2 0.05 9 0.05 0.025 3+4 0.10 11+12 0.05 0.3125 6 0.3125 7 0.078 8 0.078 5+6 0.156 7+8 0.078 5+6+7+8 0.06 Table 5 gives IC 5 0 data for the inhibition of cytochrome P450 3A4. The 'C 50 values were determined by the appearance of 613-hydroxytestosterone after incubation with testosterone. All the compounds tested were significant inhibitors of testosterone 63-..
hydroxylase activity. Isomers 1, 2 and 3+4, i.e. those with 2R,4S stereochemistry in the dioxolane ring, showed notably less inhibition of the P450 3A4 enzyme than the 2S,4R isomers. Of greater clinical significance is the ratio of the IC 5 o for 3A4 to the MIC for C. albicans 44203. This value indicates the "therapeutic index" for each isomer: a compound being a weak inhibitor of 3A4 (large IC 5 0 and a potent antifungal against C.
albicans 44203 (small MIC) would give a large ICso/MIC ratio. As seen in Table isomers 1, 2 and 3+4 showed excellent results.
Table 5. ICso against Cytochrome P450 3A4 *0 a a.
60000 0@OS
S
0* 0
C.
C r
**S
SS
*5
S*
S*
S CCC 5 a Sr 0s 1 1 0 5 Compound ICso (pM) IC 5 s/Itraconazole IC 5 o/ 0 IC Ratio Ratio 1 2.8 5.3 56 2 3.5 6.6 9 0.37 0.7 7.4 10 0.48 0.9 19 3+4 2.4 3.8 24 11+12 0.34 0.6 Itraconazole 0.53 1 In vivo activity of hydroxyitraconazole and derivatives may be compared against experimental cutaneous candidosis in guinea pigs, and vaginal candidosis in rats. The in 20 vivo activity of the compounds in vaginal candidosis is evaluated by inducing vaginal infection with C. albicans in ovariectomized and hysterectomized Wistar rats (100g) which are treated weekly with 100 jtg of estradiol undecanoate in sesame oil, subcutaneously. Animals in pseudooestrus are infected intravaginally with a fixed concentration of C. albicans in saline. Control of infection or cure is estimated by taking vaginal smears at fixed days after infection. Drugs to be evaluated and compared on a mg/kg basis may be given prophylactically or therapeutically and their efficacy judged by comparing the ratio of negative animals to the total number in each drug group. In similar studies, the activity against cutaneous candidosis in guinea pigs [(Van Cutsem et al. Chemotherapy 17, 392, (1972)] provides the basis of comparison.
The compounds of the present invention allow the treatment of fungal infections..
while avoiding the adverse effects associated with itraconazole. The term "adverse effects" includes, but is not limited to, arrhythmogenicity, hepatotoxicity and elevations in serum liver enzymes, drug interactions, and hypersensitivity reactions including urticaria, nausea, vomiting, abdominal pain, headache, dizziness and the like.
The potential for promoting arrhythmia is evaluated by examining the effects of the isomers of hydroxyitraconazole on cardiac action potential and contractility in human, canine and rabbit hearts. Torsades de Pointes is a well known side effect of antiarrhythmic drugs, such as quinidine, sotalol and acetyl-procainamide, which cause a 10 prolongation of cardiac repolarization. All of these drugs have in common the ability to iblock a cellular potassium channel called the delayed rectifier and it is generally assumed that this is mechanistically linked to their ability to induce the syndrome of Torsades de Pointes. [See, Zehender et al. Cardiovascular Drugs Ther., 5 515-530 (1991).] Increases in QT duration and action potential duration in isolated guinea pig or "15 rabbit hearts can therefore be used to indicate an arrhythmogenic effect. Hearts are perfused with an oxygenated Tyrode's solution, containing 0.0, 1.0, 5.0, 10.0 or 30.0 pM of racemic itraconazole. QT duration and action potential duration (APD) are measured from cardiac electrodes.
To observe the effects in vivo, mongrel dogs of either sex weighing 5-20 kg are anesthetized and instrumented by standard techniques for blood pressure and EKG. A solid state transducer for dP/dT is placed in the left cardiac ventricle, and an epicardial electrode is put into place. The test compound is infused at progressively higher doses, beginning at 1 jg/kg/min for 15 to 30 minutes and increased incrementally until a cardiovascular collapse ensues. Parameters measured are: blood pressure, heart rate, dP/dT, and the QT-interval. Measurements of hemodynamics and electrical activity, including QTc interval, are made in response to the test compound and compared.
The potential for promoting hepatotoxicity is assessed in vitro in human hepatic microsomes, human lymphocytes and other cell culture systems. Hepatic microsomes are prepared from human liver. Tissue is thawed and then homogenized in 0.15 M KCI in a Polytron homogenizer. The homogenate is centrifuged and the pellet is resuspended and homogenized in 0.15 M KCI. Aliquots are frozen and stored at -70 C. Human lymphocytes are aseptically isolated from fresh, heparinized human blood. Blood is diluted with Eagle's minimal essential medium and layered on Ficoll-Paque. The samples are centrifuged, and lymphocytes are then removed from the aqueous-Ficoll interface and suspended in medium (15Mm HEPES, pH, The cells are then centrifuged, washed once in the HEPES medium, and resuspended.
Cytotoxicity is assessed by the conversion of 3-(4,5 dimethylthiazol-2-yl)-2,5diphenyltetrazolium bromide (MTT) to a purple formazan. The conversion of MTT to :i 10 dye is done in multiwell plates. After preparation, hepatic microsomes or lymphocytes are incubated alone or with the test compound in a concentration range from I to 400 pM at 370 C in a humidified incubator. After incubation, the microsomes/cells are washed with 5% albumin in HEPES-buffered medium and resuspended. The microsomes/cells are then incubated at 37 C in a humidified incubator. After the incubation, 125 jig of MTT is added to each well. The plates are incubated at 370 C and centrifuged. After centrifugation, 100 tL of isopropanol is added and, after incubation, the optical density is determined using an automated plate-reader.
o The isomers of cis hydroxyitraconazole were tested for water solubility against a M standard solution of hydroxyitraconazole. Table 6 lists the absorbance of eachisomer at 260 nm and its corresponding solubility. All the isomers except for 3+4 and 11+12 showed absorbances below the limit of detection (LOD) of the instrument, which is 5000 pV*sec., (2 and 10 were not even detected), allowing only order-of-magnitude accuracy. The values for 3+4 and 11+12 are barely above the LOD, giving accuracies of 10%. In every case, solubilities were in the 10' to 102 ng/ml range. Itraconazole itself has such low water solubility that it could not be determined by this method.
10 a r.
a. a a.
a Table 6.
Water solubility of Hydroxyitraconazole Isomers and Itraconazole.
Compound Area (IiV*sec) Solubility (ng/mIl) 1 1499 51 2 not detected 9 1332 not detected 3+4' 54534 1970 11+12' 16449 590 +Itraconazole- not detected 'Measured against a Hydroxyitraconazole std of 482 jig/mI, Area=13364520 mV*sec The synthesis of the phosphate and sulfate esters of cis hydroxyitraconazole isomers and their corresponding WICs against three fungal strains are shown below.
Scheme 4. Synthesis of the Sulfate Ester of 2
CI
0 9H N jH 2 Cl pyridine/S0 3 _t C 1 0 CH3 N- N~ (Att0 N H 0N N OS0 3
H
2-sulfate ester Scheme 5. Synthesis of the Phosphate Ester of 2
CI
Cl O H 3 "o N N HIN N 2 1) (BnO) 2 PN(iPr) 2 I 2) t-BuOOH 3) H 2 -Pd/C N N C I 0 N H
XNH
N *CHy-O KJ N N
Y
N
N
N 0P0 3
H
2 2-phosphate ester Table 7.
Antifungal MICs (pg/ml) Organism 2-sulfate 2-phosphate C. albicans 6.25 12.5 Cr. neoformens 3.13 12.5 S. cerevisae 12.5 12.5 The phosphate analogs of the isomers with the largest ICo/MIC ratios, 1, 2 and 3+4 were further investigated for hydrolysis in plasma and tissues. Sodium, potassium and calcium salts were prepared, but the bis-NMG salt was the most stable. The A% conversion (as determined by HPLC A% analysis) of the phosphate ester to the hydroxy compound was determined in three systems: human plasma, equine pancreas and porcine brain. The results indicate that the phosphate ester of the 2R4SSR isomer 2 [both as the free acid and as the bis(N-methylglucamine) salt] was the most readily hydrolyzed compound in all three systems, and the bis-NMG salts were more or equally rapidly hydrolyzed when compared to the free acid counterparts. Hydrolysis proceeded to >10% overnight at 37 0 C in pancreas and brain, and 3% in human plasma. The hydrolyses of the three phosphates of 1, 2 and 3+4 were also studied in various rat tissues and plasma. In all cases, the jejunum and liver gave the greatest conversion to hydroxyitraconazole, ranging from 20% to 40% for the free acids.
The water solubility of the NMG salts of the phosphate esters of isomers 1, 2 and 3+4 were investigated and compared to the corresponding hydroxyitraconazole isomers 1, 2 and 3+4. The phosphate esters were as much as 10' times more water soluble than the corresponding hydroxyitraconazole isomer.
10 Table 8.
a
C
C,
15 Water solubility of Hydroxyitraconazole Phosphate Isomers Compound Solubility (mg/mL) 1 5.1x10- 2 <1.0xl0- 3+4 2.0x10- 3 1-PO,-NMG 2 >100 2-PO,-NMG 2 >100 3+4-PO,-NMG, >100 The biological assays shown above establish that the hydroxyitraconazole isomers are of equal or greater potency than their itraconazole counterparts. All the hydroxyitraconazole isomers inhibited CYP3A4, their inhibition being a function of the absolute stereochemistry of the dioxolane moiety. In addition, the 2R,4S isomers of hydroxyitraconazole showed dramatically improved ICso/MIC ratios when compared to racemic itraconazole. The sulfate and phosphate esters of hydroxyitraconazole isomers, although highly water-soluble, showed lessened antifungal activity.
Nonetheless, the hydrolysis of the phosphate esters to hydroxyitraconazole allows them to be considered water-soluble prodrugs of useful potency.
The concentrations of hydroxyitraconazole in plasma, CSF and brain tissue were examined in the rat. Fasted male rats of the Crl:CD®(SD)IGS BR strain from Charles River Laboratories were given the test compounds by oral gavage at 20 mg/kg. The rats were fasted for an additional 4 hours after dosing and were sacrificed by cardiac puncture under halothane anesthesia at the time shown in the tables below. The blood, CSF and brain tissue were collected and the samples were analyzed by HPLC for hydroxyitraconazole concentration. The following isomers were administered: Test Group stereochemical descriptor isomer number 0' 0 0*t 0**b 0 1 1 2 2 3 1:1 mixture of 3+4
(R,S,S,S)
4 3 5 4 Because of the low solubility of the medicament in the vehicle, the compounds were administered as suspension/solutions of 10 mgof test compound per milliliter in an aqueous vehicle containing 0.5% carboxymethylcellulose and 0.1% Tween® 80. The suspensions were centrifuged and the actual amount of medicament in suspension (the insoluble portion) and in the supernatant (the soluble portion) were determined. The data from the dose concentration and solubility analyses show that the concentrations of the doses were very close to the nominal dose and that only the mixed isomer dosage had much solubility in the 0.5% CMC/O.I% Tween 80 in water solvent.
I, Table 9 Groups 1-5 Dose Concentration Test Material Spmtn Hydroxy-Actual as itraconazole Whole Whole Supernatant of enantiomers Nominal Actual of Actual Whole Group (mg/mE) Nominal (mglmL) Actual IRSSR 10 10.1 101 0.009 0.1 2 RSRS 10 1.0.4 104 0.094 0.9 3RSRR/RSSS 10 10.5 105 0.943 4 RSRR 10 10.3 103 0.014 0.1 5 RISSS 10 10.2 102 013 0.3 Table Concentrations (ng/mL) of Hydroxyitraconazole in Rat Plasma I Group 2 Group 3 Group 4 Group Treatment RS,S,R R,,S,RS RS,R,R/ RS,RR RS,S,S R,S,S,S Predose BLQ* BLQ BLQ BLQ BLQ hr 29.8 288 1220' 113 50.0 1 hr 46.6 5092 14501 281 331 2 hr 74.3 8601 5060' 1140' 10001 4 hr 55.7 1090' 68001 2260' 1880' 6 hr 272 468 2 10800' 1380' 1910' 8 hr 101 346 8220' 941' 1320' 12 hr 32.2 26.7 3020' 750' 579' 24 hr _1.67 2.85 66.5 49.9 4.44 *BLQ Below the Limit of Quantification Original value exceeded calibration curve; reassaved at partial volume.
2 Estimated value reported; exceeded calibration curve.
-28- Table IlI Concentrations (ng/mL) of Hydroxyitraconazole in Rat CSF
C..
C
Table 12 Concentrations (nglg) Of HYdroxyitraconazole in Rat Brain In plasma, the data in Table 2 show that the concentration of hydroxyitraconazole. .was highest in rats treated with the 1:1 mixture of isomers 3 4 (Group with concentrations reaching 10,800 ng/mL at 6 hours postdose. Rats treated with the (R,SR,R) single isomer 3 or single isomer 4 hydroxyitraconazole (Groups 4 and 5, respectively) had Cma values approximately five times lower than rats given the 1:1 mixture of isomers 3 4.
Rats given the hydroxyitraconazole isomer 2 (Group 2) had Cm. values approximately ten times lower than rats given the 1:1 mixture of isomers 3 4, while rats given the isomer 1 hydroxyitraconazole had C, values approximately forty times lower than rats given the 1:1 mixture ofisomers 3 4. The for all five treatment 10 groups was either 4 or 6 hours postdose.
In cerebrospinal fluid (Table the concentrations of hydroxyitraconazole found were near or below the limit of detection in almost all the samples; the highest concentration and widest appearance of hydroxyitraconazole over time were seen in samples from Group 3 (the 1:1 mixture of isomers 3 4) between two and twelve hours postdose.
15 In brain tissue (Table the concentrations of hydroxyitraconazole were near or below the limit of detection in almost all the samples in all treatment groups except for Group 3 (the 1:1 mixture of isomers 3 4) between two and twelve hours postdose, matching the CSF samples that had quantifiable levels of hydroxvitraconazole.
The results of this study demonstrate that the administration of a mixture of isomers results in unexpectedly higher levels of the medicament in plasma, CSF and brain tissue than those seen with single isomers. Since antifungal efficacy is a function of drug concentration, one may conclude from these studies that a mixture of isomers would exhibit improved efficacy, particularly in treating infections previously inaccessible to itraconazole therapy.
The magnitude of a prophylactic or therapeutic dose of hydroxyitraconazole or.
derivative in the acute or chronic management of disease will vary with the severity of the condition to be treated, and the route of administration. The dose, and perhaps the dose frequency, will also vary according to the age, body weight, and response of the individual patient. In general, the total daily dose range, for hydroxyitraconazole or a derivative, for the conditions described herein, is from about 50 mg to about 1200 mg, in single or divided doses. Preferably, a daily dose range should be between about 100 mg to about 800 mg, in single or divided doses, while most preferably, a daily dose range should be between about 200 mg and 400 mg, in divided doses. In managing the patient, the therapy should be 10 initiated at a lower dose, perhaps about 100 mg to about 200 mg, and increased up to about .400 mg or higher depending on the patient's global response. It is further recommended that children, and patients over 65 years, and those with impaired renal, or hepatic function, initially receive low doses, and that they be titrated based on individual response(s) and blood level(s). It may be necessary to use dosages outside these ranges in some cases as 15 will be apparent to those skilled in the art. Further, it is noted that the clinician or treating physician will know how and when to interrupt, adjust, or terminate therapy in conjunction with individual patient response. An amount sufficient to alleviate or prevent infections but insufficient to cause adverse effects is encompassed by the above-described dosage amounts and dose frequency schedule.
20 Any suitable route of administration may be employed for providing the patient with an effective dosage ofhydroxyitraconazole or derivative. For example, oral, rectal, parenteral (subcutaneous, intramuscular, intravenous), transdermal, topical, pulmonary and like forms of administration may be employed. Dosage forms include tablets, troches, dispersions.
suspensions, solutions, capsules, patches, ointments, creams, shampoos, aerosols for inhalation and the like.
The pharmaceutical compositions of the present invention comprise hydroxyitraconazole or derivative as the active ingredient, or a pharmaceutically acceptable salt thereof, and may also contain a pharmaceutically acceptable carrier, and optionally, other therapeutic ingredients.
The terms "pharmaceutically acceptable salts" or "a pharmaceutically acceptable salt thereof' refer to salts prepared from pharmaceutically acceptable non-toxic acids or bases including inorganic acids and bases and organic acids and bases. Since the hydroxy compound of the present invention is basic, salts may be prepared from pharmaceutically acceptable non-toxic acids including inorganic and organic acids.
Suitable pharmaceutically acceptable acid addition salts for the compound of the present invention include acetic, benzenesulfonic (besylate), benzoic, camphorsulfonic, citric, ethenesulfonic, fumaric, gluconic, glutamic, hydrobromic, hydrochloric, isethionic, lactic, maleic, malic, mandelic, methanesulfonic (mesylate), mucic, nitric, pamoic, '10 pantothenic, phosphoric, succinic, sulfuric, tartaric, p-toluenesulfonic, and the like. The phosphate, being acidic, allows for the preparation of salts of bases as well as internal salts. Suitable pharmaceutically acceptable base addition salts for the compounds of the present invention include metallic salts made from aluminum, calcium, lithium, magnesium, potassium, sodium and zinc or organic salts made from lysine, N,N'- 15 dibenzylethylenediamine, chloroprocaine, choline, diethanolamine, ethylenediamine, meglumine (N-methylglucamine) and procaine. N-methyl-glucamine salts are particularly preferred, inasmuch as they exhibit superior long term stability in parenteral and oral formulations.
The compositions of the present invention include compositions such as suspensions, 20 solutions, elixirs, aerosols, and solid dosage forms. Carriers such as starches, sugars, microcrystalline cellulose, diluents, granulating agents, lubricants, binders, disintegrating agents, and the like, are commonly used in the case of oral solid preparations (such as powders, capsules, and tablets), with the oral solid preparations being preferred over the oral liquid preparations. The most preferred oral solid preparation is tablets.
Because of their ease of administration, tablets and capsules represent the most advantageous oral dosage unit form, in which case solid pharmaceutical carriers are employed. If desired, tablets may be coated by standard aqueous or nonaqueous techniques.
A second preferred route of administration is topically, for which creams, ointments, shampoos, and the like are well suited.
In addition to the common dosage forms set out above, the compounds of the present invention may also be administered by controlled release means and/or delivery devices and, because of their solubility, may also be employed in parenteral solutions, such as for intravenous administration. Because they reduce peak plasma concentrations, controlled release dosage forms are particularly useful for providing a ~therapeutic plasma concentration of 2 R,4S-itraconazole while avoiding the side effects I associated with peak plasma concentrations.
10 Pharmaceutical compositions of the present invention suitable for oral administration may be presented as discrete units such as capsules, cachets, or tablets, or aerosol sprays, each containing a predetermined amount of the active ingredient, as a powder or granules, or as a solution or a suspension in an aqueous liquid, a non-aqueous liquid, an Sgo* oil-in-water emulsion, or a water-in-oil liquid emulsion. Such compositions may be 15 prepared by any of the methods of pharmacy, but all methods include the step of ~bringing into association the active ingredient with the carrier which constitutes one or more necessary ingredients. In general, the compositions are prepared by uniformly and intimately admixing the active ingredient with liquid carriers or finely divided solid carriers or both, and then, if necessary, shaping the product into the desired presentation.
For example, a tablet may be prepared by compression or molding, optionally, with one or more accessory ingredients. Compressed tablets may be prepared by compressing in a suitable machine the active ingredient in a free-flowing form such as powder or granules, optionally mixed with a binder, lubricant, inert diluent, surface active or dispersing agent. Molded tablets may be made by molding in a suitable machine, a mixture of the powdered compound moistened with an inert liquid diluent.
Desirably, each tablet contains from about 100 mg to about 300 mg of the active ingredient. Most preferably, the tablet, cachet or capsule contains either one of three dosages, about 50 mg, about 100 mg, or about 200 mg of the active ingredient.
Similarly, sustained or controlled release formulation is well known in the art.
Chapter 94 of a standard pharmacy school text, Remington: The Science and Practice of Pharmacy, 19th edition, entitled "Sustained-Release Drug Delivery Systems," describes the more common types of oral and parenteral controlled-release dosage forms (pages 1660-1675.) Controlled release means and delivery devices are also described in U.S.
Patent Numbers 3,845,770; 3,916,899; 3,536,809; 3,598,123; and 4,008,719, and in S PCT application WO 92/20377. The relevant portions of the foregoing textbook and patent documents are incorporated herein by reference.
For topical application, there are employed as non-sprayable forms, viscous to semisolid or solid forms comprising a carrier compatible with topical application and having a dynamic viscosity preferably greater than water. Suitable formulations include but are not limited to solutions, suspensions, emulsions, creams, ointments, powders, liniments, salves, aerosols, etc., which are, if desired, sterilized or mixed with auxiliary agents, e.g., 15 preservatives, stabilizers, wetting agents, buffers or salts for influencing osmotic pressure, etc. For topical application, also suitable are sprayable aerosol preparations wherein the active ingredient, preferably in combination with a solid or liquid inert carrier material, is packaged in a squeeze bottle or in admixture with a pressurized volatile, normally gaseous propellant, a freon.
Formulations for parenteral administration include aqueous and non-aqueous sterile injection solutions which may contain anti-oxidants, buffers, bacteriostats and solutes which render the formulation isotonic with the blood of the intended recipient.
Formulations for parenteral administration also include aqueous and non-aqueous sterile suspensions, which may include suspending agents and thickening agents. The formulations may be presented in unit-dose of multi-dose containers, for example sealed ampules and vials, and may be stored in a freeze-dried (lyophilized) condition requiring only the addition of a sterile liquid carrier, for example saline, phosphate-buffered saline (PBS) or the like, immediately prior to use. Extemporaneous injection solutions and .t suspensions may be prepared from sterile powders, granules and tablets of the kind previously described.
The invention is further defined by reference to the following examples describing in detail the preparation of the compositions of the present invention as well as their utility.
It will be apparent to those skilled in the art that many modifications, both to materials and methods may be practiced without departing from the purpose and interest of this invention.
Example 1 Oral Formulation Capsules Quantity per capsule in mg Formula A B C Hydroxyitraconazole 50 100 200 Lactose 380 330 230 Cornstarch 65 65 Magnesium Stearate 5 5 Compression Weight 500 500 500 The active ingredient, hydroxyitraconazole or derivative, is sieved and blended with the excipients. The mixture is filled into suitably sized two-piece hard gelatin capsules using suitable machinery. Other doses may be prepared by altering the fill weight and if necessary, changing the capsule size to suit.
Example 2 Oral Formulation Tablets Quantity per tablet in mg Formula A B C Hydroxyitraconazole 50 100 200 Lactose 109 309 209 Cornstarch 30 30 Water (per thousand 300 mL 300 300 tabs)* mL mL Cornstarch 60 60 Magnesium Stearate 1 1 1 Compression Weight 250 500 500 '9 *The water evaporates during manufacture The active ingredient is blended with the lactose until a uniform blend is formed.
The smaller quantity of cornstarch is blended with the water to form the resulting cornstarch paste. This is then mixed with the uniform blend until a uniform wet mass is 15 formed and the remaining cornstarch is added and mixed until uniform granules are obtained. The granules are screened through a suitable milling machine using a 1/4" stainless steel screen. The milled granules are dried in a suitable drying oven and milled through a suitable milling machine again. The magnesium stearate is then blended and the resulting mixture is compressed into tablets of desired shape, thickness, hardness and disintegration.
Example 3 Aqueous Suspension for Injection A suspending vehicle is prepared from the following materials: Polyethylene glycol 4000 30 gm.
Potassium chloride 11.2 gm.
Polysorbate 80 2 gm.
Methylparaben 0.2 gn.
Water for injection q.s. 1000 mL.
The parabens are added to a major portion of the water and are dissolved therein by stirring and heating to 650 C. The resulting solution is cooled to room temperature and the remainder of the ingredients are added and dissolved. The balance of the water to make up the required volume is then added and the solution sterilized by filtration. The sterile vehicle thus prepared is then mixed with 3 gm of 2R,4S-itraconazole or a phosphate salt thereof, which has been previously reduced to a particle size less than about 10 microns and sterilized with ethylene oxide gas. The mixture is passed through a sterilized colloid mill and filled under aseptic conditions into sterile containers which are then sealed.
0 Example 4 Oral formulation Controlled Release Composition per tablet: (+)-hydroxyitraconazole 50 mg lactose 100 mg dibasic calcium phosphate 100 mg hydroxypropylmethylcellulose 120 mg polyethylene oxide 100,000 MW 20 mg polyethylene oxide 200,000 MW 20 mg magnesium stearate 4 mg Total 414mg EXAMPLE 4 All of the ingredients, except the magnesium stearate, are blended for 10 minutes.
The mixture is screened through a 30-mesh (500 pM) screen and reblended for a further minutes. The magnesium stearate is screened through a 30-mesh (500 pM) screen, added to the mixture and blended for five minutes. The resultant blend is made up into tablets, each weighing 414 mg, on a rotary tableting machine. Tablets of other strengths may be prepared by altering the ratio of active ingredient to the excipients or to the final weight of the tablet.
*0 •9

Claims (14)

  1. 2. A 1: 1: 1:1 mixture according to claim 1. 39
  2. 3. The 1:1 mixture of isomers 1 and 2 according to claim 1.
  3. 4. The mixture of any two isomers according to claim 2. The mixture according to any one of claims 1 to 4, wherein R is hydrogen.
  4. 6. The mixture according to any one of claims 1 to 5, wherein R is phosphate.
  5. 7. The mixture according to claim 6 in the form of an N-methylglucamine salt
  6. 8. A 1:1 mixture of isomers of formulae: CI Sc_ 0 CH3 0 and Cl CH 3 CH 2 -O N Q N I OH NI 6 H NH or a salt thereof.
  7. 9. A pharmaceutical composition comprising a mixture according to any one of claims 1 to 9 and a pharmaceutically acceptable carrier, excipient, diluent and/or adjuvant. 20 10. A pharmaceutical composition according to claim 9 wherein said pharmaceutically acceptable carrier is a carrier for parenteral administration.
  8. 11. A controlled release pharmaceutical composition according to claim 9 wherein said pharmaceutically acceptable carrier comprises a carrier for controlled release.
  9. 12. A pharmaceutical composition, substantially as hereinbefore described with reference to Examples 1, 2 or 4.
  10. 13. A method of treating a fungal, yeast or dermatophyte infection comprising administering to a mammal suffering from said infection a therapeutically effective amount of the mixture of any one of claims 1 to 8 or the composition of any one of claims 9 to 12.
  11. 14. The method according to claim 13, wherein said infection is central candidiasis.
  12. 15. The method according to claim 13 or claim 14, wherein said mixture or composition is administered orally.
  13. 16. The method according to claim 13 or claim 14, wherein said mixture or composition is administered parenterally.
  14. 17. The method according to claim 16, wherein said administration is intravenous. DATED THIS FIRST DAY OF FEBRUARY 2002 SEPRACOR, INC. BY PIZZEYS PATENT AND TRADE MARK ATTORNEYS 0 S see* 00055555 0 Gee
AU14775/02A 1996-11-12 2002-02-01 Hydroxyitraconazole enantiomer mixtures Ceased AU773405B2 (en)

Applications Claiming Priority (7)

Application Number Priority Date Filing Date Title
US60029998 1996-11-12
US60029990 1996-11-12
US60029992 1996-11-12
US60031073 1996-11-12
US60/029740 1996-11-12
US09301627 1999-04-29
AU48027/00A AU4802700A (en) 1999-04-29 2000-04-26 2(r),4(s)-hydroxyitraconazole isomers

Related Parent Applications (6)

Application Number Title Priority Date Filing Date
AU51720/98A Division AU715999B2 (en) 1996-11-12 1997-11-11 Use of cis-hydroxyitraconazole in order to avoid side-effects of itraconazole and hydroxyintraconazole
AU54326/98A Division AU5432698A (en) 1996-11-12 1997-11-12 2r,4s,s,s- and 2s,4r,s,s-hydroxyitraconazole
AU52512/98A Division AU5251298A (en) 1996-11-12 1997-11-12 (2r,4s,s,r-) and (2s,4r,s,r-)hydroxyitraconazole
AU51789/98A Division AU5178998A (en) 1996-11-12 1997-11-12 2(r),4(s),(r),(s)- and 2(s),4(r),(r),(s)-hydroxyitraconazole- and hydroxysaperconazole derivatives
AU52513/98A Division AU5251398A (en) 1996-11-12 1997-11-12 (2r,4s,r,r-) and (2s,4r,r,r-)hydroxyitraconazole
AU48027/00A Division AU4802700A (en) 1996-11-12 2000-04-26 2(r),4(s)-hydroxyitraconazole isomers

Publications (2)

Publication Number Publication Date
AU1477502A AU1477502A (en) 2002-03-21
AU773405B2 true AU773405B2 (en) 2004-05-27

Family

ID=32398692

Family Applications (1)

Application Number Title Priority Date Filing Date
AU14775/02A Ceased AU773405B2 (en) 1996-11-12 2002-02-01 Hydroxyitraconazole enantiomer mixtures

Country Status (1)

Country Link
AU (1) AU773405B2 (en)

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0228125A1 (en) * 1985-12-23 1987-07-08 Janssen Pharmaceutica N.V. Novel derivatives of [[4-[4-(4-phenyl-1-piperazinyl)phenoxymethyl]-1,3-dioxolan-2-yl]methyl]-1H-imidazoles and 1H-1,2,4-triazoles

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0228125A1 (en) * 1985-12-23 1987-07-08 Janssen Pharmaceutica N.V. Novel derivatives of [[4-[4-(4-phenyl-1-piperazinyl)phenoxymethyl]-1,3-dioxolan-2-yl]methyl]-1H-imidazoles and 1H-1,2,4-triazoles

Also Published As

Publication number Publication date
AU1477502A (en) 2002-03-21

Similar Documents

Publication Publication Date Title
US5474997A (en) Methods and compositions of (2R,4S) itraconazole for treating fungal yeast and dermatophyte infections
US5952502A (en) 2R,4S,R, S- and 2S,4R,R,S-hydroxyitraconazole
EP0610377B1 (en) Tri-substituted tetrahydrofuran antifungals
US6040307A (en) Methods ad compositions of (-) ketoconazole for treating fungal yeast and dermatophyte infections
US6147077A (en) 2R,4S-hydroxyitraconazole isomers
AU773405B2 (en) Hydroxyitraconazole enantiomer mixtures
US6455530B1 (en) 2R,4S,S,S- and 2S,4R,S,S-hydroxyitraconazole
WO1994016701A1 (en) Methods and compositions of (2s,4r) itraconazole for treating fungal yeast and dermatophyte infections
CA2269622A1 (en) 2r,4s,r,r- and 2s,4r,r,r-hydroxyitraconazole
CA2269616A1 (en) 2r,4s,s,r- and 2s,4r,s,r-hydroxyitraconazole
AU742242B2 (en) Method and composition employing (2R,4S) itraconazole
Dadashpour et al. Chemical and biological aspects of posaconazole as a classic antifungal agent with non-classical properties: highlighting a tetrahydrofuran-based drug toward generation of new drugs
WO1994014446A1 (en) Methods and compositions of (+) ketoconazole for treating fungal, yeast and dermatophyte infections
EP0942725A1 (en) Use of cis-hydroxyitraconazole in order to avoid side-effects of itraconazole and hydroxyintraconazole

Legal Events

Date Code Title Description
FGA Letters patent sealed or granted (standard patent)