Novel Glucocorticoid Antedrugs Possessing a 17 ... - ACS Publications

Fluocinolone acetonide (8a) was the most convenient commercially available starting material for the syn- thesis of our target acetonide, butylidene, ...
0 downloads 0 Views 138KB Size
602

J. Med. Chem. 2001, 44, 602-612

Novel Glucocorticoid Antedrugs Possessing a 17β-(γ-Lactone) Ring Panayiotis A. Procopiou,* Keith Biggadike, Alan F. English, Rosanne M. Farrell, Graham N. Hagger, Ashley P. Hancock, Michael V. Haase, Wendy R. Irving, Meenu Sareen, Michael A. Snowden, Yemisi E. Solanke, Cathy J. Tralau-Stewart, Sarah E. Walton, and Jenny A. Wood Glaxo Wellcome Research and Development, Medicines Research Centre, Gunnels Wood Road, Stevenage, Hertfordshire SG1 2NY, U.K. Received July 25, 2000

The chemical synthesis and structure-activity relationships of a novel series of 17βglucocorticoid butyrolactones possessing either a 16R,17R-isopropylidene or -butylidene group are described. The sulfur-linked γ-lactone group was incorporated onto the 17β-position of the androstane nucleus via Barton ester decarboxylation and trapping the generated 17-radical with butyrolactone disulfides. The glucocorticoid butyrolactones were hydrolyzed in human plasma by the enzyme paraoxonase to the respective hydroxy acids, which were very weak glucocorticoid agonists. The rate of hydrolysis in plasma was very rapid (t1/2 ) 4-5 min) in the case of lactones possessing a sulfur atom in the R-position of the butyrolactone group, whereas carbon-linked lactones were stable in plasma. 16R,17R-Butylidenes were more potent glucocorticoid agonists than the corresponding isopropylidene derivatives. Similarly, 1,4-dien-3-ones were more potent than the corresponding 4-en-3-ones. The butyrolactones linked to the steroidal nucleus via the β-position were more potent glucocorticoid agonists than those linked through the R-position of the lactone. The most potent compounds were also shown to be stable in human lung S9 fraction, showed much lower systemic effects than budesonide in the thymus involution test, and possessed topical antiinflammatory activity in the rat ear edema model. Introduction Topical glucocorticoids are extensively used for the treatment of a variety of inflammatory diseases such as rhinitis, inflammatory bowel disease, asthma, and several dermatological diseases. Asthma is recognized as a chronic inflammatory disease of the airways which is characterized by an eosinophil- and lymphocyte-rich accumulation of activated inflammatory cells in the airway wall. Over 100 million people worldwide suffer from asthma, and the most effective antiinflammatory treatment of the disease is provided by glucocorticoids.1 Oral glucocorticoids such as dexamethasone (1) and prednisolone (2) are still used in patients with severe

asthma; however, the high doses required mean that these agents are associated with significant adverse systemic effects such as growth retardation, osteoporosis, and suppression of the hypothalamic-pituitaryadrenal function and of the immune system. Inhaled glucocorticoid therapy was introduced in 1972 with beclomethasone dipropionate (3), and this provided asthma control with much lower doses and dramatically reduced systemic effects. A large proportion of the inhaled dose (ca. 80%) is, however, swallowed and * To whom correspondence should be addressed. Tel: +44-(0)-1438 763637. Fax: +44-(0)-1438 763438. E-mail: pap1746@ glaxowellcome.co.uk.

therefore available for absorption into the systemic circulation from the gastrointestinal tract. Glucocorticoids such as 3 and budesonide (4) retain significant oral bioavailability (>10%), and together with drug absorbed via the lung still have the potential to cause systemic side effects at therapeutic doses. Fluticasone propionate (5) (launched in 1993) is very efficiently inactivated in the liver and exhibits low oral bioavailability leading to a further reduction in systemic exposure.2

The incidence of asthma in developed countries is increasing and is responsible for an increasing proportion of healthcare costs. Inhaled glucocorticoids are currently used for treatment of all types of asthma including mild forms of the disease, and for this reason the search for new and even safer glucocorticoids is continuing. The terms ‘antedrug’3 and ‘soft drug’4 were introduced to describe drugs designed to act topically at the site of application but that are transformed into inactive metabolites upon entry into the systemic circulation. A

10.1021/jm001035c CCC: $20.00 © 2001 American Chemical Society Published on Web 01/03/2001

Glucocorticoid Antedrugs with a Lactone Ring

number of publications have appeared that describe the synthesis of antiinflammatory glucocorticoid derivatives based on the antedrug concept.5-12 These have generally involved the introduction of a carboxylic ester functionality in the expectation that hydrolysis by blood esterases will give inactive steroid carboxylic acid metabolites. Recently our group has discovered that the incorporation of a γ-lactone moiety provides derivatives which are extremely rapidly inactivated in plasma but which show remarkable stability in human lung S9 fraction. Thus, the potent antiinflammatory glucocorticoid 6 is hydrolyzed in human plasma (t1/2 < 2 min) to the inactive hydroxycarboxylic acid 7 but is essentially stable in human lung S9 (t1/2 > 2 h).13 These

ideal properties make such lactone derivatives excellent lung-selective antedrug candidates for use in asthma. The enzyme responsible for the plasma hydrolysis of these γ-lactones was identified by our group as human serum paraoxonase (EC 3.1.8.1), an organophosphatedetoxifying enzyme whose natural substrate and function were unknown but which is thought to have a role in the metabolism of lipids and lipoproteins.14 As far as we are aware, paraoxonase-mediated hydrolysis of γ-lactones has not been reported prior to our disclosure. However, hydrolysis of simple γ-lactones by a mammalian enzyme described as lactonase has been reported by Fishbein.15 We believe that Fishbein’s lactonase and paraoxonase are probably the same enzyme. Very recently the enzyme human serum homocysteine thiolactone hydrolase, a detoxifying enzyme responsible for the hydrolysis of homocysteine thiolactone, has been isolated and again identified as paraoxonase.16 As part of our medicinal chemistry program we have investigated the structural requirements for paraoxonase-mediated hydrolysis of a variety of glucocorticoid lactone derivatives and have recently reported on some C16,17-fused γ-lactones.17 In this paper we report a series of C17-linked γ-lactones as analogues of the highly potent antiinflammatory 17β-thioalkylandrostane derivatives reported recently by the Rhoˆne-Poulenc Rorer group.18 Chemistry Fluocinolone acetonide (8a) was the most convenient commercially available starting material for the synthesis of our target acetonide, butylidene, and their respective 1,2-dihydro analogues. Fluocinolone acetonide was converted to the carboxylic acid 9a by the K2CO3-catalyzed air oxidation developed by Kertesz and Marx19 (Scheme 1). The carboxylic acid 9a was then activated as the mixed anhydride 10a18 before reaction with 2-mercaptopyridine N-oxide sodium salt.20 The resulting Barton ester 11a was decarboxylated in the

Journal of Medicinal Chemistry, 2001, Vol. 44, No. 4 603

Scheme 1a

a (i) PrCHO, sand, heptane, HClO ; (ii) air, K CO , EtOH, H O; 4 2 3 2 (iii) ClPO(OEt)2, Et3N, THF; (iv) 2-mercaptopyridine N-oxide sodium salt, DMF; (v) 12, DMF, light; (vi) NaOH, H2O, THF; (vii) EtOAc, TsOH, HCl.

Scheme 2a

a

(i) Light, DMF; (ii) Raney Ni, THF, H2O.

presence of the disulfide 1221 to give a 1:1 mixture of diastereoisomers of the androstane-17β-(γ-lactone) sulfides 13a and 14a. The two diastereoisomers were separated by HPLC, and the configuration of the diastereoisomer 13a with the 3.93 ppm triplet in the 1H NMR spectrum (isomer with the shorter HPLC retention time) was shown to be the S isomer by an X-ray diffraction study. Base-catalyzed hydrolysis of isomer 13a gave an 8:1 (R:S) mixture of the hydroxy acid 15a in 70% yield indicating the facile epimerization at the lactone asymmetric center. Acid-catalyzed relactonization of this hydroxy acid mixture gave predominantly the lactone 14a (9:1 14a:13a). Decarboxylation of the Barton ester 11a in the presence of R-methylene-γ-butyrolactone (16) gave the γbutyrolactone pyridyl sulfide 17 (Scheme 2) which was desulfurized with Raney nickel to provide the C-linked γ-butyrolactones 18 as a 2:1 mixture of diastereoisomers. The individual diastereoisomers were separated by preparative HPLC.

604

Journal of Medicinal Chemistry, 2001, Vol. 44, No. 4

Scheme 3a

Procopiou et al.

Scheme 4a

a (i) I , CH Cl ; (ii) Na S , H O, EtOH; (iii) 25, DMF, light; (iv) 2 2 2 2 2 2 26, DMF, light. a (i) H , (PPh ) RhCl, EtOH; (ii) ClPO(OEt) , Et N, THF; (iii) 2 3 3 2 3 2-mercaptopyridine N-oxide sodium salt, DMF; (iv) 12, DMF, light; (v) NaOH, H2O, THF; (vi) EtOAc, HCl.

The 16,17-butylidene derivatives were prepared in a fashion similar to the acetonides (Scheme 1). Thus fluocinolone acetonide 8a was converted to the (R)butylidene 8b by the Astra method22 and then oxidized to the carboxylic acid 9b.23 Activation of 9b to 10b and reaction with mercaptopyridine N-oxide as before gave Barton ester 11b which was then decarboxylated in the presence of disulfide 12 to give the γ-lactone sulfides 13b and 14b. The two diastereoisomers were separated by preparative HPLC, and the configuration of the isomer with the shorter retention time (13b, more polar) was assigned the S configuration at the lactone asymmetric center (vide infra). Selective hydrogenation of the carboxylic acids 9a,b with Wilkinson’s catalyst gave the corresponding 1,2dihydro derivatives 19a,b (Scheme 3). Conversion of the acids 19a,b to the corresponding mixed anhydrides 20a,b, followed by Barton ester formation gave 21a,b. Decarboxylation of 21a,b in the presence of 12 gave 1:1 diastereoisomeric mixtures of γ-lactone sulfides 22a,b and 23a,b which were separated by preparative HPLC. The structure of 22a with the 3.95 ppm triplet in the 1H NMR spectrum (isomer with the shorter HPLC retention time) was shown to be the S isomer by an X-ray diffraction study. The butylidenes were generally less crystalline than isopropylidenes, and crystals suitable for X-ray diffraction study to confirm the assignments of configuration could not be obtained. The butylidene 22b was assigned the S configuration at the lactone asymmetric center (isomer with shorter HPLC

retention time) by analogy with the corresponding acetonides. Base-catalyzed hydrolysis of lactone 22b gave predominantly the (R)-hydroxy acid 24b. Lactonization of the hydroxy acid mixture gave mainly the (R)lactone 23b confirming again that an epimerization occurs during hydrolysis. Additional confirmation of the configuration of diastereoisomers 22b and 23b was obtained from the chemical shift of the 16β-H of 24b (minor isomer, giving 22b on relactonization) which appeared 0.23 ppm further downfield than the major isomer (giving 23b on relactonization). The analogous minor isomer of isopropylidene 15a (S isomer) had a chemical shift 0.19 ppm further downfield than the corresponding major isomer (R isomer). Barton ester 21b was decarboxylated in the presence of disulfides 25 and 26 to give the steroidal sulfide lactones 27 and 28, respectively, as mixtures of diastereoisomers (Scheme 4). The diastereoisomers of 27 were separated by preparative HPLC, the diastereoisomers of 28, however, were separated only after extensive chromatography by preparative TLC in order to remove the 2-pyridyl sulfide impurity 29. Disulfide 25 was prepared from 3-mercapto-γ-butyrolactone (30)24 by oxidation with iodine in CH2Cl2, whereas disulfide 26 was prepared by Michael addition of disodium disulfide to 16. The homologous acetonide lactones 31 and 32 were prepared by the synthetic route shown in Scheme 5. Fluocinolone acetonide (8a) was selectively reduced to the diol 33 with NaBH4 in THF in 99% yield. The use of THF was critical for the selective reduction of the C20-carbonyl; reduction in EtOH was nonselective, reducing the C3-carbonyl as well. The diol 33 was

Glucocorticoid Antedrugs with a Lactone Ring

Journal of Medicinal Chemistry, 2001, Vol. 44, No. 4 605

Table 1. Stability of Compounds in Krebs Buffer and Human Plasma and Relative Potency in Vitro

entry

compd no.

stability in Krebs buffer (% compd remaining) after 60 min

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20

13a 14a 13b 14b 18 isomer A 18 isomer B 22a 23a 22b 23b 27 isomer A 27 isomer B 28 31 32 38 isomer A 38 isomer B 41 15a 24b

74 90 77 100 100 100 89 92 82 91 99 100 100 96 89 97 98 NT NT NT

stability in human plasma (% compd remaining) after 10 min after 60 min

Scheme 5a

24 45 0 19 100 100 20 28 0 19 28 56 100 0 24 53 17 NT NT NT

0 15 0 7 100 99 0 0 0 0 19 0 100 0 0 13 8 NT NT NT

cleaved to the aldehyde 34 with NaIO4 and then reduced to the alcohol 35 with NaBH4 in EtOH (45% for the last two steps). Reaction of alcohol 35 with trifluoromethanesulfonic acid anhydride gave the triflate 36 (93%) which was then displaced with 2-mercapto-γ-butyrolactone (37)25 or 3-mercapto-γ-butyrolactone (30) to give 31 and 32, respectively, as mixtures of diastereoisomers. The lactone 38 (Scheme 6) was obtained by alkylation of the thioketone 3926 with 2-bromo-γ-butyrolactone (40) using K2CO3 in DMF, and the isomers were separated by chromatography followed by preparative HPLC. Finally the spirocyclic analogue 41 was prepared from ketone 4227 and thioglycolic acid (43) using trimethyl-

13 2 5 1 3 19 23 8 10 1.5 2 0.5 0.2 >73 52 >250 >250 7 1200 73

t1/2 in human plasma (min) 4 4

5 >240

Scheme 6a

a

a (i) NaBH , THF; (ii) NaIO , EtOH, H O; (iii) NaBH , EtOH; 4 4 2 4 (iv) Tf2O, pyridine, CH2Cl2; (v) 30, NaH, THF; (vi) 37, NaH, THF.

rel potency (HeLa)

(i) K2CO3, 40, DMF; (ii) HSCH2CO2H (43), TMSOTf, CH2Cl2.

silyl trifluoromethanesulfonate (TMSOTf) which acts as a Lewis acid and converts 43 to its bis-trimethylsilyl derivative in situ.28 The spirocycle 41 was obtained as a single diastereoisomer whose configuration at C17 was assumed to be as drawn based on the mechanism of the reaction, where the mercapto group attacked the 17keto group to form an ene-sulfide, followed by attack by the carboxylic acid from the least hindered R-side. The structure could not, however, be confirmed by NOE experiments. Biology Compounds were tested in vitro for their stability in human plasma and for their glucocorticoid agonist activity. The stability of the compounds shown in Table 1 was examined by HPLC in Krebs buffer after incubating for 1 h and then their stability in human plasma after incubating for 10 and 60 min at 37 °C. The results are expressed as a percent (%) of compound remaining. For a selection of compounds more accurate half-lives in human plasma were determined and are shown in Table 1. The glucocorticoid agonist activity of the compounds in Table 1 was tested by the method of Stein29 in the HeLa MMTV sPAP screen, a functional

606

Journal of Medicinal Chemistry, 2001, Vol. 44, No. 4

Table 2. Inhibition of Rat Ear Edema by 13a and 14a Compared to Fluticasone Propionate test compd

dose (µg)

mean wta ((SEM) (mg)

% decrease

p

13a 13a 13a 13a 14a 14a 14a 14a 5 5 5

1.56 6.25 25.00 100.00 1.56 6.25 25.00 100.00 0.25 1.00 4.00

56.03 ( 1.41 54.86 ( 1.35 50.65 ( 1.18 44.05 ( 1.18 58.02 ( 1.46 47.85 ( 1.09 47.21 ( 1.28 43.03 ( 1.19 45.29 ( 1.16 39.79 ( 0.99 40.57 ( 1.10

0 1.46 9.01 20.87 0 14.04 15.20 22.70 18.64 28.53 27.12

NS NS 0.007