throbber
J. Med. Chem. 2006, 49, 7887-7896
`
`7887
`
`Quinol-4-ones as Steroid A-Ring Mimetics in Nonsteroidal Dissociated Glucocorticoid Agonists
`
`John Regan,*,† Thomas W. Lee,† Rene´e M. Zindell,† Younes Bekkali,† Jo¨rg Bentzien,† Thomas Gilmore,†
`Abdelhakim Hammach,† Thomas M. Kirrane,† Alison J. Kukulka,† Daniel Kuzmich,† Richard M. Nelson,† John R. Proudfoot,†
`Mark Ralph,† Josephine Pelletier,‡ Donald Souza,‡ Lijiljana Zuvela-Jelaska,‡ Gerald Nabozny,‡ and David S. Thomson†
`
`Departments of Medicinal Chemistry and Immunology & Inflammation, Boehringer Ingelheim Pharmaceuticals, 900 Ridgebury Road,
`Ridgefield, Connecticut 06877
`
`ReceiVed October 31, 2006
`
`We report on the nuclear receptor binding affinities, cellular activities of transrepression and transactivation,
`and anti-inflammatory properties of a quinol-4-one and other A-ring mimetic containing nonsteroidal class
`of glucocorticoid agonists.
`
`Introduction
`
`During the past 50 years the successful treatment of inflam-
`matory diseases has relied on the use of glucocorticoid (GC)a
`agonists such as dexamethasone (1a) and prednisolone (1b)
`(Chart 1). While effective in controlling asthma,1 rheumatoid
`arthritis,2 and other disorders, GC therapy is fraught with a
`number of severe side effects. The seriousness of the complica-
`tions often hampers high dose and chronic administration. In
`addition, cross reactivity with other steroid hormone receptors,
`such as progesterone receptor (PR) and mineral corticoid
`receptor (MR), of the commonly prescribed GCs can provoke
`off-target pharmacology. In direct response to the current state
`of GC therapy, finding drugs with good anti-inflammatory
`properties and reduced side effects remains an ongoing quest.
`The probability of identifying a GC agonist with a better
`safety profile compared to existing therapies has substantially
`increased with newer understandings of the molecular mecha-
`nism of action.3-8 After an agonist enters a target cell and binds
`to the glucocorticoid hormone receptor (GR),
`the ligand-
`activated complex (GRC) translocates into the nucleus where
`direct and indirect functional pathways can be accessed. Acting
`directly, the GRC serves as an endogenous transcription factor
`by binding to specific DNA sequences and coactivator proteins,
`thereby initiating transcription of metabolic and endocrine genes.
`GRC-mediated transactivation of these genes is believed to
`contribute to the side effect profile of GC therapy.9 Acting
`indirectly, the GRC adopts a conformation with an affinity for
`transcription factors (e.g., NF-kB and AP-1). Subsequent binding
`to these transcription factors results in the inhibition of
`expression of pro-inflammatory cytokines such as TNF-R and
`IL-6. This process, known as transrepression, is thought to
`contribute, in part, to the anti-inflammatory component of GCs.
`
`(203) 798-
`* To whom correspondence should be addressed. Phone:
`4768. Fax: (203) 791-6072. E-mail: jregan@rdg.boehringer-ingelheim.com.
`† Department of Medicinal Chemistry.
`‡ Department of Immunology & Inflammation.
`a Abbreviations: AP-1, activator protein-1; DBF, dihydrobenzofuran;
`DELFIA, dissociation-enhanced lanthanide fluorescent immunoassay; DMEM,
`Dulbecco’s modified Eagle’s medium; D-gal, D-galactose; CHAPS, 3-[(3-
`cholamidopropyl)dimethylammonio]-1-propanesulfonate; EDTA, ethylene-
`diamimetetraacetic acid; ELISA, enzyme-linked immunosorbent assay; FP,
`fluorescence polarization; GC, glucocorticoid; GRC, glucocorticoid recep-
`tor-ligand complex; GR, glucocorticoid receptor; GR-LBD, glucocorticoid
`receptor-ligand binding domain; HFF, human foreskin fibroblast; HSP,
`heat-shock protein; IL-6, interleukin-6; LPS, lipopolysaccharide; MMTV,
`mouse mammary tumor virus; MR, mineral corticoid receptor; NF, nuclear
`factor; PR, progesterone receptor; TES, N-[tris(hydroxymethyl)methyl]-2-
`aminoethanesulfonic acid; TNF-R, tumor necrosis factor-R.
`
`Therefore, the search for GC agonists with a dissociated profile
`(greater transrepression than transactivation activity) has ac-
`celerated in recent years10,11 with an appreciation of the complex
`molecular pathways and the anticipation of an improved safety
`margin.12,13
`A primary objective in our GC agonist anti-inflammatory
`program is identifying nonsteroidal anti-inflammatory agents
`with the added benefit of exhibiting dissociation. Toward this
`end, we sought to discover pharmacophores capable of playing
`the hydrogen bond acceptor’s role of the C-3 carbonyl of
`dexamethasone that could be combined with a nonsteroidal
`scaffold. Dissociated GC agonists 2a and 2b14 represent a good
`starting point for such a venture as they contain a benzoxazinone
`group as a readily replaceable A-ring mimetic.15,16 Other
`important features include the alcohol as a steroidal C-11
`hydroxy surrogate, a trifluoromethyl substituent to maintain
`agonist activity,17 and a phenyl ring which is believed to reside
`adjacent to the area of steroid D-ring binding in the glucocor-
`ticoid receptor-ligand binding domain (GR-LBD). Expanding
`the scope of this effort to include modifications to the phenyl
`ring provides an opportunity to gauge whether changes to this
`region of the scaffold are a viable option to attenuate nuclear
`receptor potency and selectivity, as well as transrepression or
`transactivation pathways. We envision preparing the target
`molecules, represented by 5, by the opening of epoxide 3 (Chart
`1) with a nucleophilic amine embedded in a lipophilic ring
`containing a hydrogen bond acceptor functionality18 (i.e., atom
`A in 4). The syntheses and biological profiles of compounds
`such as 5 are the subject of this paper.
`
`Chemistry
`
`The preparations of a quinol-4-one-containing analogue (11)
`and a piperid-4-one (14) are shown in Scheme 1 as representa-
`tive examples of 5. The key step in this sequence is the opening
`of an epoxide (i.e., 9) with a nucleophilic amine. Briefly, the
`preparation of epoxide 9 begins with the copper-mediated 1,4-
`addition of Grignard reagent 6 to 1,1,1-trifluorobut-3-en-2-one
`(7)19 to provide ketone 8. Trimethylsulfoxonium iodide and NaH
`transform 8 to epoxide 9. Quinolone 11 is obtained by reacting
`9 with 4-hydroxyquinoline (10) and sodium ethoxide in etha-
`nol.20 Conversion of the methoxy group in 11 to a hydroxy in
`12 is accomplished with BBr3. Piperidin-4-one analogue 14 is
`prepared from the reaction of 9, cis-3,5-dimethyl-4-piperidone
`hydrochloride (13)21,22 and K2CO3 in DMF.
`
`10.1021/jm061273t CCC: $33.50 © 2006 American Chemical Society
`Published on Web 12/21/2006
`
`Downloaded via UNIV OF PENNSYLVANIA on January 29, 2021 at 03:36:57 (UTC).
`
`See https://pubs.acs.org/sharingguidelines for options on how to legitimately share published articles.
`
`Liquidia's Exhibit 1023
`IPR2020-00770
`Page 1
`
`

`

`7888 Journal of Medicinal Chemistry, 2006, Vol. 49, No. 26
`
`Regan et al.
`
`Chart 1
`
`Scheme 1a
`
`a Reagents and conditions: (a) CuI (1.1 equiv)/THF, -20 (cid:176)C, 30 min, then 7 (1.1 equiv), room temperature overnight; (b) trimethylsulfoxonium iodide/
`NaH/DMSO; (c) 10/NaOEt/ethanol, heat; (d) BBr3 (10 equiv)/CH2Cl2, room temperature overnight; (e) 13 (2 equiv)/K2CO3 (5 equiv)/DMF, 100 (cid:176)C, 1.5 h.
`
`Results and Discussion
`The nuclear receptor binding potency of the target compounds
`is determined by their ability to compete for receptor binding
`with tetramethylrhodamine-labeled dexamethasone (GR, MR)
`or mifepristone (PR).23 The transrepression potential is measured
`in human foreskin fibroblast (HFF) cells. Inhibition of IL-1
`stimulated IL-6 production, and the percent efficacy is compared
`with that of dexamethasone.24,25 In an effort
`to establish
`transactivation activity, compounds are counterscreened for the
`induction of aromatase in HFF cells26 and the ability to activate
`the MMTV promoter in HeLa cells transfected with an MMTV
`luciferase contruct.27 Comparisons with dexamethasone establish
`a basis for identifying compounds with a dissociated profile.
`Table 1 shows the effects of A-ring replacements in scaffold
`5. Morpholine 15, pyridin-4-one 17, and imidazole 18 are devoid
`of activity, while piperidin-4-one 16 demonstrates weak GR
`binding affinity (IC50 ) 1400 nM). Introducing methyl groups
`on the ring is an opportunity to increase the lipophilicity and
`
`perhaps provide a water shield to the hydrogen bond network
`consisting of the A-ring mimetic acceptor and the GR-LBD
`donor. The outcome is a significant enhancement in GR binding
`affinity. For example, cis-dimethylpiperidin-4-one 14 exhibits
`a 140-fold gain in GR binding (IC50 ) 10 nM). Other methylated
`analogues include cis-dimethylmorpholine 20, dimethylpyridin-
`4-one 21, and dimethylimidazole 22 with IC50 values of 38,
`120, and 650 nM, respectively. Introducing methyl groups onto
`the A-ring replacements has no effect on PR binding affinity,
`whereas modest activity against MR is seen. In the transrepres-
`sion anti-inflammatory cellular assay, compounds 14 and 21
`inhibit IL-6 release from HFF with IC50 values of 75 and 53
`nM, respectively, and 87-89% efficacy compared to dexa-
`methasone.
`Further increasing the lipophilicity of the A-ring mimetic from
`methyl substitutions to fusing an aromatic nucleus to the
`hydrogen bond acceptor ring provides analogues shown in Table
`2. Changes in GR binding avidity with this modification vary
`
`Liquidia's Exhibit 1023
`IPR2020-00770
`Page 2
`
`

`

`Quinol-4-ones as Steroid A-Ring Mimetics
`
`Journal of Medicinal Chemistry, 2006, Vol. 49, No. 26 7889
`
`Table 1. Methyl-Substituted A-Ring Mimetics
`
`a IC50 values were determined from duplicate 11-point concentration-response curves. See ref 23. b IC50 values represent the mean of at least two independent
`determinations. See ref 25. c Not tested.
`
`Table 2. Phenyl-Fused A-Ring Mimetics
`
`a IC50 values were determined from duplicate 11-point concentration-response curves. See ref 23. b IC50 values represent the mean of at least two independent
`determinations. See ref 25. c Not tested.
`
`between 2- and 10-fold. For example, compare 14 with 24, 22
`with 26, and 21 with 11. However, of these compounds, only
`quinolone 11 potently inhibits cellular IL-6 production (IC50 )
`21 nM and 82% efficacy). These results reinforce the often
`observed lack of correlation between GR binding affinity and
`
`transrepression activity.10,28,29 A likely explanation is that the
`flexibility of the GR-LBD can accommodate these ligands but,
`in doing so, is rendered incapable of effectively interacting with
`anti-inflammatory transcription factors.30,31 Support for the
`critical role of the hydrogen bond accepting atom in the A-ring
`
`Liquidia's Exhibit 1023
`IPR2020-00770
`Page 3
`
`

`

`7890 Journal of Medicinal Chemistry, 2006, Vol. 49, No. 26
`
`Regan et al.
`
`Table 3. Phenyl Ring Modifications
`
`compd
`no.
`
`R2
`
`R4
`
`R5
`
`a
`
`GR IC50
`(nM)
`
`a
`
`PR IC50
`(nM)
`
`MR IC50
`a
`(nM)
`
`IL-6 inhib in
`b (nM)
`HFF IC50
`(efficacy, % vs
`dexamethasone)
`
`OMe H F
`OH
`H F
`H
`H H
`OH
`H H
`Me
`H H
`Me
`H F
`OH
`H Me
`Me
`H OMe
`Me
`H OH
`OH
`F
`H
`
`11
`27 (82)
`120
`475
`10
`12
`6 (92)
`580
`960
`5
`30
`78 (48)
`350
`106
`28
`31
`6 (90)
`450
`555
`6
`32
`61 (63)
`150
`240
`22
`33
`34 (66)
`120
`240
`10
`34
`5.6 (93)
`160
`160
`8
`35
`17 (86)
`91
`525
`11
`>2000
`>2000
`36
`ntc
`780
`37
`3 (88)
`880
`340
`3
`38d
`11 (86)
`98
`48
`4
`a IC50 values were determined from duplicate 11-point concentration-
`response curves. See ref 23. b IC50 values represent the mean of at least
`two independent determinations. See ref 25. c Not tested. d See Table 5 for
`the structure.
`
`replacements is highlighted with tetrahydroquinoline 25. A factor
`of 200 decrease in GR binding affinity, compared to that of
`dihydroquinolone 24, underscores the importance of achieving
`a hydrogen bond network in the GR-LBD with this series. The
`3-fold difference in GR binding affinity between benzo[1,4]-
`oxazine 23 and dihydroquinolone 24 may reflect a stronger
`hydrogen bond acceptor pharmacophore and an advantageous
`distance for 24 between the hydrogen bond donor of the protein
`and the acceptor group on the ligand. One consequence of fusing
`a phenyl ring to this scaffold is an erosion of the GR/PR ratio
`from greater than 75-fold for compounds without a fused
`aromatic nucleus (14 and 20) to less than 50-fold for those with
`a phenyl ring attached (11, 23, 24). The GR/MR binding ratio
`was nearly unchanged. Interestingly, benzimidazole 26 does not
`show a loss of nuclear receptor selectivity. Additional changes
`to the A-ring mimetic involve embedding a nitrogen atom into
`the aromatic nucleus as a goal to define polarity limits. Table
`2 describes the consequences of a quinazolinone (27) and
`naphthyridin-4-ones (28, 29) acting as A-ring replacements.
`Compared to 11, a significant diminution in GR binding is
`observed for 27 and less so with 28 and 29. Nevertheless, both
`28 and 29 retain satisfactory transrepression activity. These data
`suggest the lipophilic environment of the A-ring binding domain
`in GR can tolerate ligands with somewhat increased polarity
`whereas the PR-LBD does not readily accommodate these
`changes. Of the phenyl-fused A-ring replacements shown in
`Table 2, compounds 11, 23, 24, 26, 28, and 29 achieve the
`highest GR binding affinity, but only 11, 28, and 29 reach IC50
`values <100 nM in the transrepression assay.
`Molecular modeling suggests that, upon binding to the GR-
`LBD, the phenyl ring of 2b resides adjacent to the steroid D-ring
`domain.15,16 The contribution to GR binding and transrepression
`activity of simple groups attached to the phenyl ring in the
`quinolone-containing scaffold is summarized in Table 3.
`Conversion of the methoxy group at C-2 (11, R2 ) OMe) to a
`hydroxy (12, R2) OH) improves the transrepression activity.
`Removal of the hydroxy group and fluorine atom from 12
`(compound 30) weakens the GR binding affinity (IC50 ) 28
`nM) and decreases the agonist activity in the cellular assay (IC50
`) 78 nM and 48% efficacy). Also seen is a decline in the GR/
`PR ratio. Maintaining only a hydroxy group (31) restores the
`binding (GR IC50 ) 6 nM) and cellular activity (IC50 ) 6 nM
`
`and 90% efficacy). In addition, a good GR/PR binding ratio is
`re-established. Switching the hydroxy group at C-2 to a methyl
`(i.e., R2 ) Me, 32 and 33) maintains the GR binding affinity
`but lessens the GR/MR ratio. Reductions in IL-6 inhibition (IC50
`values of 30-60 nM) and lower efficacies are also observed
`(63-66%) for this change. Substituting an electronegative atom
`at C-5 for an electropositive atom (34) retains the GR binding
`and cellular activity with an increase in PR and MR binding
`affinities compared to those of 12. Further changing the
`electronic character at C-5 by introducing a methoxy improves
`the agonist activity compared to that of 33. However, placing
`a hydroxy group at C-5 (36) causes a significant loss of GR
`binding, implying a negative interaction of a hydrogen bond
`donor at this site. Phenyl ring analogues involving substitution
`at C-4 reveal a very narrow range of acceptable pharmacophores.
`Only fluorine at this site provides good GR binding affinity
`and cellular activity (37). Groups larger than fluorine (methyl,
`methoxy, hydroxy) generally lead to a significant loss of binding
`affinity (data not shown). Incorporating the oxygen atom at C-2
`into a dihydrobenzofuran (DBF) ring (38) significantly dimin-
`ishes the nuclear receptor selectivity. The transrepression activity
`is comparable to that of 11. The above survey of substituents
`on the phenyl ring of the quinolone-containing steroid mimetic
`generally demonstrates a range of nuclear receptor binding
`affinity smaller than a factor of 10. However, pharmacophores
`such as methoxy or hydroxy at C-2 (11, 12, 31, 34, and 37)
`and DHB (38) are required for achieving the best agonist profile.
`Another general trend (data not shown) for this series is a
`hydroxy group at C-2 displays better IL-6 inhibition and efficacy
`compared to a methoxy despite similar GR binding affinities
`(e.g., 11 vs 12). Also important are the substitution patterns at
`C-2 and C-5 (34 vs 35) and steric constraints at C-4. Taken
`together, these results highlight the important relationships of
`size and polarity required for favorable interactions adjacent to
`the steroid D-ring domain within the GR-LBD. In addition,
`similar observations are noted with phenyl ring substitutions
`for molecules containing A-ring mimetics other than quinol-4-
`one. See Table 5 for examples.
`A primary goal of this project is identifying GC agonists that
`can distinguish between transrepression and transactivation
`pathways. Two assays are used as a counterscreen to determine
`the extent of transactivation. Dexamethasone activates the
`MMTV promoter in HeLa cells transfected with an MMTV
`luciferase construct. In addition, dexamethasone induces expres-
`sion of aromatase in HFF.32 Several compounds were evaluated
`for their transactivation potential to activate MMTV and induce
`aromatase expression (EC50 and percent efficacy). A compound
`can be characterized as dissociated with either weak activity or
`low efficacy, or both, in the transactivation assays compared to
`the transrepression assay. The role that phenyl ring substitutions
`play in defining dissociation is seen in Table 4. In the MMTV
`assay, 2-hydroxyphenyl compounds 12 and 37 are partial
`agonists (EC50 ) 15-37 nM and 20-35% efficacy) while the
`other compounds are devoid of activity compared to dexa-
`methasone. In the aromatase induction assay, 12 and 37 exhibit
`pronounced activity with EC50 ) 15-17 nM and 65-74%
`efficacies. Alternatively, 2-methoxyphenyl, 2-methylphenyl, and
`DBF analogues (11, 32, and 38) display low potency or efficacy
`in the aromatase assay. Of the modifications at C-5 of the phenyl
`ring, methoxy 35 displays moderate activity against aromatase
`(EC50 ) 95 nM, 65% efficacy) while hydrogen or fluorine (32
`and 33) are weakly potent and efficacious derivatives. These
`data suggest groups on the phenyl ring have an important
`influence on the degree of dissociation and parallel the observa-
`
`Liquidia's Exhibit 1023
`IPR2020-00770
`Page 4
`
`

`

`Quinol-4-ones as Steroid A-Ring Mimetics
`
`Journal of Medicinal Chemistry, 2006, Vol. 49, No. 26 7891
`
`Table 4. Dissociation Profiles
`
`Table 5. In Vivo Activities of Dissociated Agonists
`
`compd
`no.
`
`GR IC50
`a
`(nM)
`
`IL-6 inhib in
`b (nM)
`HFF IC50
`(efficacy, % vs
`dexamethasone)
`
`MMTV activation in
`HeLa EC50
`c (nM)
`(efficacy, % vs
`dexamethasone)
`
`aromatase inhib in
`HFF EC50
`d (nM)
`(efficacy, % vs
`dexamethasone)
`
`1a
`1.9 (100)
`17 (100)
`0.5 (100)
`3.4
`1b
`19 (92)
`16 (91)
`6.6 (96)
`15
`>2000 (14)
`2a
`91 (56)
`59 (80)
`3
`>2000 (10)
`11
`220 (67)
`27 (82)
`10
`12
`14 (74)
`37 (20)
`6 (92)
`5
`14
`240 (93)
`nte
`75 (89)
`10
`>2000 (3)
`21
`170 (77)
`53 (87)
`120
`28
`ipf (41)
`nt
`56 (78)
`77
`>2000 (1)
`29
`ip (39)
`35 (72)
`34
`>2000 (29)
`>2000 (1)
`32
`61 (63)
`22
`>2000 (2)
`33
`ip (22)
`34 (66)
`10
`>2000 (6)
`35
`95 (65)
`17 (86)
`11
`37
`17 (82)
`15 (35)
`3 (88)
`3
`38g
`ip (60)
`nt
`11 (86)
`4
`39g
`ip (26)
`nt
`39 (77)
`34
`>2000 (14)
`40g
`nt
`47 (59)
`48
`>2000 (20)
`41g
`nt
`70 (71)
`125
`a IC50 values were determined from duplicate 11-point concentration-
`response curves. See ref 23. b IC50 values represent the mean of at least
`two independent determinations. See ref 25. c EC50 values represent the
`mean of at least two independent determinations. See ref 27. d EC50 values
`represent the mean of at least two independent determinations. See ref 26.
`e Not tested. f Due to the flat shape of the dose-response curve, an EC50
`value beyond 2000 nM could not be calculated. g See Table 5 for the
`structures.
`
`tion that some steroids containing modified D-rings show
`dissociated activities.33 The contribution of the A-ring replace-
`ment to the dissociation profile is seen in Table 4. Comparing
`compounds with the 2-methoxy-5-fluorophenyl ring, dimeth-
`ylpiperidin-4-one 14, dimethylpyridin-4-one 21, and naphthy-
`ridin-4-one 28 demonstrate a somewhat weaker transactivation
`response compared to quinol-4-one 12. However, the 2-methyl-
`5-fluorophenyl and DBF rings impart a much more dissociated
`profile for dimethylpyridin-4-one (39 and 41), naphthyridin-4-
`one (40), and quinol-4-one (33 and 38) A-ring mimetics. Thus,
`a distinction between potent transrepression activity and dimin-
`ished transactivation is achieved with the proper choice of A-ring
`replacements and phenyl substitution with this class of com-
`pounds.
`Selected dissociated compounds were evaluated for their anti-
`inflammatory properties in an LPS-stimulated mouse model of
`TNF-R production. Test compounds were administered orally
`in Cremophor 60 min prior to LPS challenge. TNF-R levels
`were measured 60 min later. Table 5 summarizes the GR binding
`and cellular and in vivo transrepression profiles of various
`combinations of A-ring (dimethylpyridin-4-one, quinol-4-one,
`and naphthyridin-4-one) and phenyl ring analogues. When dosed
`at 10 mg/kg, the nondissociated analogue 12 and dissociated
`compounds 33 and 38-41 inhibit TNF-R production (48-58%).
`
`Conclusion
`Several new types of pharmacophores can serve as A-ring
`mimetics with good binding affinity for GR for this scaffold.
`These include dimethylpiperidin-4-one (14), quinolone (11), cis-
`dimethylmorpholine (20), benzo[1,4]oxazine (23), dihydroqui-
`nolone (24), benzimidazole (26), and naphthyridin-4-one (29).
`In addition, good nuclear receptor selectivity is achieved. Of
`these, 11, 29, and dimethylpyridin-4-one (21) exhibit good
`transrepression activity in a cellular assay of IL-6 production.
`The substitution patterns on the phenyl ring play an important
`role in defining GR binding affinity and the degree of trans-
`repression. Compounds with a hydroxy group at C-2 obtain the
`highest efficacy levels of transrepression, while a C-5 hydroxy
`is deleterious toward activity. Methyl and methoxy substituents
`
`compd
`no.
`
`A
`
`phenyl
`ring
`
`IL-6 inhib in
`b (nM)
`HFF IC50
`(efficacy, % vs
`dexamethasone)
`
`a
`
`GR IC50
`(nM)
`
`inhib of
`TNF-R in micec
`(%) (n ) 8)
`(10 mg/kg)
`84 ( 8
`1bd
`7 (96)
`15
`see Chart 1
`55 ( 6
`12
`6 (92)
`5
`see Table 3
`36 ( 10
`32
`61 (63)
`22
`II
`II
`50 ( 17
`33
`34 (66)
`10
`I
`II
`49 ( 10
`38
`11 (86)
`4
`III
`II
`49 ( 6
`39
`39 (77)
`34
`III
`I
`58 ( 12
`40
`47 (59)
`48
`I
`III
`48 ( 11
`41
`70 (71)
`125
`I
`I
`a IC50 values were determined from duplicate 11-point concentration-
`response curves. See ref 23. b IC50 values represent the mean of at least
`two independent determinations. c See the Experimental Section. d 3 mg/
`kg, po.
`
`at C-2 and C-5 are well tolerated. The dissociated profile of
`this series in either the MMTV or aromatase assays was
`dependent upon the choice of the A-ring mimetic and the
`substitution on the phenyl ring. As A-ring replacements, fused
`rings 11, 28, and 29 exhibit a superior dissociated profile due
`to differences in either potency or efficacy in the transactivation
`assays compared to monocyclic rings 14 and 21. In general,
`methyl and methoxy groups at C-2 of the phenyl ring demon-
`strate weaker transactivation activities than hydroxy at this site.
`In an animal model of inflammation, compounds 12, 33, and
`38-41 potently inhibit TNF-R production. Thus, the nuclear
`receptor binding affinities, the cellular levels of transrepression
`and transactivation, and the anti-inflammatory properties have
`been demonstrated with the quinol-4-one-containing class of
`GR agonists. Further work in this series will be reported in due
`course.
`
`Experimental Section
`All solvents and reagents were obtained from commercial sources
`and used without further purification. Flash chromatography was
`performed according to the procedure of Still et al. (EM Science
`Kieselgel 60, 70-230 mesh). Melting points were obtained from a
`Mel-temp 3.0 or Fisher-Johns melting point apparatus and are
`uncorrected. The 1H NMR spectra were recorded on a Bruker DPX
`400 spectrometer. Chemical shifts are reported in parts per million
`((cid:228)) from the tetramethylsilane resonance in the indicated solvent.
`Infrared spectra were recorded without solvent on a Nicolet Impact
`410. Mass spectra were obtained from a Finnigan-SSQ7000
`spectrometer. Samples were generally introduced by a particle beam
`and ionized with NH4Cl. The analytical HPLC purity was estab-
`lished with the following protocols. Method A: Gilson chromato-
`graphic system, Waters SureFire reversed-phase C18 analytical
`column (4.6 (cid:2) 50 mm, 5 (cid:237)m, part number 186002557), flow rate
`5 mL/min, detection with a Gilson UV/VIS-155 at 254 nm, sample
`
`Liquidia's Exhibit 1023
`IPR2020-00770
`Page 5
`
`

`

`7892 Journal of Medicinal Chemistry, 2006, Vol. 49, No. 26
`
`Regan et al.
`
`size 25 (cid:237)L, gradient 5% acetonitrile in water with 0.1% TFA to
`95% acetonitrile in water with 0.1% TFA over 6 min and held.
`Method B: Rainin chromatography system, model SD-200, Tosoh
`Bioscience TSK Super ODS column (4.6 mm (cid:2) 10 cm, 2 (cid:237)M,
`part number G0030-90F), flow rate 1.2 mL/min, detection with
`Dynamax absorbance detector at 254 nM, gradient 5% acetonitrile
`in water with 0.05% TFA to 95% acetonitrile in water with 0.05%
`TFA over 10 min and held. Method C: Rainin chromatography
`system, model SD-200, Whatman PartiSphere C-8 column (4.6 (cid:2)
`125 mm, 5 (cid:237)m, part number 4621-0503), detection with Dynamax
`absorbance detector at 254 nM, gradient 5% acetonitrile in water
`with 0.1% TFA to 95% acetonitrile in water with 0.1% TFA.
`1,1,1-Trifluoro-4-methylpent-3-en-2-one (7). To a 0-5 (cid:176)C
`mixture of N,O-dimethylhydroxylamine hydrochloride (15.8 g) in
`CH2Cl2 (400 mL) was added dropwise trifluoroacetic anhydride
`(21.7 mL). Pyridine (37 mL) was added to the mixture dropwise.
`The resulting mixture was stirred at 0-5 (cid:176)C for 0.5 h and diluted
`with water. The organic layer was washed with water, 1 N aqueous
`HCl, water, and brine and dried (MgSO4). Removal of the volatiles
`in vacuo for 5 min provided 2,2,2-trifluoro-N-methoxy-N-methyl-
`acetamide as a colorless oil.
`To a 0-5 (cid:176)C mixture of 2,2,2-trifluoro-N-methoxy-N-methyl-
`acetamide (3 g) in 30 mL of anhydrous ether was added (2-
`methylpropenyl)magnesium bromide in THF (42 mL of a 0.5 M
`solution). The reaction was stirred at 0-5 (cid:176)C for 0.5 h, warmed to
`room temperature, and stirred overnight. The reaction was quenched
`with saturated aqueous NH4Cl and extracted with ether three times.
`The organic layers were combined, washed with water and brine,
`and dried (MgSO4). Most of the volatiles were removed at
`atmospheric pressure, providing 7 as an approximately 75% pure
`solution containing ether and THF that was used without further
`purification.
`1,1,1-Trifluoro-4-(5-fluoro-2-methoxyphenyl)-4-methylpentan-
`2-one (8). To a 0-5 (cid:176)C solution of 7 (24 g, approximately 75%
`pure solution containing ether and THF) and CuI (30.1 g) in ether
`(300 mL) was added (5-fluoro-2-methylphenyl)magnesium bromide
`(315 mL, 157 mmol, 0.5 M solution in THF). The mixture was
`warmed to room temperature, stirred overnight, and diluted with
`saturated aqueous NH4Cl (15 mL) and ethyl acetate. The organic
`layer was washed with water and brine and dried (MgSO4).
`Removal of the volatiles in vacuo provided a residue which was
`purified by flash silica gel chromatography using 0-5% ethyl
`acetate in hexanes as the eluent. The product-rich fractions were
`collected and the volatiles removed in vacuo to yield 20.3 g (46%)
`of 8 as an oil.
`2-[2-(5-Fluoro-2-methoxyphenyl)-2-methylpropyl]-2-(trifluo-
`romethyl)oxirane (9). To a suspension of trimethylsulfoxonium
`iodide (4.76 g, 21.5 mmol) in DMSO (25 mL) was added NaH
`(0.863 g of a 60% dispersion in oil, 21.5 mmol) in four portions.
`After being stirred for 0.5 h, the solution was added to 8 (5.00 g,
`17.9 mmol) in DMSO (20 mL). After 2 h the mixture was diluted
`with water (150 mL) and extracted with ether (2 (cid:2) 125 mL). The
`combined organic layers were washed with water and brine and
`dried (MgSO4). Removal of the volatiles in vacuo provided 9 as
`an oil which was used without further purification.
`1-[4-(5-Fluoro-2-methoxyphenyl)-2-hydroxy-4-methyl-2-(tri-
`fluoromethyl)pentyl]-1H-quinolin-4-one (11). A mixture of 9
`(0.226, 0.773 mmol), 4-hydroxyquinoline (10; 0.112 g, 0.773
`mmol), sodium ethoxide (0.28 mL of a 21 wt % solution in ethanol,
`0.77 mmol), and anhydrous ethanol (4 mL) was heated at 85 (cid:176)C
`for 6 h, cooled to room temperature, diluted with ethyl acetate and
`acetic acid, washed with water and brine, and dried (MgSO4).
`Removal of the volatiles in vacuo provided a residue which was
`purified with flash silica gel chromatography using ethyl acetate
`as the eluent. The product-rich fractions were collected and the
`volatiles removed in vacuo to yield 0.13 g (38%) of 11. Mp: 198-
`200 (cid:176)C.
`1H NMR (400 MHz, CDCl3): (cid:228) 8.21 (d, 1H), 7.42 (m,
`2H), 7.25 (s, 1H), 7.1 (m, 1H), 6.92 (m, 2H), 6.80 (m, 1H), 4.15
`(dd, 2H), 3.80 (s, 3H), 2.55 (dd, 2H), 1.5 (s, 3H), 1.33 (s, 3H).
`FTIR neat (cm-1): 1624, 1610. MS (ESI): m/e 438 (MH+). HPLC
`
`purity: method A 99%, retention time (tR) 3.53 min; method B
`98%, tR 7.98 min.
`1-[4-(5-Fluoro-2-hydroxyphenyl)-2-hydroxy-4-methyl-2-(tri-
`fluoromethyl)pentyl]-1H-quinolin-4-one (12). To a solution of 11
`(0.082 g, 0.19 mmol) in CH2Cl2 (3 mL) at -20 (cid:176) C was added
`dropwise BBr3 (1.9 mL of a 1 Msolution in CH 2Cl2, 1.9 mmol).
`The mixture was stirred for 1 h and quenched with saturated
`NaHCO3 and ethyl acetate. The organic layer was washed with
`saturated NaHCO3 and brine and dried (MgSO4). Removal of the
`volatiles in vacuo left a residue which was purified by trituration
`in ether and ethyl acetate and provided 0.041 g of product. Mp:
`222-223 (cid:176)C.
`1H NMR (400 MHz, DMSO-d6): (cid:228) 9.98 (s, 1H),
`8.30 (d, 1H), 7.87 (d, 1H), 7.75 (t, 1H), 7.52 (t, 1H), 7.33 (m, 2H),
`7.13 (m, 1H), 7.05 (m, 1H), 7.48 (s, 1H), 6.20 (d, 1H), 4.21 (m,
`2H), 3.24 (d, 1H), 2.15 (d, 1H), 1.75 (s, 3H), 1.55 (s, 3H). MS
`(ESI): m/e 424 (MH+). HPLC purity: method A 98%, tR 3.11 min;
`method B 95%, tR 7.16 min.
`cis-1-[4-(5-Fluoro-2-methoxyphenyl)-2-hydroxy-4-methyl-2-
`(trifluoromethyl)pentyl]-3,5-dimethylpiperidin-4-one Hydro-
`chloride (14). A mixture of 9 (0.092 g, 0.32 mmol), cis-3,5-
`dimethylpiperidin-4-one hydrochloride (13; 0.103 g, 0.64 mmol),
`and powdered K2CO3 (0.218 g, 1.58 mmol) in DMF (3 mL) was
`heated at 100 (cid:176)C for 1.5 h, cooled to room temperature, diluted
`with ether, washed thoroughly with water and brine, and dried
`(MgSO4). Removal of the volatiles in vacuo provided a residue
`which was treated with ethereal HCl. The mixture was filtered and
`the solid washed with ether and dried in vacuo to provide 0.015 g
`of product. Mp: 77-80 (cid:176)C.
`1H NMR (400 MHz, CDCl3): (cid:228) 7.02
`(dd, 1H), 6.78 (m, 1H), 6.68 (m, 1H), 3.75 (s, 3H), 1.52 (s, 3H),
`1.28 (s, 3H), 0.78 (d, 6H). MS (ESI): m/e 420 (MH+). HPLC
`purity: method C 92%, tR 10.25 min.
`1,1,1-Trifluoro-4-(5-fluoro-2-methoxyphenyl)-4-methyl-2-(mor-
`pholin-4-ylmethyl)pentan-2-ol Hydrochloride (15). Mp: 195-
`200 (cid:176)C.
`1H NMR (400 MHz, CDCl3): (cid:228) 6.97 (dd, 1H), 6.78 (m,
`1H), 6.68 (m, 1H), 4.55 (s, 1H), 3.75 (s, 3H), 3.46 (br s, 4H), 2.33
`(m, 4H), 2.14 (d, 2H), 1.97 (d, 2H), 1.52 (s, 3H), 1.32 (s, 3H). MS
`(ESI): m/e 380 (MH+). HPLC purity: method A 99%, tR 2.41 min;
`method B 99%, tR 6.35 min.
`1-[4-(5-Fluoro-2-methoxyphenyl)-2-hydroxy-4-methyl-2-(tri-
`fluoromethyl)pentyl]piperidin-4-one Hydrochloride (16). Mp:
`134-136 (cid:176) C. 1H NMR (400 MHz, DMSO-d6): (cid:228) 6.8 (m, 3H), 4.55
`(br s, 1H), 3.62 (s, 3H), 2.7-2.0 (m, 12H), 1.36 (s, 3H), 1.18 (s,
`3H). MS (ESI): m/e 392 (MH+). HPLC purity: method C 94%, tR
`7.42 min.
`1-[4-(5-Fluoro-2-methoxyphenyl)-2-hydroxy-4-methyl-2-(tri-
`fluoromethyl)pentyl]-1H-pyridin-4-one (17). Mp: 205-206 (cid:176)C.
`1H NMR (400 MHz, DMSO-d6): (cid:228) 7.48 (d, 2H), 7.12 (m, 3H),
`6.47 (s, 1H), 6.07 (d, 2H), 3.92 (s, 3H), 3.81 (d, 1H), 3.60 (d, 1H),
`2.92 (d, 1H), 2.18 (d, 1H), 1.65 (s, 3H), 1.45 (s, 3H). IR (cm-1):
`1639. MS (ESI): m/e 388 (MH+). HPLC purity: method A 99%,
`tR 2.72 min; method B 99%, tR 6.84 min.
`1,1,1-Trifluoro-4-(5-fluoro-2-methoxyphenyl)-2-(imidazol-1-
`ylmethyl)-4-methylpentan-2-ol (18). Mp: 166-167 (cid:176) C. 1H NMR
`(400 MHz, CDCl3): (cid:228) 8.1 (br s, 1H), 7.2-6.7 (m, 6H), 3.94 (dd,
`2H), 3.78 (s, 3H), 2.45 (s, 2H), 1.44 (s, 3H), 1.33 (s, 3H). MS
`(ESI): m/e 361 (MH+). HPLC purity: method A 99%, tR 2.56 min;
`method B 98%, tR 6.59 min; method C 96%, tR 8.67 min.
`2-[(trans-2,6-Dimethylmorpholin-4-yl)methyl]-1,1,1-trifluoro-
`4-(5-fluoro-2-methoxyphenyl)-4-methylpentan-2-ol Hydrochlo-
`ride (19). Mp: 50-55 (cid:176)C. Free base
`1H NMR (400 MHz,
`CDCl3): (cid:228) 7.02 (m, 1H), 6.81 (m, 1H), 6.69 (m, 1H), 4.68 (br d,
`1H), 3.76 (m, 5H), 2.26 (m, 4H), 2.1-1.85 (m, 4H), 1.51 (s, 3H),
`1.30 (s, 3H), 1.03 (br s, 6H). MS (ESI): m/e 408 (MH+). HPLC
`purity: method A 99%, tR 3.16 and 3.61 min; method B 99%, tR
`8.19 and 9.23 min.
`2-[(cis-2,6-Dimethylmorpholin-4-yl)methyl]-1,1,1-trifluoro-4-
`(5-fluoro-2-methoxyphenyl)-4-methylpentan-2-ol Hydrochloride
`(20). Mp: 133-135 (cid:176)C.
`1H NMR (400 MHz, DMSO-d6): (cid:228) 6.8
`(m, 3H), 4.13 (br s, 1H), 3.74 (br s, 2H), 3.58

This document is available on Docket Alarm but you must sign up to view it.


Or .

Accessing this document will incur an additional charge of $.

After purchase, you can access this document again without charge.

Accept $ Charge
throbber

Still Working On It

This document is taking longer than usual to download. This can happen if we need to contact the court directly to obtain the document and their servers are running slowly.

Give it another minute or two to complete, and then try the refresh button.

throbber

A few More Minutes ... Still Working

It can take up to 5 minutes for us to download a document if the court servers are running slowly.

Thank you for your continued patience.

This document could not be displayed.

We could not find this document within its docket. Please go back to the docket page and check the link. If that does not work, go back to the docket and refresh it to pull the newest information.

Your account does not support viewing this document.

You need a Paid Account to view this document. Click here to change your account type.

Your account does not support viewing this document.

Set your membership status to view this document.

With a Docket Alarm membership, you'll get a whole lot more, including:

  • Up-to-date information for this case.
  • Email alerts whenever there is an update.
  • Full text search for other cases.
  • Get email alerts whenever a new case matches your search.

Become a Member

One Moment Please

The filing “” is large (MB) and is being downloaded.

Please refresh this page in a few minutes to see if the filing has been downloaded. The filing will also be emailed to you when the download completes.

Your document is on its way!

If you do not receive the document in five minutes, contact support at support@docketalarm.com.

Sealed Document

We are unable to display this document, it may be under a court ordered seal.

If you have proper credentials to access the file, you may proceed directly to the court's system using your government issued username and password.


Access Government Site

We are redirecting you
to a mobile optimized page.





Document Unreadable or Corrupt

Refresh this Document
Go to the Docket

We are unable to display this document.

Refresh this Document
Go to the Docket