discovery of a potent and selective steroidal …...discovery of a potent and selective steroidal...

18
Discovery of a Potent and Selective Steroidal Glucocorticoid Receptor Antagonist (ORIC-101) Yosup Rew,* Xiaohui Du, John Eksterowicz, Haiying Zhou, Nadine Jahchan, Liusheng Zhu, Xuelei Yan, Hiroyuki Kawai, Lawrence R. McGee, Julio C. Medina, Tom Huang, Chelsea Chen, Tatiana Zavorotinskaya, Dena Sutimantanapi, Joanna Waszczuk, Erica Jackson, Elizabeth Huang, Qiuping Ye, Valeria R. Fantin, and Daqing Sun* ORIC Pharmaceuticals, 240 East Grand Avenue, Fl2, South San Francisco, California 94080, United States * S Supporting Information ABSTRACT: The glucocorticoid receptor (GR) has been linked to therapy resistance across a wide range of cancer types. Preclinical data suggest that antagonists of this nuclear receptor may enhance the activity of anticancer therapy. The rst- generation GR antagonist mifepristone is currently undergoing clinical evaluation in various oncology settings. Structure-based modication of mifepristone led to the discovery of ORIC-101 (28), a highly potent steroidal GR antagonist with reduced androgen receptor (AR) agonistic activity amenable for dosing in androgen receptor positive tumors and with improved CYP2C8 and CYP2C9 inhibition prole to minimize drugdrug interaction potential. Unlike mifepristone, 28 could be codosed with chemotherapeutic agents readily metabolized by CYP2C8 such as paclitaxel. Furthermore, 28 demonstrated in vivo antitumor activity by enhancing response to chemotherapy in the GR + OVCAR5 ovarian cancer xenograft model. Clinical evaluation of safety and therapeutic potential of 28 is underway. INTRODUCTION Glucocorticoids (GCs) such as cortisol are steroid hormones secreted by the adrenal gland in a circadian and stress- associated manner to regulate metabolism, cell growth, apoptosis and dierentiation, inammation, mood, and cognitive function. 1 GCs signal through the glucocorticoid receptor (GR), a member of the superfamily of nuclear receptors expressed across a wide variety of tissues. Upon ligand binding, GR translocates to the nucleus where it triggers transcription of a spectrum of genes that mediate its pleiotropic biological eects. Dysregulation of GR signaling has been associated with a variety of pathological states including Cushings syndrome, psychotic depression, obesity, and cancer. 2 Therefore, there has been considerable interest to target this nuclear receptor for therapeutic purposes. Several lines of evidence point to GR as a potential mechanism of therapy resistance in cancers of epithelial origin. 3 Elevated levels of GR protein have been correlated with poor prognosis in ESR1 breast cancer 4 and endometrial cancer patients 5 as well as to poor outcome in castration- resistant prostate cancer (CRPC) patients treated with enzalutamide. 6 In addition, cumulative preclinical data over the past decade have functionally linked GR activation with resistance to a variety of chemotherapeutic agents across an array of solid tumors that include ovarian cancer, triple- negative breast cancer (TNBC), prostate cancer, pancreatic cancer, nonsmall cell lung cancer, urological cancers, etc. 713 At the molecular level, GR drives resistance to therapy, at least in part, by driving transcriptional activation of mediators of cell survival, antiapoptosis, cell adhesion, and invasion. 9,1417 Collectively, these studies have established that genetic ablation or pharmacologic inhibition of GR reverses the pro- survival eects mediated by this nuclear receptor and enhances chemotherapy and antiandrogen activity in cancer mod- els. 69,14,18 These data have also provided rationale to evaluate the ecacy of GR antagonists in combination with chemo- therapeutic agents as well as enzalutamide in the clinic. Furthermore, it is well established that GR is expressed in a variety of immune cells. 19 Endogenous and synthetic Received: May 8, 2018 Published: August 9, 2018 Article pubs.acs.org/jmc Cite This: J. Med. Chem. 2018, 61, 7767-7784 © 2018 American Chemical Society 7767 DOI: 10.1021/acs.jmedchem.8b00743 J. Med. Chem. 2018, 61, 77677784 Downloaded via ORIC PHARMACEUTICALS INC on September 14, 2018 at 19:56:33 (UTC). See https://pubs.acs.org/sharingguidelines for options on how to legitimately share published articles.

Upload: others

Post on 21-Aug-2020

6 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: Discovery of a Potent and Selective Steroidal …...Discovery of a Potent and Selective Steroidal Glucocorticoid Receptor Antagonist (ORIC-101) Yosup Rew,* Xiaohui Du, John Eksterowicz,

Discovery of a Potent and Selective Steroidal GlucocorticoidReceptor Antagonist (ORIC-101)Yosup Rew,* Xiaohui Du, John Eksterowicz, Haiying Zhou, Nadine Jahchan, Liusheng Zhu, Xuelei Yan,Hiroyuki Kawai, Lawrence R. McGee, Julio C. Medina, Tom Huang, Chelsea Chen,Tatiana Zavorotinskaya, Dena Sutimantanapi, Joanna Waszczuk, Erica Jackson, Elizabeth Huang,Qiuping Ye, Valeria R. Fantin, and Daqing Sun*

ORIC Pharmaceuticals, 240 East Grand Avenue, Fl2, South San Francisco, California 94080, United States

*S Supporting Information

ABSTRACT: The glucocorticoid receptor (GR) has been linked to therapy resistance across a wide range of cancer types.Preclinical data suggest that antagonists of this nuclear receptor may enhance the activity of anticancer therapy. The first-generation GR antagonist mifepristone is currently undergoing clinical evaluation in various oncology settings. Structure-basedmodification of mifepristone led to the discovery of ORIC-101 (28), a highly potent steroidal GR antagonist with reducedandrogen receptor (AR) agonistic activity amenable for dosing in androgen receptor positive tumors and with improvedCYP2C8 and CYP2C9 inhibition profile to minimize drug−drug interaction potential. Unlike mifepristone, 28 could becodosed with chemotherapeutic agents readily metabolized by CYP2C8 such as paclitaxel. Furthermore, 28 demonstrated invivo antitumor activity by enhancing response to chemotherapy in the GR+ OVCAR5 ovarian cancer xenograft model. Clinicalevaluation of safety and therapeutic potential of 28 is underway.

■ INTRODUCTION

Glucocorticoids (GCs) such as cortisol are steroid hormonessecreted by the adrenal gland in a circadian and stress-associated manner to regulate metabolism, cell growth,apoptosis and differentiation, inflammation, mood, andcognitive function.1 GCs signal through the glucocorticoidreceptor (GR), a member of the superfamily of nuclearreceptors expressed across a wide variety of tissues. Uponligand binding, GR translocates to the nucleus where it triggerstranscription of a spectrum of genes that mediate itspleiotropic biological effects. Dysregulation of GR signalinghas been associated with a variety of pathological statesincluding Cushing’s syndrome, psychotic depression, obesity,and cancer.2 Therefore, there has been considerable interest totarget this nuclear receptor for therapeutic purposes.Several lines of evidence point to GR as a potential

mechanism of therapy resistance in cancers of epithelialorigin.3 Elevated levels of GR protein have been correlatedwith poor prognosis in ESR1− breast cancer4 and endometrialcancer patients5 as well as to poor outcome in castration-resistant prostate cancer (CRPC) patients treated with

enzalutamide.6 In addition, cumulative preclinical data overthe past decade have functionally linked GR activation withresistance to a variety of chemotherapeutic agents across anarray of solid tumors that include ovarian cancer, triple-negative breast cancer (TNBC), prostate cancer, pancreaticcancer, nonsmall cell lung cancer, urological cancers, etc.7−13

At the molecular level, GR drives resistance to therapy, at leastin part, by driving transcriptional activation of mediators of cellsurvival, antiapoptosis, cell adhesion, and invasion.9,14−17

Collectively, these studies have established that geneticablation or pharmacologic inhibition of GR reverses the pro-survival effects mediated by this nuclear receptor and enhanceschemotherapy and antiandrogen activity in cancer mod-els.6−9,14,18 These data have also provided rationale to evaluatethe efficacy of GR antagonists in combination with chemo-therapeutic agents as well as enzalutamide in the clinic.Furthermore, it is well established that GR is expressed in a

variety of immune cells.19 Endogenous and synthetic

Received: May 8, 2018Published: August 9, 2018

Article

pubs.acs.org/jmcCite This: J. Med. Chem. 2018, 61, 7767−7784

© 2018 American Chemical Society 7767 DOI: 10.1021/acs.jmedchem.8b00743J. Med. Chem. 2018, 61, 7767−7784

Dow

nloa

ded

via

OR

IC P

HA

RM

AC

EU

TIC

AL

S IN

C o

n Se

ptem

ber

14, 2

018

at 1

9:56

:33

(UT

C).

Se

e ht

tps:

//pub

s.ac

s.or

g/sh

arin

ggui

delin

es f

or o

ptio

ns o

n ho

w to

legi

timat

ely

shar

e pu

blis

hed

artic

les.

Page 2: Discovery of a Potent and Selective Steroidal …...Discovery of a Potent and Selective Steroidal Glucocorticoid Receptor Antagonist (ORIC-101) Yosup Rew,* Xiaohui Du, John Eksterowicz,

glucocorticoids are potent regulators of inflammation.20,21

Glucocorticoid binding to GR activates or represses tran-scription of a variety of pro- and anti-inflammatory genes, andthe functional consequences of glucocorticoid action differ bycell type. GR activation has been shown to promote expansionof regulatory T cells and myeloid derived suppressor cells,22−24

stimulation of Th2 differentiation,25 as well as suppression ofactivated CD8+ T cells,26 and most of these effects werereversed by the GR antagonist mifepristone (also known asRU486). Importantly, promotion of immunosuppressive cellsand T cell exhaustion have been implicated in cancer immuneescape as well poor response to immune therapy.27

Furthermore, disruption in cortisol secretion has beencorrelated with poor prognosis and immunosuppression acrossa variety of advanced solid tumors including breast, ovarian,and lung cancer.28−30 In addition, extra-adrenal production ofcortisol by colorectal cancer, squamous skin cancer, andmelanoma cells has been associated with decreased CD8+ Tcell proliferation and activity,31,32 suggesting that aberrantactivation of GR could contribute to local tumor immunosup-pression. Altogether these observations point to theimmunomodulatory potential of GR and provide a rationalbasis for the clinical evaluation of GR antagonists incombination with emerging immunotherapies.The marketed drug mifepristone (RU-486) is a steroidal GR

antagonist, discovered in the 1980s as a potent progesteronereceptor (PR) antagonist.33 Because of its potent GRinhibitory activity, mifepristone has also been evaluated forconditions associated with alterations in GR activity.34

Mifepristone was approved in 2011 for the treatment ofCushing’s syndrome and is currently undergoing clinicaltesting in various solid tumors.Compared to recently discovered synthetic nonsteroidal GR

antagonists and allosteric inhibitors,35−40 mifepristone (1)stands out for its potency. However, mifepristone is a partialAR agonist, and its agonistic activity is sufficient to stimulatethe proliferation of CRPC LNCaP/AR-luc (LNAR) cells bothin vitro and in vivo. In addition, mifepristone has also beenshown to induce AR target gene expression in AR+ TNBCMDA-MB-453 cells.41 Besides prostate cancer, previous studieshave shown the oncogenic roles of AR and androgen signalingin TNBC and ovarian cancer.42−45 In fact, the therapeuticpotential of the antiandrogen enzalutamide is currently beingclinically evaluated in AR+ TNBC and ovarian cancerpatients.45−47 Clearly, partial AR agonism activity is anunwanted feature for any GR antagonist with potential clinicalapplication in AR-driven cancers. We envisioned that theundesirable AR agonism of mifepristone could be eliminatedwhile retaining its GR antagonistic potency by structure-basedmodification of mifepristone. We report here the results of ourstructure−activity relationship (SAR) studies, which led to thediscovery of ORIC-101 (28) (Figure 1), a highly potent GRantagonist with significantly reduced AR agonism andimproved cytochrome P450 (CYP) inhibition profiles. Thesefeatures of 28 make this novel compound an attractivecombination partner to antiandrogens and paclitaxel in AR+

cancers including CRPC as well as a subset of TNBC andovarian cancers.14,44,46,48 In addition, the favorable CYPinhibition profile of 28 makes this compound combinablewith therapeutic agents that are metabolized by CYP2C8 andCYP2C9.

■ RESULTS AND DISCUSSIONWith the goal of developing a potent, steroidal GR antagonistwith reduced AR agonism as compared to 1, an analysis of thebinding mode of mifepristone to AR and GR was initiated. Theprotein cocrystal structure of 1 bound to GR, but not to AR,has been solved (Figure 2).49,50 Because the two nuclearreceptors are closely related, a homology model of AR wasconstructed based on the GR structure (pdb 3H52). Figure 2Ashows a view of the mifepristone (1)/GR binding siteillustrating the key hydrogen bond interactions of the steroidcarbonyl with Arg611 and Gln570. Outside of the interactionwith these two polar residues, the majority of binding siteresidues are hydrophobic in nature and form favorable van derWaals interactions with the predominately hydrophobic ligand.Figure 2B shows the complementary fit between the ligand andthe binding site, with 1 almost completely buried in the steroidbinding pocket. The helices that contain residues that interactwith mifepristone and the NCoR peptide are also labeled.The agonistic and antagonistic functions of GR and AR

ultimately depend on the protein conformation,51−53 inparticular, the positions of helices 11 and 12 and the abilityor inability of the receptor to bind coactivators orcorepressors.54 To that end, structural hypotheses weredeveloped along three distinct vectors with the goal of alteringthe GR and AR structures and thus affecting the agonist andantagonist profiles of the ligands. Using the mifepristonescaffold as a foundation, modification of the C11 4-dimethylaminophenyl group provided a vector to alter theposition of helix 12 and also to directly challenge copeptidebinding. Modification of the C17 hydroxyl and introduction ofC16 substituents afforded the opportunity to disrupt thepositions of both helices 11 and 12 directly,55 and modificationof the C17 1-propynyl group provided a cascading structuraleffect where disruption of helix 7 in turn altered the position ofhelices 11 and 12.Initial investigation of 1 began with replacement of the 4-

dimethylaminophenyl group at the C11 position in the steroidcore with various substituents (Table 1, compounds 2−6).56−58 To measure antagonist and agonist activities of thesecompounds on GR and AR, luciferase reporter assays wereemployed.59,60 Substitution of the 4-dimethylaminophenylgroup with a small alkyl group, such as an ethyl, switchedthe GR activity, converting antagonism to agonism (2 vs 1).Increase of the substituent’s size to cyclohexyl or unsubstitutedphenyl group restored GR antagonism and attenuated GRagonism (3 and 4 vs 2). These results imply that a phenyl or asimilar-sized cyclohexyl moiety at the C11 position is animportant feature for GR antagonism. Moving the dimethyla-mino group from the para-position to the meta-position

Figure 1. Mifepristone (1) and ORIC-101 (28). aIC50 in GRluciferase antagonist assay. bEC50 [Emax]* in AR luciferase agonistassay. * = % mifepristone.

Journal of Medicinal Chemistry Article

DOI: 10.1021/acs.jmedchem.8b00743J. Med. Chem. 2018, 61, 7767−7784

7768

Page 3: Discovery of a Potent and Selective Steroidal …...Discovery of a Potent and Selective Steroidal Glucocorticoid Receptor Antagonist (ORIC-101) Yosup Rew,* Xiaohui Du, John Eksterowicz,

reduced AR agonism dramatically (5.8% of 1) but decreasedGR antagonism by 20-fold as well (5 vs 1). The 4-isopropylphenyl derivative elicited improvement in GRantagonism (IC50 = 1.93 nM for 6). However, it also inducedmore AR agonism (200% of 1).Attention was then focused on the modification of the 1-

propynyl group at the C17 position (Table 1, compounds 7−11).56,61 Changing the 1-propynyl group to a saturated n-alkylgroup amplified AR agonism while diminishing GR antagonism(7 vs 1), suggesting that a C−C triple bond at the C17position is crucial for minimized AR agonism and enhancedGR antagonism. The introduction of an electron withdrawinggroup by replacing the 1-propynyl group with a 3,3,3-trifluoro-1-propynyl group maintained the same level of AR agonism (8vs 1). Interestingly, gradually increasing alkyl substituent sizein the 1-propynyl group led to the 3,3-dimethyl-1-butynylderivative 11, which possessed substantially reduced partialagonism in the AR luciferase assay (13% of 1). However, 11also exhibited a 4.5-fold decrease in GR antagonism potencycompared to 1 (compounds 9−11 vs 1).As part of the lead optimization process, a cocrystal structure

of 11 bound to human GR was determined at a resolution of3.05 Å (Figure 3). The ketone moiety of 11 makes keyhydrogen bond interactions with Arg611 and Gln570, andoverall, the ligand sits in a similar position as that ofmifepristone in the binding site. There is, however, a significantdifference in the protein structure in the region interactingwith the C17 substituent. As can be observed in themifepristone cocrystal structure, the 1-propynyl group sits ina hydrophobic region between helices 3, 6, and 7 (Figure 2).The orientation of these helices in turn has an effect on theposition of helices 11 and 12. The resulting overallconformation of GR is favorable for binding of the corepressorpeptide, NCoR. This is consistent with the antagonistic natureof mifepristone toward GR. The replacement of the alkynylmethyl with a t-butyl group in 11 results in significant changesin the shape and size of the binding site and the overallconformation of GR. Figure 3 illustrates a significant increasein the size and depth of this part of the binding pocket that isnecessary to accommodate 11. Consequently, significantchanges occur in the orientations of helices 6 and 7, resulting

in a shift in the positions of helices 11 and 12 as well. In fact,the helix 12 position prevents the binding of corepressor orcoactivator peptides (Supporting Information, Figure S4).62

While mifepristone does not show significant agonism towardGR (Emax relative to dexamethasone of 3.1%), 11 furtherreduces the relative Emax by more than 50%.As mentioned, the addition of t-butyl to the alkyne has a

significant effect on AR selectivity. A more than 7-foldreduction in AR agonism (Emax) is observed for 11 versusmifepristone. To date, efforts to obtain a cocrystal structure ofAR with 11 have not been successful. However, given the closestructural relationship between AR and GR, a similar structuralhypothesis can be developed through molecular modeling toexplain the reduced AR agonism of 11. The orientation of 11in the binding site of AR is expected to be similar to the oneobserved for GR, and thus a similar disruption of the positionsof helices 6, 7, 11, and 12 is expected. These largeconformational changes in turn disrupt the copeptide bindingsite further reducing the AR agonism for 11 versus mifepri-stone.A single-crystal small-molecule X-ray structure of 11 was

also obtained (Figure 4). This structure allowed forconfirmation of the absolute stereochemistry of all of thechiral centers in the molecule and was also used to evaluate thecocrystal structure binding mode. The steroid scaffold is rigid,and overall 11 has few rotatable bonds. Nevertheless, structuraldistortion of the ligand is still possible and could result in ahigh energy binding conformation. The ligand conformation of11 from the GR protein cocrystal structure is shown in B, whileC shows the overlay of the two and illustrates that theconformations are almost identical. There is no significantchange in ligand conformation upon binding to GR, and thusno additional strain is introduced into the molecule.Having identified the 3,3-dimethyl-1-butynyl group as the

optimized 1-alkynyl group at the C17 position with respect toAR selectivity, attention was turned to substituent modificationof the aryl ring at the C11 position (Table 2). Replacing the 4-dimethylamino group in 11 by small alkyl groups was toleratedin terms of GR antagonism potency, but those derivativesdisplayed greater AR agonism as the size of the alkyl groupincreased (12, 13, and 14). Replacement of the 4-

Figure 2. (A) 1 (gray) bound to GR (pdb 3H52). GR active site residues shown in blue. (B) GR protein surface with helices labeled and NCoRpeptide shown in purple.

Journal of Medicinal Chemistry Article

DOI: 10.1021/acs.jmedchem.8b00743J. Med. Chem. 2018, 61, 7767−7784

7769

Page 4: Discovery of a Potent and Selective Steroidal …...Discovery of a Potent and Selective Steroidal Glucocorticoid Receptor Antagonist (ORIC-101) Yosup Rew,* Xiaohui Du, John Eksterowicz,

dimethlyamino group with a methoxy or methyloxetanyl groupwas also well tolerated for GR antagonism (15 and 25).Shifting the dimethylamino substituent from the para-positionto the meta-position led to a drop in GR antagonism potency(16) as observed in the previous SAR with compound 5.Additional substitutions at the meta-and ortho-positions werethen evaluated while preserving the dimethylamino group atthe para position. The 2,5-difluorophenyl analogue 18 showedGR antagonism potency similar to that of 11, whereas potencyof all other analogues was diminished (17, 19, and 20). Theoptimization of the N,N-dialkyl group within 11 was attemptedto improve GR antagonism potency and to address theinherent metabolic instability of the N,N-dimethylaminogroup. Compound 21, which exchanged one N-methyl group

with an isopropyl group, was lower in GR antagonism activitycompared to 11 (IC50 = 43 nM for 21 vs IC50 = 14 nM for 11)but improved AR agonism selectivity (Emax = 8.2% of 1 for 21vs Emax = 13% of 1 for 11). Introduction of various cyclictertiary amines was also tolerated with regard to GR antagonistpotency (22, 23, and 24). Overall, no significant improvementin GR antagonist potency was observed from the aryl groupmodification at the C11 position.The X-ray cocrystal structure of 11 bound to GR protein

(Figure 3) shows that the A ring carbonyl oxygen forms directhydrogen bonds to the guanidinium of Arg611 and to the γ-amide of Gln570. The A-ring carbonyl oxygen in compound 11is α,β−γ,δ-diene carbonyl oxygen with extended conjugation,which is known to be less electronegative than α,β-unsaturated

Table 1. Exploration of Substituents at C11 and C17 Positions in Mifepristonec

aPercent dexamethasone. bPercent mifepristone. cPotency and Emax data with SD are reported as the average of at least two determinations.

Journal of Medicinal Chemistry Article

DOI: 10.1021/acs.jmedchem.8b00743J. Med. Chem. 2018, 61, 7767−7784

7770

Page 5: Discovery of a Potent and Selective Steroidal …...Discovery of a Potent and Selective Steroidal Glucocorticoid Receptor Antagonist (ORIC-101) Yosup Rew,* Xiaohui Du, John Eksterowicz,

carbonyl oxygen. Therefore, we hypothesized that reducing theC9 C−C double bond in the B ring could enhance hydrogenbond acceptor ability of the A ring carbonyl oxygen.Consequently, an effort was made to explore modification ofthe steroid core in 11 (Table 3).Of significance was the observation that 26, the correspond-

ing C9 C−C double bond reduced analogue of 11, had amarked 3-fold improvement in GR antagonist potency (IC50 =5.6 nM for 26 vs IC50 = 14 nM for 11). However, 26 showed aslight increase in AR agonist activity (Emax = 22% of 1)compared to 11 (Emax = 13% of 1). This increase is notnegligible. For example, the comparable partial agonisticactivity of bicalutamide (Emax = 23% of 1) compromises itstherapeutic efficacy as an antiandrogen relative to fullantagonists such as enzalutamide (Figure 5) and apalutamide.The 3,3-dimethyl-1-butynyl group in 26 was further

modified by replacing one methyl of the t-butyl group with amethoxy moiety. This was attempted to reduce AR agonismand improve solubility, but it also resulted in lower GRantagonism (27). On the other hand, similar to previousobservations, the replacement of one N-methyl group in 26with an isopropyl group provided 28 with significantly reducedAR agonism (Emax = 11% of 1), which is comparable to ARagonism of enzalutamide (Emax = 8.7% of 1) (Figure 5).Overall, compound 28 is a weak full AR antagonist with IC50 of484 ± 110 nM in the AR luciferase assay. Furthermore,compound 28 is equipotent to 26 and only 2-fold less potentthan mifepristone in the GR luciferase antagonism assay (IC50= 5.6 nM).

We also compared the AR agonism of compounds 1 and 28in the LNAR cells, which exhibit high AR, but low GR, and lowPR expression, as well as in the MDA-MB-453 cells with highAR, high GR, but no PR expression by RT-qPCR (Figures 6and 7). The results showed that 1, but not 28, is able tostimulate endogenous AR target gene expression in bothLNAR and MDA-MB-453 cell lines at about 100 nM. Theseexpression levels are similar to those achievable by 0.1 nMR1881, the potent synthetic androgen in the same cell lines.On the other hand, 1 μM enzalutamide completely revertedgene expression induced by R1881, but not 1, in the LNARcell line (Figure 6). Notably, endogenous AR target geneinduction by compound 28 was minimal in both cell lines(Figures 6 and 7). These data suggest that compound 28exhibits no AR agonism and might have an advantage over 1 intreating AR-driven cancers.The single-crystal small-molecule X-ray structure of 28 was

solved. Figure 8 shows the structure of 28 as compared to theprotein cocrystal structure of 11. Because the overlay showsthat the overall shape of both compounds is very similar, 28would be expected to have a very similar binding mode to thatof 11. This is consistent with the binding mode of 28, aspredicted via docking calculation with GR.To identify a molecule suitable for preclinical development,

several selected compounds were evaluated for their ratpharmacokinetic (PK) profile (Table 4). The results showedthat 13 and 23 had low clearance but poor oral bioavailability(CL = 0.41 and 0.28 L/kg/h; %F = 0 and 0.7%, respectively).Compounds 11 and 28 had moderate clearance (CL = 1.4 and1.9 L/kg/h, respectively) and acceptable oral bioavailability (%F = 13 and 24%, respectively). The rest of the compounds 21,25, and 27 showed high CL (CL = 3.0−5.0 L/kg/h), although27 exhibited good oral bioavailability (%F = 54%).Given the GR antagonist potency, low AR agonism, and

favorable rat PK profile, compound 28 was selected for furtherevaluation. The preclinical pharmacokinetics of 28 wereevaluated in mouse and minipig following single-dose intra-venous (iv) or oral administration. The molecule was found tohave low clearance and reasonable oral bioavailability in mouseand minipig (Table 5).63

Species differences in the unbound fraction of compound 28were minimal (<2−3-fold). The unbound fraction (0.14) of

Figure 3. (A) 11 (dark gray) bound to GR (pdb 6DXK). GR active site residues shown in olive. (B) GR protein surface with helices labeled.

Figure 4. Comparison of small-molecule single-crystal X-ray structureof 11 (A, orange) with the GR cocrystal X-ray structure 11 (B, darkgray, pdb 6DXK). The overlay of the two structures is shown in C.

Journal of Medicinal Chemistry Article

DOI: 10.1021/acs.jmedchem.8b00743J. Med. Chem. 2018, 61, 7767−7784

7771

Page 6: Discovery of a Potent and Selective Steroidal …...Discovery of a Potent and Selective Steroidal Glucocorticoid Receptor Antagonist (ORIC-101) Yosup Rew,* Xiaohui Du, John Eksterowicz,

compound 28 is 10-fold higher than that of compound 1 inhuman plasma. As compound 1 is reported to have highpotential for inhibition of CYP2C8, we next examined theliability of 11 and 28 in the CYP inhibition assay. It was found

that both 11 and 28 have low potential of CYP2C8 andCYP2C9 inhibition compared to 1, although the IC50 values of11 and 28 for CYP3A4 are lower than that of 1 (Table 6). Ingeneral, the replacement of the C17 alkynyl methyl in 1 with a

Table 2. Exploration of Aryl Substituents at C11 Position in Compound 11c

aPercent dexamethasone. bPercent mifepristone. cPotency and Emax data with SD are reported as the average of at least two determinations.

Journal of Medicinal Chemistry Article

DOI: 10.1021/acs.jmedchem.8b00743J. Med. Chem. 2018, 61, 7767−7784

7772

Page 7: Discovery of a Potent and Selective Steroidal …...Discovery of a Potent and Selective Steroidal Glucocorticoid Receptor Antagonist (ORIC-101) Yosup Rew,* Xiaohui Du, John Eksterowicz,

t-butyl group improves the CYP inhibition profile, especiallyagainst CYP2C8 and CYP2C9.

Furthermore, the in vivo combination PK studies of 1 or 28with paclitaxel, a CYP2C8 substrate, in female micedemonstrated that 28 showed less potential for CYP2C8driven drug−drug interaction compared to 1 (Figure 9). Theresults showed that adding 1 to paclitaxel resulted in higherpaclitaxel plasma exposure. Unlike 1, 28 did not result in theincrease in paclitaxel concentration in plasma.To evaluate the selectivity of compound 28 for GR over PR,

both 28 and 1 were tested in the PR luciferase reporter assay.The results showed that the inhibitory activity of 28 wassubstantially lower than that of 1 (IC50 = 21 ± 10 nM for 28;IC50 = 0.4 ± 0.2 nM for 1). The PR/GR ratio of 28 is 3.8,while that of 1 is 0.12. Compound 28 does not show anyagonism in the PR luciferase reporter assay. Unlike theestrogen receptor (ER) ligand 17β-estradiol and the ERantagonist tamoxifen, 28 does not physically bind to ER up to1 μM (Supporting Information, Figure S1). Similarly, 28neither induces nor inhibits aldosterone-induced interactionbetween MR and its coactivator SRC at the concentration of 5μM (Supporting Information, Figures S2 and S3), suggesting28 is not a mineralocorticoid receptor (MR) agonist orantagonist.To compare the activity of 1 and 28 on GR across multiple

species, expression of endogenous GR target genes wasevaluated using human, rat, dog, monkey, and minipigperipheral blood mononuclear cells (PBMCs) treated ex vivowith dexamethasone in the presence or absence of 1 or 28.Figure 10 shows an example of quantitative polymerase chainreaction (RT-qPCR) results measuring the dose-dependentexpression levels of two established GR target genes, GILZ andFKBP5,64 in human PBMCs treated with dexamethasone and28. Treatment with 28 significantly inhibited ligand-induced

Table 3. Modification of the Steroid Core of 11c

aPercent dexamethasone. bPercent mifepristone. cPotency and Emax data with SD are reported as the average of at least two determinations.

Figure 5. Relative AR agonism of 11 and 28 compared to 1,enzalutamide, and bicalutamide.

Figure 6. Induction of endogenous AR target genes by syntheticandrogen (R1881) and 1 in LNAR cells. Endogenous AR targetgenes, PSA and TMPRSS2, are induced by R1881 and 1 in LNARcells while those genes are not induced by 28. Enz (enzalutamide)inhibits R1881-induced expression but does not fully inhibit theexpression induced by 1.

Journal of Medicinal Chemistry Article

DOI: 10.1021/acs.jmedchem.8b00743J. Med. Chem. 2018, 61, 7767−7784

7773

Page 8: Discovery of a Potent and Selective Steroidal …...Discovery of a Potent and Selective Steroidal Glucocorticoid Receptor Antagonist (ORIC-101) Yosup Rew,* Xiaohui Du, John Eksterowicz,

target gene expression with potencies comparable to 1 in allspecies tested (Table 7).Next, we evaluated whether 28 could inhibit endogenous

GR target gene expression and compared its relative potencywith 1 in cancer cells. To this end, the GR+ human ovariancancer cell line OVCAR5, which has no detectable AR and lowPR expression levels, was stimulated with dexamethasone inthe presence or absence of 28 or 1 and mRNA levels of GRtarget genes were measured by RT-qPCR. Compound 28reduced expression of FKBP5 and GILZ in a dose-dependentmanner (IC50 = 17.2 and 21.2 nM, respectively) with similarpotency as 1 (IC50 = 10.9 and 13.0 nM, respectively) (Figure11).The ability of 28 to inhibit GR transcriptional activity was

further evaluated in vivo. The major murine glucocorticoid,corticosterone, is a weak agonist of human GR.65 Therefore,when human cancer cells are grown as tumor xenografts inmice, it is necessary to provide an exogenous ligand such ascortisol or the synthetic ligand dexamethasone to effectivelyactivate human GR. Expression of the GR target gene FKBP5was analyzed in OVCAR5 tumors isolated from animals 6 hafter administration of vehicle or dexamethasone (0.5 mg/kgresults in a plasma glucocorticoid concentration equivalent to ahuman cortisol dose of 0.100−0.467 μM corrected forpotency) in the presence or absence of a single oral dose of28 at 3 different concentrations. In the presence of 28 at 25,50, or 75 mg/kg, dexamethasone-induced FKBP5 expressionwas reduced in a dose dependent manner (Figure 12A).Expression of FKBP5 in the 75 mg/kg group was notsignificantly different than expression in the vehicle group.The unbound plasma concentration of 28 in circulating mouseplasma ranged from 0.02 to 0.12 μM across the three groups(Figure 12B). The plasma exposures of dexamethasone weresimilar in all animals that received the dose of glucocorticoid(data not shown). These data suggest that 28 is effective atinhibiting dexamethasone-induced GR target gene expressionin OVCAR5 xenograft tumors.An in vivo study was carried out to assess the efficacy of 28

in combination with gemcitabine and carboplatin in theOVCAR5 ovarian cancer xenograft model (Figure 13). As

Figure 7. Stimulation of AR activity in AR+ TNBC cells by 1. Compound 1, but not 28, stimulates AR target gene expression in the AR+ MDA-MB-453 TNBC cell line.

Figure 8. Comparison of the small-molecule single-crystal X-raystructure of 28 (A, blue) with the GR cocrystal X-ray structure 11 (B,dark gray, pdb 6DXK). The overlay of the two structures is shown inC.

Table 4. Rat Pharmacokinetic Data for SelectedCompoundsa

iv (0.5 mg/kg)b po (5 mg/kg)c

compdCL

(L/kg/h)Vss

(L/kg)t1/2(h) F (%)

Cmax(μg/L)

AUC(μg·h/L)

1 3.7 4.1 1.6 6.4 24 8511 4.0 4.9 1.9 37 72 47113 0.41 1.7 3.4 0 0 021 4.0 4.7 1.5 23 59 29023 0.28 1.5 3.6 0.7 21 16725 3.0 6.7 4.4 7.2 59 12327 5.0 7.2 2.4 54 295 54428 1.9 4.6 4.4 24 149 635

an = 3 mice for both iv and po. iv time points: one predose and 10postdose. po time points: one predose and 8 postdose. bFormulatedin 10% DMSO, 70% PEG 400, and 20% water. cFormulated in 5%DMSO, 95% 0.2% Tween 80 in 0.25% CMC.

Table 5. Pharmacokinetic Data for 28 in Mouse and Minipig

iv (0.5 mg/kg)apo (5 mg/kg for mouse and 10

mg/kg for minipig)b

speciesCL

(L/kg/h)Vss

(L/kg) t1/2 (h)F(%)

Cmax(μg/L)

AUC(μg·h/L)

mouse 1.63 5.01 4.83 21 274 644minipig 1.1 7.8 12.2 20 153 1721

aFormulation: 28 was in 5% DMA, 10% EtOH, 40% PEG400, and45% D5W for mouse iv study; and in 5% DMA, 10% EtOH, 40% PEG400, 43.8% D5W, pH 5, for minipig iv study. bFormulation: 28 was in0.25% CMC, 0.2% Tween 80, and 5% DMSO for mouse oral study,and 28 HCl salt was in 2% HPMC and 0.2% Tween 80 for minipigoral study.

Table 6. CYP Inhibition of Compounds 1, 11, and 28

IC50 (μM)

P450 isoform substrate 1 11 28

3A4 midazolam 9.5 2.9 1.62C8 amodiaquine 1.5 >10 >102C9 diclofenac 4.9 >10 >10

Journal of Medicinal Chemistry Article

DOI: 10.1021/acs.jmedchem.8b00743J. Med. Chem. 2018, 61, 7767−7784

7774

Page 9: Discovery of a Potent and Selective Steroidal …...Discovery of a Potent and Selective Steroidal Glucocorticoid Receptor Antagonist (ORIC-101) Yosup Rew,* Xiaohui Du, John Eksterowicz,

previously mentioned, corticosterone is the prevailing circulat-ing glucocorticoid in rodents and is a weak activator of humanGR. Therefore, to achieve proper activation of GR in thehuman xenograft transplant model, cortisol was administeredchronically in the drinking water at 100 mg/L before tumorimplantation and for the duration of the study in the groupsindicated. The resulting plasma cortisol levels are consistentwith reported mean plasma cortisol levels in cancer patientsthroughout the day, which range from 0.100 to 0.496 μM.66,67

Daily treatment with 28 did not affect the growth of thetumors grown under chronic cortisol. Gemcitabine andcarboplatin induced tumor growth inhibition in groups withor without 28 compared to vehicle alone in the presence ofcortisol. However, addition of 28 significantly enhanced theantitumor effect of chemotherapy. Tumor volumes on day 21in mice receiving gemcitabine + carboplatin + cortisol weresignificantly larger than the tumor volumes in mice receivinggemcitabine + carboplatin + cortisol + 28. These dataindicated that 28 was effective in reversing the GR mediatedresistance to chemotherapy driven by cortisol in vivo.

■ CHEMISTRYCompounds 2−23 were prepared in a straightforward manner,through the route outlined in Scheme 1.61 Their synthesis wasinitiated by incorporation of C17 substituent in thecommercially available ethylene deltanone 29. Grignardreaction of ketone 29 with alkynylmagnesium bromide oralkynyllithium provided 17β-hydroxy ketal 30 exclusively.Treatment of 30 with 30% hydrogen peroxide and1,1,1,3,3,3-hexafluoropropan-2-one trihydrate in the presenceof sodium hydrogen phosphate buffer provided epoxide 31 as amajor diastereomer (dr >3:1). Copper-catalyzed addition of analkyl or aryl Grignard reagent to the vinylic epoxide 31,followed by hydrolysis with 70% acetic acid at 55 °C, gavecompounds 2−23 in moderate to high yields.Synthesis of compounds 24 and 25 was completed in a

similar manner but required extra steps as illustrated in Scheme2. The primary aniline derivative 33a was prepared fromepoxide 31f via copper catalyzed 1,4-addition of thecommercially available (4-(bis(trimethylsilyl)amino)phenyl)magnesium bromide 34, followed by acetic acid-mediatedhydrolysis. The double Michael addition of aniline 33a todivinyl sulfone afforded 24. For synthesis of 25, a commerciallyavailable diol 35 was converted to cyclic ketal 36 with 2,2-dimethoxypropane. 1,4-Addition of in situ generated Grignardreagent of 36 to epoxide 31f, followed by hydrolysis with 70%

Figure 9. In vivo combination pharmacokinetics of 1 and 28 with paclitaxel in mice.

Figure 10. GR target gene expression in PBMCs. Representative RT-qPCR measuring FKBP5 and GILZ in PBMCs from a healthy humandonor treated with dexamethasone at 30 nM and 28 at dose rangingconcentrations for 6 h.

Table 7. Cross-Species Evaluation of the Effect of 1 and 28 on Endogenous Target Gene Expression in PBMCs

human rat dog monkey minipig

GILZ FKBP5 GILZ FKBP5 GILZ FKBP5 GILZ FKBP5 GILZ FKBP5

IC50 of 1 (nM) 17.8 14.8 NDa NDa 32.6 20.2 13.9 21.9 25.7 19.7IC50 of 28 (nM) 24.9 19.5 59.4 45.6 34.3 24.5 14.9 11.1 181 39.3

aNot determined.

Figure 11. Inhibitory effects of 28 and 1 on GR-mediated target geneexpression in ovarian cancer OVCAR5 cells.

Journal of Medicinal Chemistry Article

DOI: 10.1021/acs.jmedchem.8b00743J. Med. Chem. 2018, 61, 7767−7784

7775

Page 10: Discovery of a Potent and Selective Steroidal …...Discovery of a Potent and Selective Steroidal Glucocorticoid Receptor Antagonist (ORIC-101) Yosup Rew,* Xiaohui Du, John Eksterowicz,

acetic acid gave diol 33b. Finally, intramolecular Mitsunobureaction of 33b yielded 25.68

Scheme 3 describes the synthesis of compounds 26, 27, and28. The C17 ketone group in 29 was reduced by sodiumborohydride to provide 17β-hydroxy ketal 37 as a singleisomer, which was used in the next step without furtherpurification. Oxidation of 37 with hydrogen peroxide led to amixture of epoxides with the desired epoxide as major (dr =4:1). Copper-catalyzed 1,4-addition of the Grignard reagent toepoxide 38 followed by acetic acid-mediated hydrolysisresulted in dienone 39. Birch reduction of conjugated α,β-γ,δ-unsaturated ketone 39 into unconjugated β,γ-unsaturatedketone 40,69 followed by strong acid catalyzed double bondmigration, provided α,β-unsaturated ketone 41.70 Enone 41was then protected as thioketal 42 under acidic conditionswithout double bond migration.70 The hydroxyl group in 42

was oxidized to ketone through Oppenauer oxidation. Alkynyllithium addition to the ketone 43 followed by deprotection of

Figure 12. Effect of 28 on dexamethasone-induced target gene expression in OVCAR5 xenograft tumors. (A) mRNA levels of the GR target gene,FKBP5, in OVCAR5 tumors relative to vehicle 6 h after treatment (n = 4 mice per group). Dexamethasone (Dex) was administered byintraperitoneal injection at 0.5 mg/kg, and a single dose of 28 was administrated orally by gavage at 25, 50, and 75 mg/kg. (B) Bar graph showingthe mean unbound plasma concentrations of 28 in the three treated groups 6 h after treatment with Dex at 0.5 mg/kg (n = 4 mice per group).Significance of effects on FKBP5 expression was determined by One-Way ANOVA using Dunnett’s test to correct for multiple comparisons. The p-values compared to the vehicle group are 0.0004 (***) for the dexamethasone group, 0.0036 (**) for the Dex + 28 at 25 mg/kg group, and notsignificant (ns) for the Dex + 28 at 50 mg/kg group (p = 0.1111) and the Dex + 28 at 75 mg/kg group (p = 0.7506).

Figure 13. Effects of 28 on gemcitabine and carboplatin response inOVCAR5 xenograft tumor in cortisol-treated mice. Tumor growthcurves for mice with established OVCAR5 xenografts grown in thepresence of cortisol at 100 mg/L (n = 10 mice per group). Animalswere dosed with gemcitabine 100 mg/kg and carboplatin 60 mg/kgevery 3 days for 4 doses. 28 was dosed at 75 mg/kg twice a day byoral gavage for the duration of the study. Data are displayed as mean± SEM. The star (*) in both panels indicates statistical significanceon day 21 between the two groups indicated, with p < 0.05.

Scheme 1a

aReagents and conditions: (a) for 30a, CH3CCMgBr, THF, 0 to 25°C; for 30b−30f, R2Li, −78 to 25 °C, 24−87%; (b) 35% H2O2,CF3COCF3, Na2HPO4, DCM, 42−94%; (c) R1 MgBr, CuI, THF, 0−25 °C, 59−83%; (d) 70% AcOH, 55 °C, 1−2 h, 46−96%.

Journal of Medicinal Chemistry Article

DOI: 10.1021/acs.jmedchem.8b00743J. Med. Chem. 2018, 61, 7767−7784

7776

Page 11: Discovery of a Potent and Selective Steroidal …...Discovery of a Potent and Selective Steroidal Glucocorticoid Receptor Antagonist (ORIC-101) Yosup Rew,* Xiaohui Du, John Eksterowicz,

the thioketal protecting group in 44 resulted in the finalcompounds 26, 27, and 28.

■ CONCLUSIONIn summary, structural modifications of mifepristone (1) at theC11 and C17 positions followed by reduction of the C9 C−Cdouble bond led to the discovery of 28 that features substantialreduction in AR agonism as compared to 1, with minimizedloss of GR antagonism potency and low potential of CYP2C8and 2C9 inhibition. Compound 28 demonstrated antitumoractivity by overcoming GR-mediated chemoresistance in theOVCAR5 ovarian cancer xenograft model and is currentlyundergoing clinical testing.71

■ EXPERIMENTAL SECTIONGeneral Chemistry. All reactions were conducted under an inert

gas atmosphere (nitrogen or argon) using a Teflon-coated magneticstir bar at the temperature indicated. Commercial reagents andanhydrous solvents were used without further purification. Analyticalthin layer chromatography (TLC) and flash chromatography wereperformed on Teledyne RediSep Rf Flash silica gel columns. Removalof solvents was conducted by using a rotary evaporator, and residualsolvent was removed from nonvolatile compounds using a vacuummanifold maintained at approximately 1 Torr. All yields reported areisolated yields. Preparative reversed-phase high pressure liquidchromatography (RP-HPLC) was performed using an Agilent 1100series HPLC and Phenomenex Gemini C18 column (5 μm, 100 mm× 21.2 mm i.d.), eluting with a binary solvent system A and B using agradient elusion [A, H2O with 0.1% trifluoroacetic acid (TFA); B,CH3CN with 0.1% TFA] with UV detection at 220 nm. All final

compounds were purified to ≥95% purity as determined by an Agilent1100 series HPLC with UV detection at 220 nm using the followingmethod: Phenomenex Gemini 5 μ C18 110A column (3.5 μm, 150mm × 4.6 mm i.d.); mobile phase, A = H2O with 0.1% TFA, B =CH3CN with 0.1% TFA; gradient: 5−95% B (0.0−15.0 min); flowrate, 1.5 mL/min. Low-resolution mass spectral (MS) data weredetermined on an Agilent 1100 series LCMS with UV detection at254 nm and a low resolution electrospray mode (ESI). NMR spectrawere obtained on a Bruker 400 spectrometer. Chemical shifts (δ) arereported in parts per million (ppm) relative to residual undeuteratedsolvent as an internal reference. The following abbreviations wereused to explain the multiplicities: s = single; d = doublet, t = triplet, q= quartet, dd = doublet of doublets, dt = doublet of triplets, m =multiplet, br = broad.

(8S,13S,14S,17S)-17-(3,3-Dimethylbut-1-yn-1-yl)-13-methyl-1,2,4,6,7,8,12,13,14,15,16,17-dodecahydrospiro[cyclopenta[a]-phenanthrene-3,2′-[1,3]dioxolan]-17-ol (30f). To a solution of 3,3-dimethylbut-1-yne (3.34 g, 40.7 mmol) in THF (85 mL) at −78 °Cwas added dropwise n-butyllithium (1.6 M in hexanes, 24.6 mL, 39.4mmol). After it was stirred at −78 °C for 25 min, a solution of 29 (8g, 25.4 mmol) in THF (100 mL) was added slowly. The mixture wasstirred at −78 °C for 17 min and then at rt for 40 min. The reactionmixture was quenched by slow addition of aq NH4Cl solution andconcentrated to remove the majority of THF. The residue wasextracted with EtOAc (400 mL). The organic phase was washed withbrine and dried over anhydrous Na2SO4. The mixture was thenconcentrated under reduced pressure, and the crude residue waspurified by silica gel chromatography (SiO2, 5−10% EtOAc inhexanes, gradient elution) to provide 30f (7.7 g, 18.9 mmol, 74%yield) as a white solid. 1H NMR (400 MHz, CDCl3) δ ppm 7.27 (2H, s), 5.51−5.69 (1 H, m), 3.92−4.09 (4 H, m), 2.64 (1 H, br dd, J =18.0 Hz), 2.53 (1 H, m), 2.29 (2 H, m), 1.90−2.11 (4 H, m), 1.62−1.87 (5 H, m), 1.70−1.75 (1 H, m), 1.40−1.50 (1 H, 1m), 1.20−1.30(1 H, m), 1.20 (9 H, s), 0.83 (3 H, s). m/z (ESI, +ve ion) 397.3 (M +H)+.

(5′R,8′S,10′R,13′S,14′S,17′S)-17′-(3,3-Dimethylbut-1-yn-1-yl)-13′-methyl-1′,2′,7′,8′,12′,13′,14′,15′,16′,17′-decahydro-4′H,6′H-spiro[[1,3]dioxolane-2,3′-[5,10]epoxycyclopenta[a]phenanthren]-17′-ol (31f). To a solution of 30f (7.5 g, 18.9 mmol) in DCM (120mL) at 0 °C was added Na2HPO4 (5.37 g, 37.8 mmol), followed byaddition of CF3COCF3-3H2O (2.6 mL, 18.9 mmol) and H2O2 (35%aq, 4.36 mL, 56.7 mmol). The mixture was stirred at 0 °C for 10 minand then at rt for 2 h. Aqueous Na2S2O3 solution was added, and themixture was stirred at rt for 30 min and extracted with EtOAc. Theorganic phase was washed with brine and dried over anhydrousNa2SO4. The mixture was then concentrated under reduced pressure,and the crude residue was purified by silica gel chromatography (SiO2,10−15% EtOAc in hexanes, gradient elution) to provide 31f (6.9 g,16.7 mmol, 88% yield) as a white solid. 1H NMR (400 MHz, CDCl3)δ ppm 6.07−6.11 (1 H, m), 3.87−3.99 (4 H, m), 2.45−2.65 (2 H, brs), 2.20−2.30 (2 H, m), 2.10 (1 H, m), 1.86−2.10 (4 H, m), 1.60−1.80 (5 H, m), 1.46−1.57 (1 H, m), 1.20−1.45 (2 H, m), 1.19 (9 H,s), 0.83 (3 H, s).

(5R,8S,11R,13S,14S,17S)-11-(4-(Dimethylamino)phenyl)-17-(3,3-dimethylbut-1-yn-1-yl)-13-methyl-1,2,6,7,8,11,12,13,14,15,16,17-dodecahydrospiro[cyclopenta[a]phenanthrene-3,2′-[1,3]-dioxolane]-5,17(4H)-diol (32j). A mixture of 31f (80 mg, 0.19 mmol)and copper(I) iodide (36.9 mg, 0.19 mmol) in THF (3 mL) wasdegassed with Ar. After cooling in an ice bath, (4-(dimethylamino)phenyl) magnesium bromide (0.5 M in THF, 3.1 mL, 1.55 mmol)was added dropwise. The resulting solution was maintained at rt for 1h, then quenched with NH4Cl solution and extracted with EtOAc.The organic phase was washed with brine and dried over anhydrousNa2SO4. The mixture was then concentrated under reduced pressureand the crude residue was purified by silica gel chromatography toprovide 32j (90 mg, 0.17 mmol, 89% yield) as a white solid. 1H NMR(400 MHz, CDCl3) δ ppm 7.07 (2 H, d, J = 8.5 Hz), 6.63−6.67 (2 H,m), 4.41 (1 H, d, J = 1.2 Hz), 4.13 (1 H, d, J = 7.2 Hz), 3.90−4.05 (4H, m), 2.91 (6 H, s), 2.45 (1 H, m), 2.10−2.40 (4 H, m), 2.00−2.05(2 H, m), 1.85−2.00 (2 H, m), 1.75−1.80 (1H, m), 1.60−1.75 (4 H,

Scheme 2a

aReagents and conditions: (a) (i) 34, CuI, THF, 0−25 °C, (ii) 70%AcOH, 55 °C, 1 h, 23% for two steps; (b) divinyl sulfone, IPA/H2O,115 °C, 5 h, 26%; (c) 2,2-dimethoxypropane, PTSA, DMF, 77%; (d)(i) 36, Mg, 60 °C, 30 min, CuI, THF, 0 to 25 °C, 98%, (ii) 70%AcOH, 60 °C, 1.5 h, 73%; (e) PPh3, DEAD, Ziram, THF, toluene,69%.

Journal of Medicinal Chemistry Article

DOI: 10.1021/acs.jmedchem.8b00743J. Med. Chem. 2018, 61, 7767−7784

7777

Page 12: Discovery of a Potent and Selective Steroidal …...Discovery of a Potent and Selective Steroidal Glucocorticoid Receptor Antagonist (ORIC-101) Yosup Rew,* Xiaohui Du, John Eksterowicz,

m), 1.50 (1 H, m), 1.30−1.40 (3 H, m), 1.24 (9 H, s), 0.49 (3 H, s).m/z (ESI, +ve ion) 534.3 (M + H)+.(8S,11R,13S,14S,17S)-11-(4-(Dimethylamino)phenyl)-17-(3,3-di-

m e t h y l b u t - 1 - y n - 1 - y l ) - 1 7 - h y d r o x y - 1 3 - m e t h y l -1,2,6,7,8,11,12,13,14,15,16,17-dodecahydro-3H-cyclopenta[a]-phenanthren-3-one (11). A solution of 32j (90 mg, 0.17 mmol) in1:2 acetic acid/water (16.5 mL) was stirred at 55 °C for 90 min. Thereaction mixture was cooled to rt and slowly added aq NaHCO3solution. After extraction with EtOAc, the organic phase was washedwith brine, dried over anhydrous Na2SO4, and concentrated underreduced pressure. The crude residue was purified by silica gelchromatography to provide 11 (54 mg, 0.12 mmol, 70% yield) as apale-yellow solid. 1H NMR (400 MHz, CDCl3) δ ppm 7.03 (2 H, d, J= 8.6 Hz), 6.67 (2 H, d, J = 8.6 Hz), 5.77 (1 H, s), 4.36 (1 H, br d, J =6.3 Hz), 2.93 (6 H, s), 2.76 (1 H, s), 2.59 (2 H, dd, J = 7.4, 3.1 Hz),2.27−2.49 (6 H, m), 2.17−2.25 (1 H, m), 1.86−2.06 (2 H, m), 1.64−1.78 (3 H, m), 1.47 (1 H, br s), 1.34 (1 H, br dd, J = 11.7, 5.6 Hz),1.20−1.29 (9 H, s), 0.56 (3 H, s). 13C NMR (101 MHz, CD2Cl2) δppm 199.5, 157.5, 149.2, 147.3, 132.9, 129.6, 128.1 (2 C), 122.9,113.1 (2 C), 95.5, 82.4, 80.2, 50.5, 47.5, 41.0 (2 C), 40.1, 39.8, 39.6,39.4, 37.5, 31.6, 31.4 (3 C), 28.1, 27.9, 26.4, 23.8, 14.1. m/z (ESI, +veion) 472.4 (M + H)+.2−10 and 12−23 were prepared from 29 according to a similar

procedure described for the synthesis of 11.(8S,11S,13S,14S,17S)-11-Ethyl-17-hydroxy-13-methyl-17-(prop-

1-yn-1-yl)-1,2,6,7,8,11,12,13,14,15,16,17-dodecahydro-3H-cyclopenta[a]phenanthren-3-one (2). 1H NMR (400 MHz, CDCl3)δ ppm 5.70 (1 H, s), 2.87−2.98 (2 H, m), 2.54−2.62 (1 H, m), 2.33−2.51 (5 H, m), 2.25 (1 H, ddd, J = 13.9, 9.5, 5.8 Hz), 1.78−2.06 (7 H,m), 1.45−1.72 (4 H, m), 1.20−1.43 (2 H, m) 1.05 (3 H, s), 0.95 (3H, t, J = 7.3 Hz). m/z (ESI, +ve ion) 339.1 (M + H)+.

(8S,11R,13S,14S,17S)-11-Cyclohexyl-17-hydroxy-13-methyl-17-(prop-1-yn-1-yl)-1,2,6,7,8,11,12,13,14,15,16,17-dodecahydro-3H-cyclopenta[a]phenanthren-3-one (3). 1H NMR (400 MHz, CDCl3)δ ppm 5.70 (1 H, s), 3.74−3.77 (1 H, m), 2.55−2.83 (3 H, m), 2.21−2.49 (6 H, m), 1.75−2.25 (7 H, m), 1.50−1.75 (3 H, m), 1.05−1.50(11 H, m), 0.71−0.92 (3 H, m).

(8S,11R,13S,14S,17S)-17-Hydroxy-13-methyl-11-phenyl-17-(prop-1-yn-1-yl)-1,2,6,7,8,11,12,13,14,15,16,17-dodecahydro-3H-cyclopenta[a]phenanthren-3-one (4). 1H NMR (400 MHz, CDCl3)δ ppm 7.22−7.35 (2 H, m), 7.13−7.22 (3 H, m), 5.77 (1 H, s), 4.44(1 H, br d, J = 4.0 Hz), 2.65−2.85 (1 H, m), 2.50−2.63 (2 H, m),2.13−2.50 (7 H, m), 1.90−2.15 (2 H, m), 1.90 (3 H, s), 1.62−1.88 (3H, m), 1.28−1.62 (2 H, m), 0.50 (3 H, s).

(8S,11R,13S,14S,17S)-11-(3-(Dimethylamino)phenyl)-17-hy-d r o x y - 1 3 - m e t h y l - 1 7 - ( p r o p - 1 - y n - 1 - y l ) -1,2,6,7,8,11,12,13,14,15,16,17-dodecahydro-3H-cyclopenta[a]-phenanthren-3-one (5). 1H NMR (400 MHz, CDCl3) δ ppm 7.12 (1H, m), 6.51−6.57 (3 H, m), 5.76 (1 H, s), 4.39 (1 H, br d, J = 4.0Hz), 2.91 (6 H, s), 2.70−2.85 (1 H, m), 2.18−2.65 (9 H, m), 1.91−2.18 (2 H, m), 1.90 (3 H, s), 1.70−1.85 (2 H, m), 1.28−1.65 (2 H,m), 0.58 (3 H, s); m/z (ESI, +ve ion) 430.4 (M + H)+.

(8S,11R,13S,14S,17S)-17-Hydroxy-11-(4-isopropylphenyl)-13-methyl-17-(prop-1-yn-1-yl)-1,2,6,7,8,11,12,13,14,15,16,17-dodeca-hydro-3H-cyclopenta[a]phenanthren-3-one (6). 1H NMR (400MHz, CDCl3) δ ppm 7.05−7.18 (4 H, m), 5.73 (1 H, s), 4.37 (1H, br d, J = 4.0 Hz), 2.71−2.89 (2 H, m), 2.15−2.68 (9 H, m), 1.88−2.15 (2 H, m), 1.88 (3 H, s), 1.20−1.81 (5 H, m), 1.25 (6 H, d, J =4.0 Hz), 0.58 (3 H, s). m/z (ESI, +ve ion) 429.4 (M + H)+.

(8S,11R,13S,14S,17S)-11-(4-(Dimethylamino)phenyl)-17-(hex-1-yn-1-yl)-17-hydroxy-13-methyl-1,2,6,7,8,11,12,13,14,15,16,17-do-decahydro-3H-cyclopenta[a]phenanthren-3-one (7). 1H NMR(400 MHz, CDCl3) δ ppm 7.02 (2 H, d, J = 8.2 Hz), 6.66 (2 H, d,

Scheme 3a

aReagents and conditions: (a) NaBH4, THF, MeOH, 0−25 °C, 100%; (b) 35% H2O2, CF3COCF3, Na2HPO4, DCM, 100%; (c) (i) ArMgBr, CuI,THF, 0−25 °C, 53−78%, (ii) 70% AcOH, 55 °C, 1 h, 77−80%; (d) liquid NH3, Li, THF, −78 °C, 74−100%; (e) for 41a, TsOH, benzene, reflux,46%; for 41b, 37% HCl, MeOH, 65 °C, 73%; (f) HSCH2CH2SH, TsOH, AcOH, 36−61%; (g) Al(OiPr)3, cyclohexanone, toluene, 105 °C, 60−67%; (h) n-BuLi, alkyne, −78 to 25 °C, 59−97%; (i) 4 N HCl, glyoxylic acid, AcOH, 38−43%.

Journal of Medicinal Chemistry Article

DOI: 10.1021/acs.jmedchem.8b00743J. Med. Chem. 2018, 61, 7767−7784

7778

Page 13: Discovery of a Potent and Selective Steroidal …...Discovery of a Potent and Selective Steroidal Glucocorticoid Receptor Antagonist (ORIC-101) Yosup Rew,* Xiaohui Du, John Eksterowicz,

J = 8.9 Hz), 5.76 (1 H, s), 4.35 (1 H, br d, J = 6.6 Hz), 2.92 (6 H, s),2.70−2.80 (1H, m), 2.45−2.60 (3 H, m), 2.30−2.48 (2 H, m), 2.12−2.28 (1 H, m), 1.95−2.10 (3 H, m), 1.50−1.62 (2 H, m), 1.32−1.46(5 H, m), 1.22−1.29 (3 H, m), 0.95−1.05 (2 H, m), 0.58 (3 H, s). m/z (ESI, +ve ion) 448.4 (M + H)+.(8S,11R,13S,14S,17S)-11-(4-(Dimethylamino)phenyl)-17-hy-

d roxy - 13 -me thy l - 17 - ( 3 , 3 , 3 - t r ifluo rop rop -1 - yn - 1 - y l ) -1,2,6,7,8,11,12,13,14,15,16,17-dodecahydro-3H-cyclopenta[a]-phenanthren-3-one (8). 1H NMR (400 MHz, CDCl3) δ ppm 7.02 (2H, m, J = 8.6 Hz), 6.67 (2 H, m, J = 8.4 Hz), 5.78 (1 H, s), 4.40 (1 H,br d, J = 6.8 Hz), 2.93 (6 H, s), 2.77 (1 H, m), 2.55−2.62 (2 H, m),2.44−2.54 (2 H, m), 2.29−2.40 (4 H, m), 2.23 (1 H, br d, J = 7.2Hz), 1.83−2.11 (4 H, m), 1.35−1.57 (3 H, m), 0.59 (3 H, s). m/z(ESI, +ve ion) 484.3 (M + H)+.(8S,11R,13S,14S,17S)-17-(But-1-yn-1-yl)-11-(4-(dimethylamino)-

phenyl)-17-hydroxy-13-methyl-1,2,6,7,8,11,12,13,14,15,16,17-do-decahydro-3H-cyclopenta[a]phenanthren-3-one (9). 1H NMR(400 MHz, CDCl3) δ ppm 7.15−7.22 (1 H, m), 7.31 (1 H, m),6.75−6.80 (1 H, m), 6.65 (1 H, m), 5.77 (1 H, s), 4.37 (1 H, m),3.05−3.15 (4 H, m), 3.0 (1 H, m), 2.93 (6 H, s), 2.55−2.62 (1 H, m),2.30−2.45 (4 H, m), 2.00−2.10 (1 H, m), 1.90−2.00 (1 H, m), 1.65−1.75 (3 H, m), 1.30−1.40 (2 H, m), 1.25 (1 H, m), 1.17 (3 H, t, J =7.5 Hz), 0.56 (3 H, s). m/z (ESI, +ve ion) 444.4 (M + H)+.(8S,11R,13S,14S,17S)-11-(4-(Dimethylamino)phenyl)-17-hy-

d r o x y - 1 3 - m e t h y l - 1 7 - ( 3 - m e t h y l b u t - 1 - y n - 1 - y l ) -1,2,6,7,8,11,12,13,14,15,16,17-dodecahydro-3H-cyclopenta[a]-phenanthren-3-one (10). 1H NMR (400 MHz, CDCl3) δ ppm 7.02(2 H, m, J = 8.0 Hz), 6.66 (2 H, m, J = 8.0 Hz), 5.57 (1 H, s), 4.35 (1H, br d, J = 8.0 Hz), 2.92 (6 H, s), 2.70−2.80 (1 H, m), 2.50−2.70 (2H, m), 2.15−2.50 (7 H, m), 1.85−2.15 (2 H, m), 1.65−1.85 (4 H,m), 1.20−1.60 (2 H, m), 1.17 (6 H, d, J = 4.0 Hz), 0.56 (3 H, s); m/z(ESI, +ve ion) 458.3 (M + H)+.(8S,11R,13S,14S,17S)-17-(3,3-Dimethylbut-1-yn-1-yl)-17-hy-

droxy-13-methyl-11-(p-tolyl)-1,2,6,7,8,11,12,13,14,15,16,17-dodec-ahydro-3H-cyclopenta[a]phenanthren-3-one (12). 1H NMR (400MHz, CDCl3) δ ppm 7.01−7.15 (4 H, m), 5.78 (1 H, s), 4.40 (1 H,br d, J = 7.0 Hz), 2.76 (1 H, br d, J = 14.8 Hz), 2.56−2.63 (2 H, m),2.40−2.49 (1 H, m), 2.18−2.38 (9 H, m), 1.87−2.12 (2 H, m), 1.61−1.80 (3 H, m), 1.42−1.56 (1 H, m), 1.35 (1 H, br dd, J = 11.7, 5.9Hz), 1.25 (9 H, s), 0.53 (3 H, s). m/z (ESI, +ve ion) 443.5 (M + H)+.(8S,11R,13S,14S,17S)-17-(3,3-Dimethylbut-1-yn-1-yl)-11-(4-eth-

ylphenyl)-17-hydroxy-13-methyl-1,2,6,7,8,11,12,13,14,15,16,17-do-decahydro-3H-cyclopenta[a]phenanthren-3-one (13). 1H NMR(400 MHz, CDCl3) δ ppm 7.10 (4 H, s), 5.78 (1 H, s), 4.41 (1 H,br d, J = 6.3 Hz), 2.77 (1 H, br d, J = 14.5 Hz), 2.56−2.66 (4 H, m),2.28−2.50 (6 H, m), 2.18−2.26 (1 H, m), 1.89−2.12 (2 H, m), 1.60−1.79 (3 H, m), 1.43−1.57 (1 H, m), 1.29−1.42 (1 H, m), 1.14−1.29(12 H, m), 0.52 (3 H, s); m/z (ESI, +ve ion) 457.4 (M + H)+.(8S,11R,13S,14S,17S)-17-(3,3-Dimethylbut-1-yn-1-yl)-17-hy-

d r o x y - 1 1 - ( 4 - i s o p r o p y l p h e n y l ) - 1 3 - m e t h y l -1,2,6,7,8,11,12,13,14,15,16,17-dodecahydro-3H-cyclopenta[a]-phenanthren-3-one (14). 1H NMR (400 MHz, CDCl3) δ ppm 7.04−7.20 (4 H, m), 5.78 (1 H, s), 4.41 (1 H, br d, J = 6.4 Hz), 2.87 (1 H, t,J = 6.9 Hz), 2.75 (1 H, m), 2.54−2.62 (2 H, m), 2.27−2.50 (6 H, m),2.21 (1 H, m), 2.01 (1 H, m), 1.95 (1 H, m), 1.59−1.82 (3 H, m),1.28−1.45 (2 H, m), 1.17−1.27 (15 H, m), 0.52 (3 H, s). m/z (ESI,+ve ion) 471.4 (M + H)+.(8S,11R,13S,14S,17S)-17-(3,3-Dimethylbut-1-yn-1-yl)-17-hy-

d r o x y - 1 1 - ( 4 - m e t h o x y p h e n y l ) - 1 3 - m e t h y l -1,2,6,7,8,11,12,13,14,15,16,17-dodecahydro-3H-cyclopenta[a]-phenanthren-3-one (15). 1H NMR (400 MHz, CDCl3) δ ppm 7.09(2 H, d, J = 8.4 Hz), 7.00 (1 H, d, J = 8.8 Hz), 5.78 (1 H, s), 4.39 (1H, br d, J = 7.8 Hz), 3.79 (3 H, s) 2.77 (1 H, br d, J = 14.6 Hz), 2.54−2.67 (2 H, m) 2.27−2.53 (6 H, m), 2.18−2.25 (1 H, m),1.95 (1 H, brd, J = 1.0 Hz) 1.59−1.85 (3 H, m) 1.30−1.56 (2 H, m), 1.25 (9 H, s),0.53 (3 H, s). m/z (ESI, +ve ion) 459.4 (M + H)+.(8S,11R,13S,14S,17S)-11-(3-(Dimethylamino)phenyl)-17-(3,3-di-

m e t h y l b u t - 1 - y n - 1 - y l ) - 1 7 - h y d r o x y - 1 3 - m e t h y l -1,2,6,7,8,11,12,13,14,15,16,17-dodecahydro-3H-cyclopenta[a]-phenanthren-3-one (16). 1H NMR (400 MHz, CDCl3) δ ppm 7.13(1 H, t, J = 7.9 Hz), 6.53−6.60 (3 H, m), 5.77 (1 H, s), 4.40 (1 H, brs), 2.93 (6 H, s), 2.73−2.82 (1 H, m), 2.53−2.62 (2 H, m), 2.31−2.52

(6 H, m), 2.18−2.26 (1 H, m), 1.92−2.06 (2 H, m), 1.60−1.79 (3 H,m), 1.49 (1 H, br d, J = 6.3 Hz), 1.31−1.44 (1 H, m), 1.25 (9 H, s),0.59 (3 H, s). m/z (ESI, +ve ion) 472.4 (M + H)+.

(8S,11R,13S,14S,17S)-11-(4-(Dimethylamino)-3,5-difluorophen-yl)-17-(3,3-dimethylbut-1-yn-1-yl)-17-hydroxy-13-methyl-1,2,6,7,8,11,12,13,14,15,16,17-dodecahydro-3H-cyclopenta[a]-phenanthren-3-one (17). 1H NMR (400 MHz, CDCl3) δ ppm 6.68(1 H, s), 6.65 (1 H, s), 5.81 (1 H, s), 4.32 (1 H, br d, J = 6.8 Hz), 2.87(6 H, t, J = 1.6 Hz), 2.75 (1 H, br d, J = 14.3 Hz), 2.56−2.62 (2 H,m), 2.29−2.48 (5 H, m), 2.15−2.26 (2 H, m), 1.89−2.08 (2 H, m),1.61−1.80 (3 H, m), 1.43−1.57 (1 H, m), 1.29−1.43 (1 H, m), 1.24(9 H, s), 0.55 (3 H, s). m/z (ESI, +ve ion) 508.4 (M + H)+.

(8S,11S,13S,14S,17S)-11-(4-(Dimethylamino)-2,6-difluorophen-yl)-17-(3,3-dimethylbut-1-yn-1-yl)-17-hydroxy-13-methyl-1,2,6,7,8,11,12,13,14,15,16,17-dodecahydro-3H-cyclopenta[a]-phenanthren-3-one (18). 1H NMR (400 MHz, CDCl3) δ ppm 6.01−6.24 (2 H, m), 5.70 (1 H, s), 4.52 (1 H, d, J = 8.4 Hz), 2.92 (6 H, s),2.69 (2 H, dt, J = 15.0, 5.3 Hz), 2.39−2.59 (3 H, m), 2.16−2.38 (4 H,m), 1.87−2.06 (3 H, m), 1.70−1.84 (1 H, m), 1.33−1.57 (3 H, m),1.24 (9 H, s), 1 0.84 (3 H, s). m/z (ESI, +ve ion) 508.4 (M + H)+.

(8S,11R,13S,14S,17S)-11-(4-(Dimethylamino)-3-methoxyphen-yl)-17-(3,3-dimethylbut-1-yn-1-yl)-17-hydroxy-13-methyl-1,2,6,7,8,11,12,13,14,15,16,17-dodecahydro-3H-cyclopenta[a]-phenanthren-3-one (19). 1H NMR (400 MHz, CDCl3) δ ppm 6.81(1 H, d, J = 8.2 Hz), 6.73 (1 H, s), 6.64 (1 H, d, J = 7.6 Hz), 5.78 (1H, s), 4.39 (1 H, br d, J = 6.4 Hz), 3.86 (3 H, s), 2.71−2.81 (7 H, m),2.53−2.64 (2 H, m), 2.30−2.50 (6 H, m), 2.18−2.28 (1 H, m), 1.88−2.12 (2 H, m), 1.61−1.79 (4 H, m), 1.31−1.56 (2 H, m), 1.20−1.28(9 H, s), 0.56 (3 H, s). m/z (ESI, +ve ion) 502.4 (M + H)+.

(8S,11R,13S,14S,17S)-11-(4-(Dimethylamino)-3-methylphenyl)-17-(3 ,3-dimethylbut-1-yn-1-y l ) -17-hydroxy-13-methyl -1,2,6,7,8,11,12,13,14,15,16,17-dodecahydro-3H-cyclopenta[a]-phenanthren-3-one (20). 1H NMR (400 MHz, CDCl3) δ ppm 6.99(1 H, s), 6.89 (2 H, m), 5.78 (1 H, s), 4.33−4.38 (1 H, m), 2.75 (1 H,m), 2.67 (6 H, s), 2.54−2.64 (2 H, m), 2.26−2.47 (8 H, m), 2.18−2.24 (1 H, m), 1.88−2.07 (2 H, m), 1.60−1.77 (3 H, m), 1.47 (1 H,br dd, J = 12.9, 9.4 Hz), 1.31−1.40 (1 H, m), 1.24 (9 H, s), 0.54 (3 H,s). m/z (ESI, +ve ion) 486.4 (M + H)+.

(8S,11R,13S,14S,17S)-17-(3,3-Dimethylbut-1-yn-1-yl)-17-hy-droxy-11-(4-(isopropyl(methyl)amino)phenyl)-13-methyl-1,2,6,7,8,11,12,13,14,15,16,17-dodecahydro-3H-cyclopenta[a]-phenanthren-3-one (21). 1H NMR (400 MHz, CDCl3) δ ppm 6.95−7.06 (2 H, m), 6.71 (2 H, m, J = 8.8 Hz), 5.77 (1 H, s), 4.35 (1 H, brd, J = 5.7 Hz), 3.98−4.09 (1 H, m), 2.76 (1 H, m), 2.70 (3 H, s),2.53−2.61 (2 H, m), 2.33−2.49 (4 H, m), 2.27−2.30 (1 H, m), 2.17−2.25 (1 H, m), 1.84−2.09 (2 H, m), 1.54−1.80 (3 H, m), 1.30−1.54(2 H, m), 1.24 (9 H, s), 1.10−1.38 6 H, m), 0.56 (3 H, s). m/z (ESI,+ve ion) 500.5 (M + H)+.

(8S,11R,13S,14S,17S)-17-(3,3-Dimethylbut-1-yn-1-yl)-17-hy-d r o x y - 1 3 - m e t h y l - 1 1 - ( 4 - m o r p h o l i n o p h e n y l ) -1,2,6,7,8,11,12,13,14,15,16,17-dodecahydro-3H-cyclopenta[a]-phenanthren-3-one (22). 1H NMR (400 MHz, CDCl3) δ ppm 7.03−7.13 (2 H, m), 6.75−6.91 (2 H, m), 5.78 (1 H, s), 4.34−4.40 (1 H,m), 3.79−3.90 (4 H, m), 3.08−3.18 (4 H, m), 2.76 (1 H, br d, J =15.0 Hz), 2.54−2.62 (2 H, m), 2.27−2.49 (5 H, m), 2.18−2.25 (1 H,m), 1.90−2.07 (2 H, m), 1.64−1.78 (3 H, m), 1.48 (2 H, br d, J = 9.8Hz), 1.24 (9 H, s), 0.53 (3 H, s). m/z (ESI, +ve ion) 514.4 (M + H)+.

(8S,11R,13S,14S,17S)-17-(3,3-Dimethylbut-1-yn-1-yl)-17-hy-d r o x y - 1 3 -m e t h y l - 1 1 - ( 4 - ( p y r r o l i d i n - 1 - y l ) p h e n y l ) -1,2,6,7,8,11,12,13,14,15,16,17-dodecahydro-3H-cyclopenta[a]-phenanthren-3-one (23). 1H NMR (400 MHz, CDCl3) δ ppm 6.98−7.06 (2 H, m), 6.44−6.54 (2 H, m) 5.76 (1 H, s), 4.36 (1 H, br dd, J= 5.6, 2.4 Hz), 3.27 (4 H, br t, J = 5.2 Hz), 2.73−2.81 (1 H, m),2.53−2.61 (2 H, m), 2.26−2.48 (5 H, m), 2.17−2.24 (1 H, m), 1.91−2.06 (6 H, m), 1.63−1.77 (3 H, m), 1.42−1.56 (1 H, m), 1.30−1.41(1 H, m), 1.24 (9 H, s), 0.57 (3 H, s). m/z (ESI, +ve ion) 498.4 (M +H)+.

(8S,11R,13S,14S,17S)-11-(4-Aminophenyl)-17-(3,3-dimethylbut-1-yn-1-yl)-17-hydroxy-13-methyl-1,2,6,7,8,11,12,13,14,15,16,17-do-decahydro-3H-cyclopenta[a]phenanthren-3-one (33a). Under Ar,31f (3 g, 7.27 mmol) and copper iodide (1.38 g, 7.27 mmol) in THF(40 mL) was cooled to 0 °C. (4-(Bis(trimethylsilyl)amino)phenyl)

Journal of Medicinal Chemistry Article

DOI: 10.1021/acs.jmedchem.8b00743J. Med. Chem. 2018, 61, 7767−7784

7779

Page 14: Discovery of a Potent and Selective Steroidal …...Discovery of a Potent and Selective Steroidal Glucocorticoid Receptor Antagonist (ORIC-101) Yosup Rew,* Xiaohui Du, John Eksterowicz,

magnesium bromide (34) (0.5 M in THF, 58.2 mL, 29.1 mmol) wasadded dropwise. The reaction mixture was slowly brought to rt andstirred for 2 h. The reaction was quenched (satd NH4Cl), and thereaction mixture exposed to the air and stirred for 10 min. Themixture was extracted (EtOAc) and washed (brine). The organicswere separated and dried (MgSO4) before concentration to dryness.The crude was used in next step without further purification.A solution of the crude material (641 mg g, 0.99 mmol) in 70%

acetic acid in water (5 mL) was stirred at 55 °C for 1 h. The reactionwas quenched (satd aq NaHCO3), extracted (2 × EtOAc), andwashed (brine). The combined organic layers were dried (MgSO4)and concentrated under reduced pressure. Purification of the residueby column chromatography (SiO2, 50−70% EtOAc in hexanes,gradient elution) gave 33a (100 mg, 0.23 mmol, 22% yield). 1H NMR(400 MHz, CDCl3) δ ppm 6.96 (2 H, d, J = 8.5 Hz), 6.64 (2 H, br d, J= 8.2 Hz), 5.77 (1 H, s), 4.34 (1 H, br d, J = 6.9 Hz), 2.69−2.86 (1 H,m), 2.52−2.65 (2 H, m), 2.28−2.49 (5 H, m), 2.17−2.26 (2 H, m),1.85−2.09 (2 H, m), 1.62−1.85 (3 H, m), 1.25−1.51 (2 H, m), 1.24(9 H, s), 0.506 (3 H, s). m/z (ESI, +ve ion) 444.4 [M + H]+.(8S,11R,13S,14S,17S)-17-(3,3-Dimethylbut-1-yn-1-yl)-11-(4-(1,1-

dioxidothiomorphol ino)phenyl)-17-hydroxy-13-methyl-1,2,6,7,8,11,12,13,14,15,16,17-dodecahydro-3H-cyclopenta[a]-phenanthren-3-one (24). Divinyl sulfone (0.20 mL, 1.4 mmol) wasadded to a solution of 33a (60 mg, 0.14 mmol) in isopropyl alcohol(2 mL) and water (2 mL) and the mixture was refluxed at 115 °C for5 h. The reaction was quenched (satd aq NH4Cl), extracted (2 ×EtOAc), and washed (brine). The combined organic layers were dried(MgSO4) and concentrated under reduced pressure. Purification ofthe crude product by column chromatography (SiO2, 20−60% EtOAcin hexanes, gradient elution) gave 24 (20 mg, 0.036 mmol, 26%yield). 1H NMR (400 MHz, CDCl3) δ ppm 7.11 (2 H, d, J = 8.5 Hz),6.78−6.93 (2 H, m), 5.79 (1 H, s), 4.38 (1 H, br d, J = 7.3 Hz), 3.76−3.93 (4 H, m), 3.05−3.20 (4 H, m), 2.72−2.83 (1 H, m), 2.54−2.66(2 H, m), 2.18−2.48 (7 H, m), 1.89−2.11 (3 H, m), 1.57−1.83 (1 H,m), 1.43−1.56 (2 H, m), 1.24 (9 H, s), 0.53 (3 H, s). m/z (ESI, +veion) 562.4 [M + H]+.5-(4-Bromophenyl)-2,2,5-trimethyl-1,3-dioxane (36). To a sol-

ution of 2-(4-bromophenyl)-2-methylpropane-1,3-diol (35) (688 mg,2.81 mmol) and 4-methylbenzenesulfonic acid (24 mg, 0.14 mmol) inDMF (12 mL) was added 2,2-dimethoxypropane (0.52 mL, 4.2mmol). The solution was stirred at rt overnight and then was pouredinto saturated aq NaHCO3 and extracted with EtOAc. The combinedorganic extracts were purified by column chromatography (SiO2, 0−30% EtOAc in hexanes, gradient elution) to give 36 (618 mg, 2.22mmol, 77% yield). 1H NMR (400 MHz, CDCl3) δ ppm 7.49 (2 H, d,J = 7.42 Hz) 7.26−7.30 (2 H, m) 4.08 (2 H, d, J = 11.7 Hz) 3.80 (2H, d, J = 11.7 Hz) 1.41 (3 H, s) 1.34 (3 H, s), 1.28 (3 H, s); m/z(ESI, +ve ion) 280.3 [M + H]+.(8S,11R,13S,14S,17S)-11-(4-(1,3-Dihydroxy-2-methylpropan-2-

yl)phenyl)-17-(3,3-dimethylbut-1-yn-1-yl)-17-hydroxy-13-methyl-1,2,6,7,8,11,12,13,14,15,16,17-dodecahydro-3H-cyclopenta[a]-phenanthren-3-one (33b). A dried flask was charged withmagnesium chips (40 mg, 1.5 mmol) and a crystal of iodine. Theflask was flushed with Ar, heated briefly with a heat gun, and thenheated at 60 °C. At first, a small portion (0.5 mL) of a solution of 36(azeotroped with toluene beforehand, 373 mg, 1.31 mmol) in THF (2mL) was added. After iodine color disappeared, the remainingsolution was added, and the mixture was continued stirring for 30 minat 60 °C. The mixture was then cooled down and added to a solutionof 31f (45 mg, 0.11 mmol) and copper(I) iodide (16.6 mg, 0.09mmol) in THF (1 mL) at 0 °C. The mixture was kept at 0 °C for 10min and then raised to rt for 2 h. After the reaction was cooled to rt, itwas quenched with NH4Cl and extracted with EtOAc. The organicswere washed, dried, and concentrated. The residue was purified byCombi-flash (SiO2, 0−40% EtOAc in hexanes, gradient elution) togive the Grignard addition product (55 mg, 81%).To a flask charged the Grignard addition product (55 mg, 0.09

mmol) was added 70% acetic acid (2 mL). After the mixture washeated at 60 °C for 2 h, it was quenched with NaHCO3 and extractedwith EtOAc. The organics were washed, dried, and concentrated. The

residue was purified by Combi-flash (SiO2, 0−80% EtOAc in hexanes,gradient elution) to give 33b (32 mg, 0.06 mmol, 70% yield). 1HNMR (400 MHz, CDCl3 δ ppm 7.28−7.37 (2 H, m), 7.19 (2 H, d, J= 8.2 Hz), 5.78 (1 H, s), 4.43 (1 H, br d, J = 6.6 Hz), 3.96 (2 H, d, J =11.0 Hz), 3.83 (2 H, dd, J = 11.0, 1.2 Hz), 2.69−2.87 (1 H, m), 2.59(2 H, br dd, J = 7.1, 3.7 Hz), 2.23−2.50 (7 H, m), 1.85−2.11 (2 H,m), 1.75−1.84 (2 H, m), 1.45−1.55 (1 H, m), 1.25−1.40 (1 H, m),1.30 (3 H, s), 1.25 (9 H, s), 0.50 (3 H, s). m/z (ESI, +ve ion) 517.4(M + H)+.

(8S,11R,13S,14S,17S)-17-(3,3-Dimethylbut-1-yn-1-yl)-17-hy-droxy-13-methy l -11- (4- (3-methy loxetan-3-y l )pheny l ) -1,2,6,7,8,11,12,13,14,15,16,17-dodecahydro-3H-cyclopenta[a]-phenanthren-3-one (25). A flask was charged with 33b (33 mg, 0.07mmol) and azeotroped with toluene. Triphenylphosphine (50.3 mg,0.19 mmol), toluene (1.5 mL), THF (0.5 mL), and Ziram (39.1 mg,0.13 mmol) were added successively. Upon adding ethyl N,N-ethoxycarbonyliminocarbamate (0.03 mL, 0.19 mmol), the solutionbecame clear. The resulting solution was stirred overnight. Themixture was filtered through Celite and rinsed with EtOAc. Thefiltrate was concentrated, and the residue was purified by Combi-flash(SiO2, 0−30% EtOAc in hexanes, gradient elution) to give the crudeproduct. Purification by RP-HPLC (30−70% A/B, gradient elution)provided 25 (17 mg, 0.034 mmol, 53% yield). 1H NMR (400 MHz,CDCl3) δ ppm 7.13−7.19 (4 H, m), 5.79 (1 H, s), 4.94 (2 H, dd, J =5.6, 3.4 Hz), 4.63 (2 H, d, J = 5.6 Hz), 4.43 (1 H, br d, J = 6.7 Hz),2.77 (1 H, br d, J = 14.6 Hz), 2.60 (2 H, dd, J = 8.0, 4.1 Hz), 2.26−2.51 (6 H, m), 2.15−2.25 (1 H, m), 1.87−2.09 (2 H, m), 1.65−1.79(5 H, m), 1.43−1.56 (2 H, m), 1.25 (9 H, s), 0.52 (3 H, s). m/z (ESI,+ve ion) 499.4 (M + H)+.

(8S,13S,14S,17S)-13-Methyl-1,2,4,6,7,8,12,13,14,15,16,17-dodecahydrospiro[cyclopenta[a]phenanthrene-3,2′-[1,3]-dioxolan]-17-ol (37). To a solution of 29 (100 g, 318 mmol) in THF(500 mL) and MeOH (50 mL) was added sodium borohydride (6.14g, 159 mmol) in portions at 0 °C. The mixture was stirred at 0 °C for30 min and then rt for 1 h. The reaction was quenched with 2 mL ofsatd NaHCO3 solution and concentrated to remove MeOH. Theresidue was dissolved in EtOAc (1 L), the organic phase was washedwith satd NaHCO3 solution and then brine and dried over anhydrousNa2SO4. The organic solution was concentrated under reducedpressure to give 37 (99.5 g, 100% yield) as a colorless oil. 1H NMR(400 MHz, CDCl3) δ ppm 5.61 (1 H, m), 4.01 (4 H, s), 3.78 (1 H,m), 2.48−2.64 (1 H, m), 1.75−2.36 (14 H, m), 1.12−1.58 (3 H, m),0.78 (3 H, s). m/z (ESI, +ve ion) 317.3 [M + H]+.

( 5 ′ R , 8 ′ S , 1 0 ′ R , 1 3 ′ S , 1 4 ′ S , 1 7 ′ S ) - 1 3 ′ - M e t h y l -1′,2′,7′,8′,12′,13′,14′,15′,16′,17′-decahydro-4′H,6′H-spiro[[1,3]-dioxolane-2,3′-[5,10]epoxycyclopenta[a]phenanthren]-17′-ol (38).To a solution of 37 (43.0 g, 136 mmol) in DCM (320 mL) at 0 °Cwas added Na2HPO4 (9.65 g, 67.9 mmol), followed by addition of1,1,1,3,3,3-hexafluoropropan-2-one trihydrate (11.2 mL, 81.5 mmol)and H2O2 (35% aq, 34.2 mL, 408 mmol). The mixture was stirred at 0°C for 10 min and then at rt for 3 h. Sodium thiosulfate solution (10%aq, 200 mL) was slowly added at 0 °C, and the mixture was stirred atrt for 30 min and extracted with DCM (300 mL). The organic phasewas washed with brine and dried over anhydrous Na2SO4. Theorganic solution was then concentrated to provide 38 (43 g. 130mmol, 95% yield) as a 4:1 mixture of epimers (the desired isomer ismajor). 1H NMR (400 MHz, CDCl3) δ ppm 5.97−6.07 (1 H, m),3.84−4.00 (4 H, m), 3.65−3.79 (1 H, m), 2.38−2.57 (1 H, m), 1.75−2.36 (14 H, m), 1.12−1.58 (3 H, m), 0.74 (3 H, s).

(8S,11R,13S,14S,17S)-17-Hydroxy-11-(4-(isopropyl(methyl)-amino)phenyl)-13-methyl-1,2,6,7,8,11,12,13,14,15,16,17-dodeca-hydro-3H-cyclopenta[a]phenanthren-3-one (39b). A dried flask wascharged with magnesium chips (2.83 g, 117 mmol) and a crystal ofiodine. The flask was flushed with Ar and heated briefly with a heatgun and then put into a 60 °C of bath. A 10 mL of 4-bromo-N-isopropyl-N-methyl-aniline (26.1 g, 115 mmol, azeotroped withtoluene beforehand) in THF (120 mL) was added. After the iodinecolor disappeared, the remaining solution was added dropwise for 20min and the mixture was continued to stir at 6 °C for 2 h.

The generated Grignard reagent prepared above was cooled downand added to a solution of 38 (12.7 g, 38.2 mmol, azeotroped with

Journal of Medicinal Chemistry Article

DOI: 10.1021/acs.jmedchem.8b00743J. Med. Chem. 2018, 61, 7767−7784

7780

Page 15: Discovery of a Potent and Selective Steroidal …...Discovery of a Potent and Selective Steroidal Glucocorticoid Receptor Antagonist (ORIC-101) Yosup Rew,* Xiaohui Du, John Eksterowicz,

toluene) and copper(I) iodide (3.64 mg, 19.1 mmol) in THF (100mL) at 0 °C. The resulting mixture was allowed to warm up to rt andstir for 1 h. The reaction was quenched (satd aq NH4Cl), extracted (2× EtOAc), and washed (brine). The combined organic layer wasdried (Na2SO4) and concentrated under reduced pressure.Purification of the residue by Combi-flash (SiO2, 45−65% EtOAc/hexanes, gradient elution) provided the intermediate (9.7 g, 20.1mmol, 53% yield) as an off-white foam.A solution of the intermediate (9.7 g, 20.1 mmol) in 70% acetic

acid in water (80 mL, 1.33 mol) was stirred at 55 °C for 1 h. Thereaction was concentrated under reduced pressure to remove mostAcOH. Then the residue was diluted (EtOAc), basified (satd aqNaHCO3), extracted (2 × EtOAc), and washed (brine). Thecombined organic layer was dried (Na2SO4) and concentratedunder reduced pressure. Purification of the residue by Combi-flash(SiO2, 50−100% EtOAc in hexanes, gradient elution) gave 39b (6.80g, 16.2 mmol, 80% yield) as a white solid. 1H NMR (400 MHz,CDCl3) δ ppm 7.01 (2 H, d, J = 8.5 Hz), 6.71 (2 H, d, J = 8.9 Hz),5.76 (1 H, s), 4.29 (1 H, br d, J = 6.9 Hz), 4.06 (1 H, dt, J = 13.3, 6.6Hz), 3.62−3.73 (1 H, m), 2.73 (1 H, br s), 2.71 (3 H, s), 2.32−2.61(7 H, m), 2.07−2.17 (1 H, m), 2.02 (1 H, dq, J = 12.9, 4.4 Hz), 1.63−1.76 (2 H, m), 1.28−1.49 (5 H, m), 1.15 (3 H, d, J = 6.6 Hz), 1.14 (3H, d, J = 6.6 Hz) 0.48 (3 H, s). m/z (ESI, +ve ion) 420.4 [M + H]+.(8S,9R,11S,13S,14S,17S)-17-Hydroxy-11-(4-(isopropyl(methyl)-

amino)phenyl)-13-methyl-1,2,4,6,7,8,9,11,12,13,14,15,16,17-tetra-decahydro-3H-cyclopenta[a]phenanthren-3-one (40b). Under Ar,lithium (2.03 g, 292 mmol) in pieces was added to liquid ammonia(500 mL) at −78 °C; when the color changed to dark blue, a solutionof 39b (50.0 g, 119 mmol) in THF (500 mL) was added over 15 min(kept the internal temperature below −60 °C). The mixture wasstirred at −78 °C for 5 min before the reaction was quenched byadding 10 mL of H2O, then the solution was poured into 500 mLice−water, extracted by EtOAc (500 mL × 2), and the combinedorganic layer was dried and concentrated under reduced pressure.Purification of the residue by Combi-flash (SiO2, EtOAc/DCM/hex =1:1:10 to1:1:5, gradient elution) provided 40b (37.1 g, 74% yield) aswhite foam. 1H NMR (400 MHz, CDCl3) δ ppm 7.17 (2 H, d, J = 8.6Hz), 6.65 (2 H, d, J = 8.9 Hz), 4.04 (1 H, dt, J = 13.2, 6.6 Hz), 3.60−3.71 (1 H, m), 3.44 (1 H, br s), 2.81 (2 H, br s), 2.68 (3 H, s), 2.07−2.45 (8 H, m), 1.98 (1 H, br d, J = 17.5 Hz), 1.82−1.91 (1 H, m),1.60−1.71 (2 H, m), 1.20−1.45 (5 H, m), 1.14 (3 H, d, J = 6.6 Hz),1.13 (3 H, d, J = 6.6 Hz), 0.36 (3 H, s). m/z (ESI, +ve ion) 422.4 [M+ H]+.(8R,9S,10R,11S,13S,14S,17S)-17-Hydroxy-11-(4-(isopropyl-

( m e t h y l ) a m i n o ) p h e n y l ) - 1 3 - m e t h y l -1,2,6,7,8,9,10,11,12,13,14,15,16,17-tetradecahydro-3H-cyclopenta-[a]phenanthren-3-one (41b). Under Ar, to a solution of 40b (11.3 g,26.8 mmol) in anhydrous EtOH (68 mL) was added conchydrochloric acid (90 mL), and the resulting solution was stirred at65 °C until most starting material disappeared (∼16 h). The reactionwas cooled to rt and concentrated under reduced pressure to removemost HCl. Then the residue was diluted with DCM, basified with ice-cold 2.5 M NaOH, and extracted (2 × DCM). The combined organiclayer was washed with brine, dried over Na2SO4, and concentratedunder reduced pressure. Purification of the residue by Combi-flash(SiO2, 25−50% EtOAc in hexanes, gradient elution) provided 41b(8.90 g, 21.1 mmol, 78% yield) as an off-white foam. 1H NMR (400MHz, CDCl3) δ ppm 7.24 (2 H, br d, J = 8.6 Hz), 6.71 (2 H, br d, J =8.5 Hz), 5.85 (1 H, s), 4.02−4.12 (1 H, m), 3.53−3.63 (1 H, m), 3.25(1 H, br t, J = 5.3 Hz), 2.83 (1 H, br s), 2.72 (3 H, s), 2.53 (1 H, br d,J = 14.3 Hz), 2.37 (1 H, td, J = 14.1, 5.0 Hz), 2.26 (1 H, dt, J = 16.3,3.9 Hz), 2.18 (1 H, dd, J = 13.1, 1.4 Hz), 2.07−2.14 (2 H, m), 2.01−2.05 (1 H, m), 1.86−1.96 (1 H, m), 1.65−1.74 (1 H, m), 1.60−1.65(1 H, m), 1.27−1.51 (1 H, m), 1.27−1.51 (5 H, m), 1.18 (3 H, d, J =6.4 Hz), 1.16 (3 H, d, J = 6.4 Hz), 0.98−1.12 (2 H, m), 0.59 (3 H, s).m/z (ESI, +ve ion) 422.3 [M + H]+.(8R,9S,10R,11S,13S,14S)-11-(4-(Isopropyl(methyl)amino)phenyl)-

13-methyl-1,2,6,7,8,9,10,11,12,13,14,15,16,17-tetradecahydrospiro-[cyclopenta[a]phenanthrene-3,2′-[1,3]dithiolan]-17-ol (42b). To asolution of 41b (2.80 g, 6.64 mmol) and p-toluenesulfonic acid

monohydrate (2.45 g, 12.9 mmol) in acetic acid (50 mL) was addedethane-1,2-dithiol (5.4 mL, 64 mmol). After the resulting solution wasstirred at rt under Ar for 2 h, the reaction was quenched (ice-cold 2.5N aq NaOH) and extracted (2 × EtOAc). The combined organiclayer was washed (brine), dried (MgSO4), and concentrated underreduced pressure. Purification of the residue by Combi-flash (SiO2,30−60% EtOAc in hexanes, gradient elution) provided 42b (1.17 g,2.35 mmol, 36% yield) as a white solid. 1H NMR (400 MHz, CDCl3)δ ppm 7.23 (2 H, d, J = 8.8 Hz), 6.69 (2 H, d, J = 8.9 Hz), 5.61 (1 H,d, J = 1.3 Hz), 4.01−4.11 (1 H, m), 3.26−3.41 (3 H, m), 3.15−3.24(2 H, m), 2.71 (3 H, s), 2.42−2.53 (1 H, m), 2.24−2.34 (1 H, m),2.07−2.18 (2 H, m), 1.76−2.04 (5 H, m), 1.58−1.72 (3 H, m), 1.30−1.42 (2 H, m), 1.21−1.24 (1 H, m), 1.07−1.18 (8 H, m), 0.91−1.04(2 H, m), 0.54 (3 H, s). m/z (ESI, +ve ion) 498.3 [M + H]+.

(8R,9S,10R,11S,13S,14S)-11-(4-(Isopropyl(methyl)amino)phenyl)-13-methyl-1,6,7,8,9,10,11,12,13,14,15,16-dodecahydrospiro-[cyclopenta[a]phenanthrene-3,2′-[1,3]dithiolan]-17(2H)-one (43b).To a solution of 42b (1.17 g, 2.35 mmol) in toluene (19.6 mL) wasadded cyclohexanone (7.00 mL, 6.77 mmol) and aluminum propan-2-olate (648 mg, 3.17 mmol) successively. After the reaction was heatedto 105 °C for 4 h, it was cooled to rt and quenched with saturated aqRochelle’s salt solution. The mixture was stirred for 10 min andextracted (2 × EtOAc). The combined organic layer was washed(brine), dried (MgSO4), and concentrated under reduced pressure.Purification of the residue by Combi-flash (SiO2, 15−30% EtOAc inhexanes, gradient elution) provided 43b (695 mg, 1.40 mmol, 60%yield) as a white solid. 1H NMR (400 MHz, CDCl3) δ ppm 7.22 (2H, d, J = 8.8 Hz), 6.68 (2 H, d, J = 8.8 Hz), 5.64 (1 H, d, J = 1.5 Hz),4.06 (1 H, dt, J = 13.3, 6.6 Hz), 3.25−3.41 (3 H, m), 3.16−3.25 (2 H,m), 2.71 (3 H, s), 2.37−2.55 (2 H, m), 2.33 (1 H, dt, J = 13.3, 3.1Hz), 1.94−2.21 (8 H, m), 1.62−1.80 (3 H, m), 1.25−1.37 (2 H, m),1.16 (6 H, t, J = 6.50 Hz), 1.01−1.13 (2 H, m), 0.66 (3 H, s). m/z(ESI, +ve ion) 496.4 [M + H]+.

(8R,9S,10R,11S,13S,14S,17S)-17-(3,3-Dimethylbut-1-yn-1-yl)-11-( 4 - ( i s o p r o p y l ( m e t h y l ) am i n o ) p h e n y l ) - 1 3 -m e t h y l -1,2,6,7,8,9,10,11,12,13,14,15,16,17-tetradecahydrospiro-[cyclopenta[a]phenanthrene-3,2′-[1,3]dithiolan]-17-ol (44c). To asolution of 1.6 M n-butyllithium in hexanes (8.80 mL, 14.2 mmol) inTHF (7 mL) was added 3,3-dimethylbut-1-yne (2.10 mL, 17.3 mmol)at −78 °C. After 30 min, a solution of 43b (390 mg, 0.79 mmol) inTHF (8 mL) was added slowly. After the reaction was stirred at −78°C for 1 h, it was allowed to warm to rt and stirred for 3 h. Thereaction was quenched (satd aq NH4Cl) and extracted (2 × EtOAc).The combined organic layers were washed (brine), dried (MgSO4),and concentrated under reduced pressure. Purification of the residueby Combi-flash (SiO2, 15−30% EtOAc in hexanes, gradient elution)provided 44c (440 mg, 0.76 mmol, 97% yield) as a white foam. 1HNMR (400 MHz, CDCl3) δ ppm 7.24 (2 H, br d, J = 8.6 Hz), 6.68 (2H, br d, J = 8.6 Hz), 5.61 (1 H, s), 4.06 (1 H, dt, J = 13.0, 6.6 Hz),3.32−3.41 (3 H, m), 3.27−3.31 (1 H, m), 3.17−3.25 (1 H, m), 2.71(3 H, s), 2.45−2.54 (1 H, m), 2.29 (1 H, br d, J = 13.7 Hz), 2.06−2.24 (4 H, m), 1.77−2.00 (5 H, m), 1.69 (2 H, br d, J = 10.5 Hz),1.41−1.50 (1 H, m), 1.29−1.38 (2 H, m), 1.25 (9 H, s), 1.16 (3 H, d,J = 6.5 Hz), 1.16 (3 H, d, J = 6.5 Hz), 1.03−1.11 (2 H, m), 0.99 (1 H,br d, J = 12.0 Hz), 0.62 (3 H, s). m/z (ESI, +ve ion) 578.5 [M + H]+.

(8R,9S,10R,11S,13S,14S,17S)-17-(3,3-Dimethylbut-1-yn-1-yl)-17-hydroxy-11-(4-(isopropyl(methyl)amino)phenyl)-13-methyl-1,2,6,7,8,9,10,11,12,13,14,15,16,17-tetradecahydro-3H-cyclopenta-[a]phenanthren-3-one (28). To a solution of 44c (114 mg, 0.20mmol) in acetic acid (1.7 mL) was added glyoxylic acid monohydrate(908 mg, 9.86 mmol) at rt, and the mixture was stirred at rt for 10min. Then 4 N HCl in water (0.89 mL, 3.6 mmol) was added and theresulting solution was stirred at rt for 20 h. The reaction wasquenched (satd aq NaHCO3), extracted (2 × EtOAc), and washed(brine). The combined organic layers were dried (Na2SO4) andconcentrated under reduced pressure. Purification of the residue withCombi-flash (SiO2, 20−40% EtOAc in hexanes, gradient elution)provided 28 (38 mg, 0.076 mmol, 38% yield) as a white foam. 1HNMR (400 MHz, CDCl3) δ ppm 7.23 (2 H, br d, J = 8.3 Hz), 6.71 (2H, br d, J = 8.3 Hz), 5.86 (1 H, s), 4.04−4.11 (1 H, m), 3.33 (1 H, brs), 2.86 (1 H, br s), 2.72 (3 H, s), 2.53 (1 H, br d, J = 14.3 Hz), 2.33−

Journal of Medicinal Chemistry Article

DOI: 10.1021/acs.jmedchem.8b00743J. Med. Chem. 2018, 61, 7767−7784

7781

Page 16: Discovery of a Potent and Selective Steroidal …...Discovery of a Potent and Selective Steroidal Glucocorticoid Receptor Antagonist (ORIC-101) Yosup Rew,* Xiaohui Du, John Eksterowicz,

2.43 (1 H, m), 2.07−2.31 (5 H, m), 2.03 (1 H, br s), 1.88−1.97 (3 H,m), 1.68−1.78 (1 H, m), 1.63 (1 H, s), 1.46−1.55 (2 H, m), 1.34−1.42 (1 H, m), 1.26−1.30 (1 H, m), 1.24 (9 H, s), 1.17 (3 H, d, J =5.6 Hz), 1.15 (3 H, d, J = 5.6 Hz), 1.06−1.12 (1 H, m), 0.67 (3 H, s).13C NMR (101 MHz, CDCl3) δ ppm 200.2, 167.7, 147.8, 131.3, 130.5(2 C), 124.4, 112.8 (2 C), 95.2, 81.9, 80.1, 52.7, 51.4, 48.9, 47.0, 42.0,38.8, 38.6, 38.3, 37.8, 36.7, 35.5, 31.9, 31.1 (3 C), 29.7, 27.5, 26.8,22.9, 19.3, 19.2, 14.8. m/z (ESI, +ve ion) 502.4 [M + H]+.(8R,9S,10R,11S,13S,14S,17S)-11-(4-(Dimethylamino)phenyl)-17-

( 3 , 3 - d ime thy l bu t - 1 - yn - 1 - y l ) - 17 -hyd roxy - 13 -me thy l -1,2,6,7,8,9,10,11,12,13,14,15,16,17-tetradecahydro-3H-cyclopenta-[a]phenanthren-3-one (26). 26 was prepared from 29 according to asimilar procedure described for the synthesis of 28. 1H NMR (400MHz, CDCl3) δ ppm 7.28 (2 H, br d, J = 8.4 Hz), 6.67 (2 H, br d, J =8.4 Hz), 5.87 (1 H, s), 3.34 (1 H, br t, J = 6.0 Hz), 2.95 (6 H, s),2.80−2.90 (1 H, m), 2.49−2.57 (1 H, m), 2.33−2.43 (1 H, m), 2.22−2.30 (2 H, m), 2.02−2.22 (4 H, m), 1.87−1.97 (3 H, m), 1.68−1.79(1 H, m), 1.60−1.67 (1 H, m), 1.47−1.54 (2 H, m), 1.38 (1 H, ddd J= 11.9, 5.7, 5.7 Hz), 1.26−1.30 (1 H, m), 1.24 (9 H, s), 0.67 (3 H, s).m/z (ESI, +ve ion) 474.4 (M + H)+.(8R,9S,10R,11S,13S,14S,17S)-11-(4-(Dimethylamino)phenyl)-17-

hydroxy-17-(3-methoxy-3-methylbut-1-yn-1-yl)-13-methyl-1,2,6,7,8,9,10,11,12,13,14,15,16,17-tetradecahydro-3H-cyclopenta-[a]phenanthren-3-one (27). 27 was prepared from 29 according to asimilar procedure described for the synthesis of 28. 1H NMR (400MHz, CDCl3) δ ppm 7.26−7.31 (2 H, m), 6.61−6.74 (2 H, m), 5.86(1 H, s), 3.37 (3 H, s), 3.32−3.36 (1 H, m), 2.96 (3 H, br s), 2.96 (3H, br s), 2.80−2.89 (1 H, m), 2.50−2.58 (1 H, m), 2.33−2.43 (1 H,m), 2.19−2.31 (3 H, m), 2.01−2.15 (3 H, m), 1.87−1.98 (3 H, m),1.72−1.80 (1 H, m), 1.70 (1 H, s), 1.52 (1 H, br d, J = 7.6 Hz), 1.48(3 H, s), 1.48 (3 H, s), 1.37−1.45 (2 H, m), 1.22−1.32 (1 H, m),1.05−1.15 (1 H, m), 0.68 (3 H, s). m/z (ESI, +ve ion) 490.4 [M +H]+.

■ ASSOCIATED CONTENT*S Supporting InformationThe Supporting Information is available free of charge on theACS Publications website at DOI: 10.1021/acs.jmed-chem.8b00743.

In vitro biological assays, in vivo study protocols, HPLCprofiles of compounds 11 and 28, ribbon view of GRcocrystal X-ray structures of 1 and 11, determination ofthe cocrystal structure of 11 with GR, and determinationof the single crystal structures of 11 and 28 (PDF)Molecular formula strings (CSV)

Accession CodesThe coordinates of 11 with GR have been deposited in theProtein Data Bank with accession code 6DXK. Authors willrelease the atomic coordinates and experimental data uponarticle publication.

■ AUTHOR INFORMATIONCorresponding Authors*For Y.R.: phone, 650-388-5612; E-mail, [email protected].*For D.S.: phone, 650-388-5607; E-mail, [email protected] Rew: 0000-0003-1155-4298NotesThe authors declare no competing financial interest.

■ ACKNOWLEDGMENTSWe thank Nigel Walker for his technical advice and guidancewith protein crystallography efforts, Proteros Biostructures

GmbH for protein production, crystallography, and structuredetermination, and Dennis Yamashita for critical reading ofmanuscript.

■ ABBREVIATIONS USEDAcCN, acetonitrile; AcOH, acetic acid; AR, androgen receptor;CL, clearance; CMC, carboxymethyl cellulose; CRPC,castration-resistant prostate cancer; CYP3A4, cytochromeP450 3A4; CYP 2C8, cytochrome P450 2C8; CYP2C9,cytochrome P450 2C9; DCM, dichloromethane; DMA,dimethylacetamide; DEAD, diethyl azodicarboxylate; DMSO,dimethyl sulfoxide; ER, estrogen receptor; EtOAc, ethylacetate; FKBP5, FK506 binding protein 5; GILZ, glucocorti-coid-induced leucine zipper; GCs, glucocorticoids; GR,glucocorticoid receptor; HPMC, hydroxypropyl methylcellu-lose; IPA, isopropyl alcohol; LNAR, LNCaP/AR-luc; NCoR,nuclear receptor corepressor; MR, mineralocorticoid receptor;OVCAR5, human ovarian carcinoma; PBMC, peripheral bloodmononuclear cell; PEG, polyethylene glycol; PR, progesteronereceptor; PTSA, p-toluenesulfonic acid; QD, once a daydosing; RT-qPCR, quantitative reverse transcription polymer-ase chain reaction; SEM, standard error of mean; TNBC,triple-negative breast cancer; TFA, trifluoroacetic acid; THF,tetrahydrofuran.

■ REFERENCES(1) Azher, S.; Azami, O.; Amato, C.; McCullough, M.; Celentano,A.; Cirillo, N. The non-conventional effects of glucocorticoids incancer. J. Cell. Physiol. 2016, 231, 2368−2373.(2) Clark, R. D. Glucocorticoid receptor antagonists. Curr. Top. Med.Chem. 2008, 8, 813−838.(3) Volden, P. A.; Conzen, S. D. The influence of glucocorticoidsignaling on tumor progression. Brain, Behav., Immun. 2013, 30, S26−S31.(4) Pan, D.; Kocherginsky, M.; Conzen, S. D. Activation of theglucocorticoid receptor is associated with poor prognosis in estrogenreceptor-negative breast cancer. Cancer Res. 2011, 71, 6360−6370.(5) Tangen, I. L.; Veneris, J. T.; Halle, M. K.; Werner, H. M.; Trovik,J.; Akslen, L. A.; Salvesen, H. B.; Conzen, S. D.; Fleming, G. F.;Krakstad, C. Expression of glucocorticoid receptor is associated withaggressive primary endometrial cancer and increases from primary tometastatic lesions. Gynecol. Oncol. 2017, 147, 672−677.(6) Arora, V. K.; Schenkein, E.; Murali, R.; Subudhi, S. K.;Wongvipat, J.; Balbas, M. D.; Shah, N.; Cai, L.; Efstathiou, E.;Logothetis, C.; Zheng, D.; Sawyers, C. L. Glucocorticoid receptorconfers resistance to antiandrogens by bypassing androgen receptorblockade. Cell 2013, 155, 1309−1322.(7) Kroon, J.; Puhr, M.; Buijs, J. T.; van der Horst, G.; Hemmer, D.M.; Marijt, K. A.; Hwang, M. S.; Masood, M.; Grimm, S.; Storm, G.;Metselaar, J. M.; Meijer, O. C.; Culig, Z.; van der Pluijm, G.Glucocorticoid receptor antagonism reverts docetaxel resistance inhuman prostate cancer. Endocr.-Relat. Cancer 2016, 23, 35−45.(8) Stringer-Reasor, E. M.; Baker, G. M.; Skor, M. N.; Kocherginsky,M.; Lengyel, E.; Fleming, G. F.; Conzen, S. D. Glucocorticoidreceptor activation inhibits chemotherapy-induced cell death in high-grade serous ovarian carcinoma. Gynecol. Oncol. 2015, 138, 656−662.(9) Skor, M. N.; Wonder, E. L.; Kocherginsky, M.; Goyal, A.; Hall,B. A.; Cai, Y.; Conzen, S. D. Glucocorticoid receptor antagonism as anovel therapy for triple-negative breast cancer. Clin. Cancer Res. 2013,19, 6163−6172.(10) Chen, Z.; Lan, X.; Wu, D.; Sunkel, B.; Ye, Z.; Huang, J.; Liu, Z.;Clinton, S. K.; Jin, V. X.; Wang, Q. Ligand-dependent genomicfunction of glucocorticoid receptor in triple-negative breast cancer.Nat. Commun. 2015, 6, 8323.(11) Zhang, C.; Mattern, J.; Haferkamp, A.; Pfitzenmaier, J.;Hohenfellner, M.; Rittgen, W.; Edler, L.; Debatin, K. M.; Groene, E.;

Journal of Medicinal Chemistry Article

DOI: 10.1021/acs.jmedchem.8b00743J. Med. Chem. 2018, 61, 7767−7784

7782

Page 17: Discovery of a Potent and Selective Steroidal …...Discovery of a Potent and Selective Steroidal Glucocorticoid Receptor Antagonist (ORIC-101) Yosup Rew,* Xiaohui Du, John Eksterowicz,

Herr, I. Corticosteroid-induced chemotherapy resistance in urologicalcancers. Cancer Biol. Ther. 2006, 5, 59−64.(12) Ge, H.; Ni, S.; Wang, X.; Xu, N.; Liu, Y.; Wang, X.; Wang, L.;Song, D.; Song, Y.; Bai, C. Dexamethasone reduces sensitivity tocisplatin by blunting p53-dependent cellular senescence in non-smallcell lung cancer. PLoS One 2012, 7, e51821.(13) Li, Z.; Chen, Y.; Cao, D.; Wang, Y.; Chen, G.; Zhang, S.; Lu, J.Glucocorticoid up-regulates transforming growth factor-β (TGF-β)type II receptor and enhances TGF-β signaling in human prostatecancer PC-3 cells. Endocrinology 2006, 147, 5259−5267.(14) Isikbay, M.; Otto, K.; Kregel, S.; Kach, J.; Cai, Y.; VanderGriend, D. J.; Conzen, S. D.; Szmulewitz, R. Z. Glucocorticoidreceptor activity contributes to resistance to androgen-targetedtherapy in prostate cancer. Horm. Cancer 2014, 5, 72−89.(15) Yin, H.; Galfalvy, H.; Pantazatos, S. P.; Huang, Y.-y.; Rosoklija,G. B.; Dwork, A. J.; Burke, A.; Arango, V.; Oquendo, M. A.; Mann, J.J. Glucocorticoid receptor-related genes: genotype and brain geneexpression relationships to suicide and major depressive disorder.Depression Anxiety 2016, 33, 531−540.(16) Sorrentino, G.; Ruggeri, N.; Zannini, A.; Ingallina, E.; Bertolio,R.; Marotta, C.; Neri, C.; Cappuzzello, E.; Forcato, M.; Rosato, A.;Mano, M.; Bicciato, S.; Del Sal, G. Glucocorticoid receptor signallingactivates YAP in breast cancer. Nat. Commun. 2017, 8, 14073.(17) Chen, Y.-X.; Wang, Y.; Fu, C.-C.; Diao, F.; Song, L.-N.; Li, Z.-B.; Yang, R.; Lu, J. Dexamethasone enhances cell resistance tochemotherapy by increasing adhesion to extracellular matrix in humanovarian cancer cells. Endocr.-Relat. Cancer 2010, 17, 39−50.(18) Hou, W. J.; Guan, J. H.; Dong, Q.; Han, Y. H.; Zhang, R.Dexamethasone inhibits the effect of paclitaxel on human ovariancarcinoma xenografts in nude mice. Eur. Rev. Med. Pharmacol. Sci.2013, 17, 2902−2908.(19) Cain, D. W.; Cidlowski, J. A. Immune regulation byglucocorticoids. Nat. Rev. Immunol. 2017, 17, 233−247.(20) Bamberger, C. M.; Chrousos, G. P. The glucocorticoid receptorand RU 486 in man. Ann. N. Y. Acad. Sci. 1995, 761, 296−310.(21) Coutinho, A. E.; Chapman, K. E. The anti-inflammatory andimmunosuppressive effects of glucocorticoids, recent developmentsand mechanistic insights. Mol. Cell. Endocrinol. 2011, 335, 2−13.(22) Karagiannidis, C.; Akdis, M.; Holopainen, P.; Woolley, N. J.;Hense, G.; Rueckert, B.; Mantel, P.-Y.; Menz, G.; Akdis, C. A.; Blaser,K.; Schmidt-Weber, C. B. Glucocorticoids upregulate FOXP3expression and regulatory T cells in asthma. J. Allergy Clin. Immunol.2004, 114, 1425−1433.(23) Zhang, K.; Bai, X.; Li, R.; Xiao, Z.; Chen, J.; Yang, F.; Li, Z.Endogenous glucocorticoids promote the expansion of myeloid-derived suppressor cells in a murine model of trauma. Int. J. Mol. Med.2012, 30, 277−282.(24) Lu, Y.; Liu, H.; Bi, Y.; Yang, H.; Li, J.; Wang, J.; Zhang, Z.;Wang, Y.; Li, C.; Jia, A.; Han, L.; Hu, Y.; Zhao, Y.; Wang, R.; Liu, G.Glucocorticoid receptor promotes the function of myeloid-derivedsuppressor cells by suppressing HIF1α-dependent glycolysis. Cell.Mol. Immunol. 2018, 15, 618−629.(25) Liberman, A. C.; Refojo, D.; Druker, J.; Toscano, M.; Rein, T.;Holsboer, F.; Arzt, E. The activated glucocorticoid receptor inhibitsthe transcription factor T-bet by direct protein-protein interaction.FASEB J. 2007, 21, 1177−1188.(26) Xing, K.; Gu, B.; Zhang, P.; Wu, X. Dexamethasone enhancesprogrammed cell death 1 (PD-1) expression during T cell activation:an insight into the optimum application of glucocorticoids in anti-cancer therapy. BMC Immunol. 2015, 16, 39.(27) Sharma, P.; Hu-Lieskovan, S.; Wargo, J. A.; Ribas, A. Primary,adaptive, and acquired resistance to cancer immunotherapy. Cell2017, 168, 707−723.(28) Sephton, S. E.; Sapolsky, R. M.; Kraemer, H. C.; Spiegel, D.Diurnal cortisol rhythm as a predictor of breast cancer survival. J. Natl.Cancer Inst. 2000, 92, 994−1000.(29) Sephton, S. E.; Lush, E.; Dedert, E. A.; Floyd, A. R.; Rebholz,W. N.; Dhabhar, F. S.; Spiegel, D.; Salmon, P. Diurnal cortisol rhythm

as a predictor of lung cancer survival. Brain, Behav., Immun. 2013, 30,S163−S170.(30) Schrepf, A.; Thaker, P. H.; Goodheart, M. J.; Bender, D.;Slavich, G. M.; Dahmoush, L.; Penedo, F.; DeGeest, K.; Mendez, L.;Lubaroff, D. M.; Cole, S. W.; Sood, A. K.; Lutgendorf, S. K. Diurnalcortisol and survival in epithelial ovarian cancer. Psychoneuroendocri-nology 2015, 53, 256−267.(31) Sidler, D.; Renzulli, P.; Schnoz, C.; Berger, B.; Schneider-Jakob,S.; Fluck, C.; Inderbitzin, D.; Corazza, N.; Candinas, D.; Brunner, T.Colon cancer cells produce immunoregulatory glucocorticoids.Oncogene 2011, 30, 2411−2419.(32) Cirillo, N.; Morgan, D. J.; Pedicillo, M. C.; Celentano, A.; LoMuzio, L.; McCullough, M. J.; Prime, S. S. Characterisation of thecancer-associated glucocorticoid system: key role of 11β-hydroxyste-roid dehydrogenase type 2. Br. J. Cancer 2017, 117, 984−993.(33) Spitz, I. M.; Bardin, C. W. Mifepristone (RU 486) - amodulator of progestin and glucocorticoid action. N. Engl. J. Med.1993, 329, 404−412.(34) Fleseriu, M.; Molitch, M. E.; Gross, C.; Schteingart, D. E.;Vaughan, T. B., 3rd; Biller, B. M. K. A new therapeutic approach inthe medical treatment of Cushing’s syndrome: glucocorticoid receptorblockade with mifepristone. Endocr. Pract. 2013, 19, 313−26.(35) Hunt, H. J.; Belanoff, J. K.; Walters, I.; Gourdet, B.; Thomas, J.;Barton, N.; Unitt, J.; Phillips, T.; Swift, D.; Eaton, E. Identification ofthe clinical candidate (R)-(1-(4-fluorophenyl)-6-((1-methyl-1H-pyr-azol-4-yl)sulfonyl)-4,4a,5,6,7,8-hexahydro-1H-pyrazolo[3,4-g]-isoquinolin-4a-yl)(4-(trifluoromethyl)pyridin-2-yl)methanone(CORT125134): a selective glucocorticoid receptor (GR) antagonist.J. Med. Chem. 2017, 60, 3405−3421.(36) Hunt, H. J.; Belanoff, J. K.; Golding, E.; Gourdet, B.; Phillips,T.; Swift, D.; Thomas, J.; Unitt, J. F.; Walters, I. 1H-Pyrazolo[3,4-g]hexahydro-isoquinolines as potent GR antagonists with reducedhERG inhibition and an improved pharmacokinetic profile. Bioorg.Med. Chem. Lett. 2015, 25, 5720−5725.(37) Clark, R. D.; Ray, N. C.; Williams, K.; Blaney, P.; Ward, S.;Crackett, P. H.; Hurley, C.; Dyke, H. J.; Clark, D. E.; Lockey, P.;Devos, R.; Wong, M.; Porres, S. S.; Bright, C. P.; Jenkins, R. E.;Belanoff, J. 1H-Pyrazolo[3,4-g]hexahydro-isoquinolines as selectiveglucocorticoid receptor antagonists with high functional activity.Bioorg. Med. Chem. Lett. 2008, 18, 1312−1317.(38) Hunt, H. J.; Ray, N. C.; Hynd, G.; Sutton, J.; Sajad, M.;O’Connor, E.; Ahmed, S.; Lockey, P.; Daly, S.; Buckley, G.; Clark, R.D.; Roe, R.; Blasey, C.; Belanoff, J. Discovery of a novel non-steroidalGR antagonist with in vivo efficacy in the olanzapine-induced weightgain model in the rat. Bioorg. Med. Chem. Lett. 2012, 22, 7376−7380.(39) Luz, J. G.; Carson, M. W.; Condon, B.; Clawson, D.; Pustilnik,A.; Kohlman, D. T.; Barr, R. J.; Bean, J. S.; Dill, M. J.; Sindelar, D. K.;Maletic, M.; Coghlan, M. J. Indole glucocorticoid receptor antagonistsactive in a model of dyslipidemia act via a unique association with anagonist binding site. J. Med. Chem. 2015, 58, 6607−6618.(40) Christodoulou, M. S.; Dapiaggi, F.; Ghiringhelli, F.; Pieraccini,S.; Sironi, M.; Lucafo, M.; Curci, D.; Decorti, G.; Stocco, G.;Chirumamilla, C. S.; Vanden Berghe, W.; Balaguer, P.; Michel, B. Y.;Burger, A.; Beccalli, E. M.; Passarella, D.; Martinet, N. Imidazo[2,1-b]benzothiazol derivatives as potential allosteric inhibitors of theglucocorticoid receptor. ACS Med. Chem. Lett. 2018, 9, 339−344.(41) Zhou, H.; Jahchan, N.; Tran, C.; McWeeney, D.; Balbas, M.;Schenkein, E.; Sutimantanapi, D.; Zavorotinskaya, T.; Jackson, E.;Medina, J.; Sun, D.; Rew, Y.; Du, X.; Yan, X.; Zhu, L.; Ye, Q.; Singh,M.; Fantin, V. Activation of AR signaling by Mifepristone enhancesprostate cancer growth and impairs enzalutamide response.Proceedings of the American Association for Cancer Research, WashingtonDC, April 1−5, 2017.(42) Malinen, M.; Toropainen, S.; Jaaskelainen, T.; Sahu, B.; Janne,O. A.; Palvimo, J. J. Androgen receptor- and PIAS1-regulated geneprograms in molecular apocrine breast cancer cells. Mol. Cell.Endocrinol. 2015, 414, 91−98.(43) Cochrane, D. R.; Bernales, S.; Jacobsen, B. M.; Cittelly, D. M.;Howe, E. N.; D’Amato, N. C.; Spoelstra, N. S.; Edgerton, S. M.; Jean,

Journal of Medicinal Chemistry Article

DOI: 10.1021/acs.jmedchem.8b00743J. Med. Chem. 2018, 61, 7767−7784

7783

Page 18: Discovery of a Potent and Selective Steroidal …...Discovery of a Potent and Selective Steroidal Glucocorticoid Receptor Antagonist (ORIC-101) Yosup Rew,* Xiaohui Du, John Eksterowicz,

A.; Guerrero, J.; Gomez, F.; Medicherla, S.; Alfaro, I. E.; McCullagh,E.; Jedlicka, P.; Torkko, K. C.; Thor, A. D.; Elias, A. D.; Protter, A. A.;Richer, J. K. Role of the androgen receptor in breast cancer andpreclinical analysis of enzalutamide. Breast Cancer Res. 2014, 16, R7.(44) Lehmann, B. D.; Bauer, J. A.; Schafer, J. M.; Pendleton, C. S.;Tang, L.; Johnson, K. C.; Chen, X.; Balko, J. M.; Gomez, H.; Arteaga,C. L.; Mills, G. B.; Sanders, M. E.; Pietenpol, J. A. PIK3CA mutationsin androgen receptor-positive triple negative breast cancer confersensitivity to the combination of PI3K and androgen receptorinhibitors. Breast Cancer Res. 2014, 16, 406.(45) Park, B. Y.; Grisham, R. N.; den Hollander, B.; Thapi, D.;Berman, T.; de Stanchina, E.; Zhou, Q.; Iyer, G.; Aghajanian, C.;Spriggs, D. R. Tumor inhibition by enzalutamide in a xenograft modelof ovarian cancer. Cancer Invest. 2016, 34, 517−520.(46) Traina, T. A.; Miller, K.; Yardley, D. A.; Eakle, J.; Schwartzberg,L. S.; O’Shaughnessy, J.; Gradishar, W.; Schmid, P.; Winer, E.; Kelly,C.; Nanda, R.; Gucalp, A.; Awada, A.; Garcia-Estevez, L.; Trudeau, M.E.; Steinberg, J.; Uppal, H.; Tudor, I. C.; Peterson, A.; Cortes, J.Enzalutamide for the treatment of androgen receptor-expressingtriple-negative breast cancer. J. Clin. Oncol. 2018, 36, 884−890.(47) NCT01974765. Enzalutamide in Patients With AndrogenReceptor Positive (AR+) Ovarian, Primary Peritoneal or FallopianTube Cancer and One, Two or Three Prior Therapies. Clinical-Trials.gov; U.S. National Institutes of Health: Bethesda, MD, 2013;https://www.clinicaltrials.gov/ct2/show/NCT01974765?term=NCT01974765&rank=1.(48) Feng, Z.; Wen, H.; Bi, R.; Ju, X.; Chen, X.; Yang, W.; Wu, X. Aclinically applicable molecular classification for high-grade serousovarian cancer based on hormone receptor expression. Sci. Rep. 2016,6, 25408.(49) Schoch, G. A.; D’Arcy, B.; Stihle, M.; Burger, D.; Bar, D.; Benz,J.; Thoma, R.; Ruf, A. Molecular switch in the glucocorticoidreceptor: active and passive antagonist conformations. J. Mol. Biol.2010, 395, 568−577.(50) Kauppi, B.; Jakob, C.; Farnegardh, M.; Yang, J.; Ahola, H.;Alarcon, M.; Calles, K.; Engstrom, O.; Harlan, J.; Muchmore, S.;Ramqvist, A.-K.; Thorell, S.; Ohman, L.; Greer, J.; Gustafsson, J.-A.;Carlstedt-Duke, J.; Carlquist, M. The three-dimensional structures ofantagonistic and agonistic forms of the glucocorticoid receptor ligand-binding domain: RU-486 induces a transconformation that leads toactive antagonism. J. Biol. Chem. 2003, 278, 22748−22754.(51) Kojetin, D. J.; Burris, T. P. Small molecule modulation ofnuclear receptor conformational dynamics: implications for functionand drug discovery. Mol. Pharmacol. 2013, 83, 1−8.(52) Huang, P.; Chandra, V.; Rastinejad, F. Structural overview ofthe nuclear receptor superfamily: insights into physiology andtherapeutics. Annu. Rev. Physiol. 2010, 72, 247−272.(53) Li, Y.; Lambert, M. H.; Xu, H. E. Activation of nuclearreceptors. a perspective from structural genomics. Structure 2003, 11,741−746.(54) Helices 11 and 12 are mobile and can be positioned tofavorably bind coactivators resulting in agonism, and corepressors orno copeptide, both of which result in antagonism.(55) Detailed SAR studies on the modification of the C17 hydroxyyland introduction of the C16 substituents will be reported in separatecommunications.(56) Several known mifepristone analogues (2, 4, 5, 6, and 10) weresynthesized to test AR agonism. Since its original discovery in 1981,the majority of medicinal chemistry research on mifepristone and itsanalogues has been focused on exploring the SAR around PR and GRselectivity.(57) Belanger, A.; Philibert, D.; Teutsch, G. Regio and stereospecificsynthesis of 11β-substituted 19-norsteroids: influence of 11β-substitution on progesterone receptor affinity - (1). Steroids 1981,37, 361−382.(58) Teutsch, G.; Gaillard-Moguilewsky, M.; Lemoine, G.; Nique,F.; Philibert, D. Design of ligands for the glucocorticoid and progestinreceptors. Biochem. Soc. Trans. 1991, 19, 901−908.

(59) Ray, D. W.; Davis, J. R. E.; White, A.; Clark, A. J. L.Glucocorticoid receptor structure and function in glucocorticoid-resistant small cell lung carcinoma cells. Cancer Res. 1996, 56, 3276−3280.(60) Soden, J.; Stevens, A.; Ray, D. W. Genetic engineering of theglucocorticoid receptor by fusion with the herpes viral protein VP22causes selective loss of transactivation. J. Endocrinol. 2002, 172, 615−625.(61) Hazra, B. G.; Basu, S.; Pore, V. S.; Joshi, P. L.; Pal, D.;Chakrabarti, P. Synthesis of 11β-(4-dimethylaminophenyl)-17β-hydroxy-17α-(3-methyl-1-butynyl)-4,9-estradien-3-one and 11β-(4-acetophenyl)-17β-hydroxy-17α-(3-methyl-1-butynyl)-4,9-estradien-3-one: two new analogs of mifepristone (RU-486). Steroids 2000, 65,157−162.(62) These large changes in the protein structure are localized tohelices 6, 7, 11, and 12 while the remaining protein maintains astructure similar to that of when mifepristone is bound to GR. Thelarge movement of helix 12 results in an opening of the binding site,and the crystal packing shows the side chain of Trp557 from anothermonomer of GR directly interacting with 11 through this opening. Anumber of new GR constructs were designed to reduce or remove thispacking interaction but crystallization attempts were not successful.(63) Metabolite profiles of compound 28 in human, rat, and minipigare similar in both in vitro hepatocyte assay and in vivo PK study.(64) van Zuiden, M.; Geuze, E.; Willemen, H. L. D. M.; Vermetten,E.; Maas, M.; Amarouchi, K.; Kavelaars, A.; Heijnen, C. J.Glucocorticoid receptor pathway components predict posttraumaticstress disorder symptom development: a prospective study. Biol.Psychiatry 2012, 71, 309−316.(65) Giannopoulos, G.; Keichline, D. Species-related differences insteroid-binding specificity of glucocorticoid receptors in lung.Endocrinology 1981, 108, 1414−1419.(66) Touitou, Y.; Bogdan, A.; Levi, F.; Benavides, M.; Auzeby, A.Disruption of the circadian patterns of serum cortisol in breast andovarian cancer patients: relationships with tumour marker antigens.Br. J. Cancer 1996, 74, 1248−1252.(67) Mazzoccoli, G.; Carughi, S.; De Cata, A.; La Viola, M.;Vendemiale, G. Melatonin and cortisol serum levels in lung cancerpatients at different stages of disease. Med. Sci. Monit. 2005, 11,CR284−CR288.(68) Suessmeier, F.; Lobell, M.; Gruenewald, S.; Haerter, M.;Buchmann, B.; Telser, J.; Joerissen, H.; Heroult, M.; Kahnert, A.;Lustig, K.; Lindner, N. Substituted Phenylimidazopyrazoles and TheirUse. U.S. Patent US 9,394,309, 2016.(69) Cleve, A.; Ottow, E.; Neef, G.; Wiechert, R. Synthesis ofoxygen-bridged antigestagens. Tetrahedron 1993, 49, 2217−2226.(70) Cleve, A.; Scheidges, C.; Neef, G.; Ottow, E.; Elger, W.; Beier,S. 11β-Aryl-4-estrenes, Process for Their Productions as well asPharmaceutical Agents. U.S. Patent US5,728,689, 1998.(71) NCT03324555. Study of ORIC-101 in Healthy Adult Subjects.ClinicalTrials.gov; U.S. National Institutes of Health: Bethesda, MD,2 0 1 7 ; h t t p s : / / w ww . c l i n i c a l t r i a l s . g o v / c t 2 / s h o w /NCT03324555?term=NCT03324555&rank=1.

Journal of Medicinal Chemistry Article

DOI: 10.1021/acs.jmedchem.8b00743J. Med. Chem. 2018, 61, 7767−7784

7784