Abstract
Soluble adenylyl cyclase (sAC: ADCY10) is an enzyme involved in intracellular signaling. Inhibition of sAC has potential therapeutic utility in a number of areas. For example, sAC is integral to successful male fertility: sAC activation is required for sperm motility and ability to undergo the acrosome reaction, two processes central to oocyte fertilization. Pharmacologic evaluation of existing sAC inhibitors for utility as on-demand, nonhormonal male contraceptives suggested that both high intrinsic potency, fast on and slow dissociation rates are essential design elements for successful male contraceptive applications. During the course of the medicinal chemistry campaign described here, we identified sAC inhibitors that fulfill these criteria and are suitable for in vivo evaluation of diverse sAC pharmacology.
Graphical Abstract

INTRODUCTION
Cyclic AMP (cAMP) is an important second messenger that plays a key role in numerous signal transduction pathways. cAMP-dependent signaling cascades regulate various cellular processes such as proliferation, apoptosis, differentiation, migration, development, ion transport, pH regulation, and gene expression.1,2 In mammals, there are two distinct types of enzyme that produce cAMP from ATP: the G-protein-regulated transmembrane adenylyl cyclases (tmACs; ADCY1–9) and a bicarbonate-regulated soluble adenylyl cyclase (sAC, ADCY10).3 Unlike tmACs, which are localized to the plasma membrane, sAC is found within the cytosol and in several compartmentalized microdomains located within cellular organelles.4–7 sAC-generated cAMP signaling has been linked to multiple physiological functions8 such as insulin release from pancreatic β-cells,9 regulation of intraocular eye pressure,10,11 lysosomal acidification,4 and, the capacitation of sperm.12,13 Due to its varied biological roles, sAC has been identified as a potential novel drug target in a number of therapeutic areas.2
In particular, recent work has increased interest in the use of sAC as a drug target for contraception.14,15 Safe, effective new contraceptive strategies for males, particularly nonhormonal methods, represent a profound unmet medical need as globally, approximately half of all pregnancies are unplanned.16,17 In males, sperm are stored in the cauda epididymis in their inactivated form (“noncapacitated”) under low bicarbonate conditions (<5 mM).18–20 Upon ejaculation, sperm are mixed with seminal fluid containing high bicarbonate levels (20–30 mM). Exposure to bicarbonate, the endogenous ligand for sAC,21 leads to an increase in intracellular cAMP, which stimulates sperm motility and converts sperm into their activated (“capacitated”) form able to undergo the acrosome reaction required for oocyte fertilization.12 Consistent with these observations, two strains of mice lacking sAC are sterile due to a male-specific sperm-motility defect.13,22 More recently, two adult human males presented as infertile with immotile sperm, a defect which was traced to an ADCY10 frameshift mutation.23 Treatment of immotile sperm from knockout mice or the two infertile adcy10 mutant men with a cell-permeable cAMP analogue restored sperm motility, which represents key confirmatory mechanistic data that their infertility is due to the absence of sAC.23 Importantly, despite sAC expression in other tissues,21 mechanism-based liabilities appear modest as both the KO mice and the humans bearing the ADCY10 frameshift mutation exhibit overall benign profiles.13,22,23 These aggregate datasets support the hypothesis that sAC represents a highly attractive target for the development of an on-demand, nonhormonal male contraceptive.
To interrogate sAC inhibition for therapeutic utility, high-quality pharmacologic tools are required. We previously described the discovery of TDI-10229, a potent and orally bioavailable inhibitor of sAC.24 Drug design efforts were aided by abundant structural information from X-ray crystallography on sAC alone and in complex with inhibitors.25–27 While sAC is similar to transmembrane adenylyl cyclases (tmACs 1–9) in overall structure, its regulatory bicarbonate binding site has unique features, which are exploited by TDI-10229.24,27 Briefly, TDI-10229 has an IC50 of 160 nM on purified human sAC protein in an in vitro sAC biochemical assay25 and a cellular IC50 of 92 nM for sAC in sAC-overexpressing rat 4–4 cells;25 is highly selective versus the closely related and widely expressed, tmACs 1–9; and exhibited reasonable druglike properties.24 However, more recent studies suggest that TDI-10229 does not provide adequate exposure for use as a male systemic contraceptive.15 This could be due to its modest affinity (160 nM in a biochemical assay) and/or the strong dilution effect that occurs when inhibitor-containing sperm enter the inhibitor-free female body. As a potential solution, we identified fast onset and slow dissociation rates as key design goals for the development of second-generation sAC inhibitors for use as male systemic contraceptives. More generally, slow dissociation rates are often a desirable property for therapeutic use.28 We now detail our optimization efforts using TDI-10229 as a starting point, leading to the identification of high-affinity sAC inhibitors with slow dissociation rates.
RESULTS AND DISCUSSION
Design and Optimization.
The crystal structure of TDI-10229 in complex with sAC provided opportunities for structure-based drug design to fulfill our goals related to the optimization of sAC binding affinity and kinetics to improve downstream activity (Figure 1).
Figure 1.

Crystal structure of sAC complex with TDI-10229. The crystal structure of sAC in complex with TDI-10229 (2.20 Å) from PDB ID 7OVD. The protein is displayed as a green cartoon with green wires and a transparent gray surface. TDI-10229 is displayed as atom-colored balls, and sticks with magenta carbons.
TDI-10229 is substituted with methyl groups at both the 1 and 5 positions of the pyrazole and we noted the presence of a promising growth vector at the 5 position. Table 1 presents the resultant structure–activity relationships (SAR) data for substitution at the 1 and 5 positions of the pyrazole.
Table 1.
Biochemical Potency of TDI-10229 and Related Analogues
| Cpd | Structure | Biochemical IC50 (nM)a |
|---|---|---|
|
1 (TDI-10229) |
|
159 ± 7 |
| 2 |
|
70% inhibition at 500 μM |
| 3 |
|
1024 ± 235 |
| 4 |
|
829 ± 166 |
| 5 |
|
469 ± 87 |
| 6 |
|
112 ± 21 |
| 7 |
|
716 ± 245 |
Compounds were tested in a minimum of three independent assays, and data are reported as the mean ± standard error of the mean. The 1 and 5 position numbering on the pyrazole ring is shown for TDI-10229 (1).
Removal of the methyl group at the 5 position leads to a small 6-fold loss of affinity (3) whilst removal of the methyl groups at both the 1 and 5 positions leads to a dramatic loss of affinity (2). Interestingly, some affinity can be regained by the replacement of the 1-methyl group with a difluoromethyl group (4) or trifluoromethyl group (5). Cyclization between the 1 and 5 positions (6) can improve binding affinity, highlighting the opportunities available for growth at these positions, though an alternate cyclization (7) reduces binding affinity. Previously reported SAR highlighted some limited ability to improve binding affinity through substitution at the 5 position of TDI-10229.24 However, further study of the crystal structure (Figure 1) and exploitation of structure-based design tools such as free-energy perturbation29,30 suggested that the space filled by the methyl group at the 5 position of TDI-10229 could also be filled by substitution of the phenyl ring at the ortho or meta position. For this reason, we explored substitution around the phenyl ring of 3. The SAR data are presented in Table 2.
Table 2.
Biochemical Potency of Ortho-, Meta-, and Para-Substituted Phenyl Analogues
| ||||
|---|---|---|---|---|
| Cpd | R1 (SAR) | R2 (SAR) | R3 (SAR) | Biochemical IC50 (nM)a |
| 3 | H | H | H | 1024 ± 235 |
| 8 | Cl | H | H | 64 ± 9 |
| 9 | H | Cl | H | 389 ± 80 |
| 10 | H | H | Cl | 2577 ± 300 |
| 11 | OMe | H | H | 67 ± 8 |
| 12 | CO2Me | H | H | 21 ± 3 |
| 13 | C(O)NHMe | H | H | 1245 ± 123 |
| 14 | C(O)N(Me)2 | H | H | 75% inhibition at 500 μM |
| 15 | H | OMe | H | 351 ± 43 |
| 16 | H | CO2Me | H | 436 ± 39 |
| 17 | H | C(O)NHMe | H | 1876 ± 208 |
Compounds were tested in a minimum of three independent assays, and data are reported as mean ± SEM.
Results for the chlorinated analogues indicate that substitution at the ortho (8) or meta (9) position leads to increases in affinity whilst substitution at the para position is not favored (10). This is in good agreement with structural data and modeling predictions, which show that the growth vector at the para position is directed toward the protein surface. Also consistent with structural data and modeling predictions, the ortho position provides the best vector for growth, with methoxy (11) and methyl ester (12) groups yielding improved IC50 values of 67 and 21 nM, respectively. However, not all substitutions are favored, with methyl amide (13) having an IC50 of 1245 nM and dimethyl amide (14) being almost inactive. The meta position is also amenable to substitution, with methoxy (15) and methyl ester (16) groups yielding improved IC50 values but methyl amide (17) having an IC50 greater than 1 μM. Considering both the SAR and the structural information, we selected alkoxy substitution at the ortho position to explore further. The SAR data are presented in Table 3.
Table 3.
Biochemical Potency of Ortho Alkoxy-Substituted Phenyl Analogues
| Cpd | R1 (SAR) | Biochemical IC50 (nM)a |
|---|---|---|
| 18 |
|
87.1 ± 11.5 |
| 19 |
|
12.3 ± 0.8 |
| 20 |
|
6.7 ± 0.5 |
| 21 |
|
7.8 ± 1.2 |
| 22 |
|
16.9 ± 3.1 |
Compounds were tested in a minimum of three independent assays, and data are reported as mean ± SEM.
Simple substitution with a phenyl group (18) does not improve biochemical IC50, but modeling indicated that an alkyl chain with a hydrophilic or weakly basic group should be favorable. Correspondingly, the morpholine compound (19) showed an improved biochemical IC50 of 12.2 nM. Analogues such as the bridgehead (20), 2-oxa-6-azaspiro[3.3]heptane (21), and piperazin-2-one (22) are also highly potent. We focused on compound 19 as a high-affinity inhibitor. Noting the potential metabolic liability31,32 of the N-methyl pyrazole group, we incorporated SAR from compound 4 (Table 1) and replaced the methyl with a difluoromethyl group.33 The resulting compound (23) showed a biochemical IC50 of 4.0 nM. As an initial check on the improved potency of 23, we examined its efficacy in an sAC cellular assay. The cellular IC50 of 23 was 2.0 nM, significantly improved from TDI-10229’s cellular IC50 of 92 nM.
Given the sAC concentration of 5.0 nM in the biochemical assay, measurement of IC50 values below 2.5 nM cannot be effectively determined, assuming all of the protein is enzymatically competent. To quantify the increased potency of compounds such as 23, we turned to biophysical measurement of compound binding using the well-established technique of surface plasmon resonance (SPR).34 This approach offered the added advantage of allowing us to investigate the kinetics of binding to assess progress made on our goal of identifying compounds with extended residence times. SPR studies have additional value because, as noted above, the biochemical assay may have reached a floor where inhibition with an IC50 better than 2.5 nM cannot be measured effectively. Table 4 shows the biochemical IC50, cellular IC50, and SPR data for 23 along with additional morpholine-modified analogues in the context of a difluoromethyl group at the pyrazole 1 position.
Table 4.
Biochemical Potency and Cell-Based Activity of Ortho Alkoxy-Substituted Phenyl Analogues
| Cpd | R1 (SAR) | Biochemical IC50(nM)a | Cell IC50(nM)a | SPR KD(nM)a | SPR Residence Time(s)a |
|---|---|---|---|---|---|
| 23 |
|
3.9 ± 0.3 | 1.9 ± 0.9 | 3.3 ± 1.8 | 1832 ± 458 |
| 24 |
|
24.2 ± 3.9 | 37.7 ± 9.3 | 4.7 ± 1.5 | 484 ± 54 |
| 25 |
|
2.1 ± 0.2 | 5.5 ± 1.7 | 5.5 ± 1.4 | 526 ± 108 |
| 26 |
|
2.4 ± 0.3 | 14.6 ± 1.3 | 2.1 ± 1.1 | 2103 ± 319 |
| 27 |
|
4.0 ± 0.5 | 16.7 ± 6.3 | 3.6 ± 1.9 | 2716 ± 893 |
| 28 |
|
2.2 ± 0.1 | 19.6 ± 2.6 | 2.5 ± 0.4 | 1712 ± 267 |
| 29 |
|
1.0 ± 0.1 | 2.5 ± 0.2 | 1.7 ± 0.5 | 3016 ± 799 |
| 30 |
|
3.3 ± 0.2 | 34.8 ± 8.5 | 1.1 ± 0.1 | 2717 ± 338 |
| 31 |
|
5.5 ± 0.8 | 22.9 ± 4.5 | 3.7 ± 1.2 | 1301 ± 239 |
| 32 |
|
4.6 ± 0.9 | 10.8 ± 2.1 | 2.3 ± 0.5 | 2579 ± 708 |
|
33 (TDI-11861) |
|
3.3 ± 0.3 | 5.5 ± 0.9 | 1.4 ± 0.3 | 3181 ± 636 |
Compounds were tested in a minimum of three independent assays, and data are reported as mean ± SEM.
All 11 compounds in Table 4 displayed enzymatic IC50, cellular IC50, and SPR-derived KD values in the nanomolar range. The variety of substituents accommodated at the ortho phenyl position offered an ability to modulate compound properties. In particular, the activity of the amide (24) indicates that the basicity of the morpholine group is not required for high-affinity binding. Table 5 presents comparative SPR traces and associated data for TDI-11861 versus TDI-10229.
Table 5.
SPR Data for TDI-10229 and TDI-11861: The SPR Data and Example Traces for TDI-10229 and TDI-11861
| Cpd | 1 (TDI-10229) | 33 (TDI-11861) |
|---|---|---|
| Structure |
|
|
| SPR KDa | 175.6 ± 23.7 nM | 1.4 ± 0.3 nM |
| SPR kona | 2.4 ± 0.4 × 105 M−1s−1 | 2.7 ± 0.2 × 105 M−1s−1 |
| SPR koffa | 39.8 ± 2.2 × 10−3 s−1 | 0.4 ± 0.1 × 10−3 s−1 |
| SPR Residence Timea | 25 ± 1 s | 3181 ± 636 s |
| Example SPR Trace |
|
|
Compounds were tested in a minimum of three independent assays, and data are reported as mean ± SEM.
The SPR data reveal a fast off-rate profile for TDI-10229, with a residence time of only 25 s. This supports the idea that, in the context of a male contraceptive indication, the risk of post-copulation female genital tract dilution represents a valid concern. SPR also confirms the significantly improved binding affinity of TDI-11861 relative to TDI-10229 (1.4 nM compared with 176.0 nM). The concomitant improvement in dissociation rates is approximately 100-fold (3181 s compared with 25 s). Notably, both compounds exhibit similarly fast on rates, demonstrating that the improvement in potency for TDI-11861 is due almost entirely to the decrease in its dissociation rate.
As an independent check on the slow dissociation of sAC inhibitors such as TDI-11861, we performed jump dilution recovery assays.35 Briefly, in these assays, sAC protein at 5× its standard assay concentration is preincubated with an inhibitor at 10× its IC50 concentration. Following a 15 min preincubation to ensure equilibration of sAC plus inhibitor, the inhibitor-protein solution is diluted 100-fold into the standard activity assay system. Cyclase activity is initiated with the addition of substrate ATP, and the rate of conversion of substrate ATP into product cAMP is measured at various time points over the next hour. Compounds with short residence times will show minimal inhibition of sAC activity after the 100× dilution step, and their rate of enzymatic conversion of ATP into cAMP will be indistinguishable from assays performed in the absence of any inhibitor. In contrast, compounds with long residence times will show a diminished enzymatic rate at early times, as inhibitor remains engaged even after dilution. The time it takes to regain the uninhibited rate of sAC activity from the initially inhibited state defines the residence time for the inhibitor tested. These jump dilution assay results illustrate that advanced analogues, such as TDI-11861, exhibit slower dissociation rates from sAC relative to TDI-10229 (Figure 2). Importantly, these experiments recapitulate the vaginal dilution effect discussed above.
Figure 2.

Jump dilution studies of sAC inhibitors. (a) Conceptual schematic illustrating jump dilution activity recovery assay protocol. (b) Comparison of residence time from SPR and jump dilution. The residence time of TDI-10229 is too short to accurately measure via jump dilution but is included for completeness. (c) Jump dilution plots of cAMP accumulation versus time.
The residence times from SPR and jump dilution are in reasonable agreement. A consistent 2- to 3-fold difference is observed and may be due to slight differences in experimental conditions (i.e., inclusion of substrate and divalent ion cofactor in jump dilution assay). Based on its biochemical potency (IC50 = 3.3 nM), cellular potency (IC50 = 5.5 nM), and SPR affinity (KD = 1.4 nM), coupled to its comparatively long residence time (3181 and 1220 s in SPR and jump dilution, respectively), TDI-11861 is among the most potent sAC inhibitors synthesized to date. To confirm the binding mode of TDI-11861, we obtained crystallographic data in complex with sAC at 1.8 Å resolution (Figure 3; Table S1).
Figure 3.

Crystal structure of sAC complex with TDI-11861. The protein is displayed as a green cartoon with green wires. TDI-11861 is displayed as atom-colored balls, and sticks with cyan carbons. Hydrogen-bonding interactions are shown as dashed yellow lines.
The protein and ligand structures are very well aligned with the crystal structure of TDI-1022924 with the key hydrogen-bonding contacts made by the aminochloropyrimidine head-group maintained from TDI-10229 and its parent LRE1.24,25 An additional hydrogen-bonding contact is seen between the ligand hydroxyl group and the sidechain carboxylate group of Asp99 (distance 2.92 Å), a key residue for binding of the substrate ATP and the divalent metal ion. The morpholine group is close to Asp99, but not in direct contact with it, in line with basicity not being required for strong binding.
We measured the pharmacokinetic (PK) properties of TDI-11861 in mice to assess its utility as a tool compound for in vivo pharmacologic studies (see Table 6 and further details in the SI). Even though the oral bioavailability of TDI-11861 in CD-1 mice was low (F% = 11 at 10 mg/kg), a higher dose of 50 mg/kg achieved unbound plasma exposures ~10-fold over its cellular IC50 (5.5 nM) out to 4 h and this would support potential in vivo examination of sAC-mediated pharmacology (Figure 4). Indeed, higher systemic levels of TDI-11861 were attained in male mice (7.04 and 0.93 μM at 1- and 4-h post-dosing, respectively, 50 mg/kg, IP), to enable successful male contraceptive studies.17
Table 6.
Pharmacokinetic Parameters of TDI-11861 (33) in Male CD-1 Mice
| parameter | IV administrationa (2 mg/kg) | oral administrationb (10 mg/kg) | oral administrationc (50 mg/kg) |
|---|---|---|---|
| AUC(0–24h) [mM-h] | 0.78 ± 0.11 | 0.41 ± 0.057 | 5.46 ± 1.5 |
| Cmax [mM] | 2.5 ± 0.21d | 0.45 ± 0.015 | 6.1 ± 1.9 |
| Tmax [h] | 0.25 | 0.25 | |
| T1/2 [h] | 0.93 ± 0.33 | ||
| Cl [mL/min/kg] | 94 ± 12 | ||
| F [%] | 11 ± 1.4 |
Dosed as a solution of the free base (0.4 mg/mL) in 20% HPBCD.
Dosed as a solution of the free base (2 mg/mL) in DMSO/PEG400/PBS (10:40:50).
Dosed as a solution of the free base (10 mg/mL) in DMSO/PEG400/PBS (10:40:50).
Initial concentration. F, bioavailability.
Figure 4.

Exposure of TDI-11861. Unbound plasma concentration of TDI-11861 (33) in CD-1 mice at 50 mg/kg PO.
Further characterization provided additional measures of TDI-11861’s suitability for interrogating sAC pharmacology in vitro and in vivo (Table 6). TDI-11861 exhibits good permeability in a parallel artificial membrane permeability assay (PAMPA), and good solubility (463 nm/s and 79 μg/mL, respectively), consistent with its Log D value (2.89). Importantly, consistent with its predecessor TDI-10229, TDI-11861 retained high selectivity for sAC vs closely related tmAC family members (Figure S1). TDI-11861 did not show appreciable activity against a panel of 322 kinases36 (Table S2) and 46 other relevant drug targets such as GPCRs, ion channels, and nuclear receptors in a Eurofins safety panel (Table S3). Furthermore, TDI-11861 showed no cytotoxicity at 20 μM (200-fold above its IC50 for sAC in cells) and showed no evidence of adduct formation for 2 h in a glutathione trapping study (Table 7).37
Table 7.
Ancillary Profile of TDI-11861 (33)
| TDI-11861 (33) | |
|---|---|
| PAMPA (nm/s) | 463 |
| HPLC Log D (pH = 7.4) | 2.89 |
| kinetic solubility (μg/mL) | 79 |
| cytotoxicity | >20 μM |
| tmACs selectivity | no activity at 10 μM against ADCY1, ADCY2, ADCY5, ADCY8, ADCY9 |
| mouse plasma protein binding (fu) | 0.13 |
| glutathione reactivity | low propensity at 5 μM |
Chemistry.
Scheme 1 illustrates the synthetic route employed for 2 which exemplifies the general protocol utilized for the formation of analogues containing the key 6-chloropyrimidine-2-amino moiety. The PMB-protected iodopyrazole 2a was converted into alcohol 2b by transmetalation (i-PrMgCl-LiCL) followed by condensation of the formed organomagnesium with benzaldehyde. The alcohol in 2b was subsequently deoxygenated (TMSCl/NaI) to furnish 2c. The ester 2c was homologated into the keto-ester 2d (ethyl acetate, NaH, THF), and subsequent condensation with guanidine carbonate provided 2e. Treatment of 2e with POCl3 provided the protected PMB pyrazole 2f. Final PMB deprotection of 2f provided 2.
Scheme 1. Synthesis of 2a.

aReagents and conditions: (a) i-PrMgCl-LiCl, benzaldehyde; (b) TMSCl, NaI; (c) ethyl acetate, NaH, THF; (d) guanidine carbonate, EtOH; (e) POCl3, dioxane; (f) CAN, acetonitrile/water.
Scheme 2 illustrates the preparation of the difluoromethylpyrazole analogue 4. The PMB group in 2c is removed via treatment of TFA to afford 4a. The NH group in 4a is converted into the difluoromethyl-substituted intermediate 4b using CF2BrC(O)OEt/NaH. The ester 4b can be transformed into 4 in a similar manner to that described for 2.
Scheme 2. Synthesis of 4a.

aReagents and conditions: (a) TFA; (b) CF2BrC(O)OEt, NaH, DMF; (c) ethyl acetate, NaH, THF; (d) guanidine carbonate, EtOH; (e) POCl3, dioxane.
The synthesis of 19 is depicted in Scheme 3. The key alcohol 19b is prepared from the transmetalation of 19a (i-PrMgCl-LiCl) followed by condensation with 2-(2-morpholinoethoxy)benzaldehyde. Alcohol 19b can be converted into 19 in a similar manner to that described previously.
Scheme 3. Synthesis of 19a.

aReagents and conditions: (a) i-PrMgCl-LiCl, 2-(2-morpholinoethoxy)benzaldehyde; (b) TMSCl, NaI; (c) ethyl acetate, LHMDS, THF; (d) guanidine carbonate, EtOH; (e) POCl3, dioxane.
The preparation of 20 is depicted in Scheme 4. Iodopyrazole 19a was converted into the benzyl intermediate 20b using conditions previously outlined ((a) metalation; (b) aldehyde condensation; (c) TMSCl/NaI deoxygenation). Ester 20b was transformed into the 6-chloropyrimidine-2-amino intermediate 20e using conditions outlined ((a) keto-ester formation; (b) condensation with guanidine carbonate; (c) POCl3). The alkene in 20e was converted into the aldehyde 20g using a two-step protocol ((a) dihydroxylation; (b) periodate cleavage). Reductive amination of aldehyde 20g with the appropriate amine provided 20.
Scheme 4. Synthesis of 20a.

aReagents and conditions: (a) i-PrMgCl-LiCl, 2-(allyloxy)benzaldehyde; (b) TMSCl, NaI; (c) ethyl acetate, LHMDS, THF; (d) guanidine carbonate, EtOH; (e) POCl3, dioxane; (f) NMO, K2OsO4; (g) NaIO4; (h) NaBH3CN, MeOH, THF, 6-oxa-3-azabicyclo[3.1.1]heptane hydrochloride.
The preparation of 33 is outlined in Scheme 5. The key NH-allyloxy-substituted pyrazole 33d was prepared from 33a following those previously depicted utilizing THP to transiently protect the pyrazole NH for the metalation step. The NH pyrazole 33d was converted into the difluoromethylpyrazole intermediate 33e using diethyl-(bromodifluoromethyl)phosphonate/KF. The ester 33e was converted into the 6-chloropyrimidine-2-amino intermediate 33h using steps like those already depicted. Aldehyde 33j was prepared from alkene 33h in two steps ((a) dihydroxylation; (b) periodate cleavage). Reductive amination of aldehyde 33j with (R)-morpholine-3-ylmethanol yielded 33 (TDI-11861).
Scheme 5. Synthesis of TDI-11861 (33)a.

aReagents and conditions: (a) DHP, PTSA, THF; (b) i-PrMgCl-LiCl, 2-(allyloxy)benzaldehyde; (c) TMSCl, NaI; (d) diethyl-(bromodifluoromethyl)phosphonate, KF, MeCN; (e) ethyl acetate, LHMDS, THF; (f) guanidine carbonate, EtOH; (g) POCl3, dioxane; (h) NMO, OsO4; (i) NaIO4; (j) NaBH(OAc)3, DCE, TEA, (R)-morpholine-3-ylmethanol hydrochloride.
CONCLUSIONS
sAC inhibition represents an interesting drug target for a number of indications.38 We have previously reported a potent inhibitor of sAC (TDI-10229) with a cellular IC50 of 92 nM and reasonable druglike properties. In this study, we have detailed the design process and experimental protocols used to engineer and evaluate improved sAC affinity through structure-based drug design, leading to the identification of TDI-11861. This includes use of both SPR and jump dilution activity recovery assays to measure inhibitors with extended residence times. Ample precedent exists showing the pharmacologic benefits of inhibitors with long residence times.39–43 The use of systemically dosed sAC inhibitors as male contraceptives may represent a limiting example where, for complete fertility inhibition, long residence times are an essential compound property. This is because, upon mating, an sAC inhibitor present in sperm and semen from injected males will be diluted when the ejaculate is deposited into the female’s inhibitor-free female genital tract. These developments are of interest because novel nonhormonal contraceptive strategies for males represent a key unmet medical need.16,17
In this study, we improved residence time in a consistent manner by optimizing the intrinsic binding affinity through increased protein-ligand contacts. This was achieved through structure-based design tools leveraging new and existing crystal structures. The application of ensemble molecular docking with Glide44 and relative binding free-energy calculations with FEP+29 will be detailed in a future study. Relative to our starting point TDI-10229, TDI-11861 shows improved sAC binding affinity (3.3 nM compared to 160.0 nM), improved sAC cellular affinity (5.5 nM relative to 92 nM), long residence time (3181 s versus to 25 s), and a higher solubility (79 μg/mL compared to 1.3 μg/mL). Overall, TDI-11861 represents a next-generation sAC inhibitor with excellent potency, extended residence time, and improved druglike properties. While enhanced bioavailability or plasma half-life may need to be improved for some applications, it is a promising lead compound for further investigation. TDI-11861 enables enhanced interrogation of sAC biology and the utility of sAC inhibition in medicine.
EXPERIMENTAL SECTION
In Vitro Cyclase Activity Assay.
Assays for sAC activity using 5 nM purified protein45 were performed at 30 °C in 100 μL reactions containing 4 mM MgCl2, 2 mM CaCl2, 1 mM ATP, 40 mM NaHCO3, 50 mM Tris pH 7.5, and 3 mM DTT. Purified sAC protein was preincubated with the indicated inhibitors for 15 min at room temperature (23 °C). Each reaction was initiated with ~1,000,000 counts of α−32P labeled ATP. Generated cAMP was purified using sequential Dowex and Alumina chromatography as previously described.46
Jump Dilution.
Jump dilutions assays were performed using a modified version of the in vitro cyclase activity assay. All assays were done in the presence of 0.03% BSA and each had a final DMSO concentration of 1%. At the start of each assay, 25 nM recombinant purified GST-tagged sACt protein was preincubated with the indicated inhibitors for 15 min at room temperature (23 °C). Each inhibitor was used at a concentration 10-fold above their IC50 values. Following the preincubation period, 1 μL of the enzyme-inhibitor solution was added to 99 μL of a reaction solution containing the following: 2 mM ATP, 10 mM Mn2+, 50 mM Tris pH 7.5, 3 mM DTT, and ~4,000,000 counts of α−32P labeled ATP. After the 100-fold dilution step, each inhibitor was present at a concentration 10-fold below their IC50 values, this was done to minimize inhibitor rebinding during the reaction. The reactions were stopped every 6 min over the course of 1 h and generated cAMP was measured using the sequential chromatography method referenced above. Using Prism (GraphPad, version 9.3.1), the data was fit to the equation: , with kobs being an estimate of the dissociation rate constant (koff). To calculate vs (uninhibited enzyme velocity) and v0 (inhibited enzyme velocity), the jump dilution assay was performed in the presence of only DMSO and an excess concentration of inhibitor, respectively.
Cell-Based Cyclase Activity Assay.
Rat 4–4 cells were generated and functionally authenticated in our laboratory as previously described9 and grown in DMEM + 10% FBS. 1.25 × 106 4–4 cells were seeded per well of a 24-well plate and incubated for 24 h at 37 °C, 5% CO2. One hour before the experiment the media was aspirated and replaced with 300 μL fresh media. For 10 min, in duplicate wells, cells were preincubated with sAC inhibitor at the indicated concentrations or 0.7% DMSO as control. For cAMP accumulation, cells were incubated with 500 μM IBMX for 5 min. To stop the reaction and to lyse the cells, the media was aspirated and replaced with 250 μL 0.1 M HCl. After shaking the plate for 5 min the cell lysate was transferred to a fresh tube and centrifuged at 1000g for 5 min. The supernatant was used for cAMP quantification using the Direct cAMP Elisa kit (Enzo) following the manufacturer’s instructions.
Measuring Binding Kinetics via Surface Plasmon Resonance.
Association (kon) and dissociation rate constants (koff) for the inhibitors were obtained at 25 °C with a Biacore 8K instrument (Cytiva) using a single-cycle kinetics protocol. In PBS-P+ buffer (1 mM KH2PO4, 150 mM NaCl, 6 mM Na2HPO4, 0.05% (w/v) P20 Surfactant), 50 μg/mL of recombinant purified His-tagged sACt protein was covalently immobilized and captured on a Series S Sensor NTA chip (Cytiva) using Ni2+-His-tag chelation followed by amine coupling with a 1:1 mixture of 1-ethyl-3-(3-dimethylaminopropyl)carbodiimide and N-hydroxysuccinimide. After coupling, 1 M ethanolamine followed by 350 mM EDTA were, respectively, used to block any remaining reactive groups on the surface of the chip and to strip the Ni2+. Following chip preparation, TBS-P+ running buffer (50 mM Tris, 150 mM NaCl, and 0.05% P20 Surfactant) supplemented with 1% DMSO was flowed over the surface of the chip. After a stable baseline was obtained, five different concentrations of the indicated inhibitors were sequentially injected into a single channel for 120 s at a flow rate of 50 μL/min and then, at the same flow rate, allowed to dissociate for 600 s in the presence of running buffer. For TDI-10229, the following concentrations were used: 5000, 1250, 312.5, 78.125, and 19.53125 nM. For all other inhibitors, the following concentrations were used: 1000, 250, 62.5, 15.625, and 4 nM. All experiments were performed in parallel in reference channels lacking immobilized protein. To process the collected data, responses from the reference channels were subtracted from the responses from the active channels. From the reference-subtracted data, fitted curves and kd and k values were determined using the Biacore 8K Insight Evaluation Software Version 2.0 (Cytiva) using a 1:1 binding kinetics model.
Log D Determination.
Log D 7.4, which is a partition coefficient between 1-octanol and aqueous buffer pH 7.4, of the compounds was measured using the chromatographic procedure whose condition was developed based on a published method.47,48
Parallel Artificial Membrane Permeability Assay (PAMPA).
Donor wells were filled with 200 μL of PRISMA HT buffer (pH 7.4, pION, Inc.) containing 10 μM test compound. The filter on the bottom of each acceptor well was coated with 4 μL of a GIT-0 Lipid Solution (pION, Inc.) and filled with 200 μL of Acceptor Sink Buffer (pION, Inc.). The acceptor filter plate was put on the donor plate and incubated for 3 h at room temperature. After the incubation, the amount of test compound in both the donor and acceptor wells was measured by LC/MS/MS.
Kinetic Solubility.
Small volumes of compound solution dissolved in DMSO were added to the aqueous buffer solution. After incubation, precipitates were separated by filtration. The solubility was determined by UV absorbance of each filtrate.
GSH (Glutathione) Reactivity Assay.
Two microliters of 1 mM compound are spiked into 398 μL of 0.1 M potassium phosphate buffer containing 5 mM GSH to achieve a final compound concentration of 5 μM. A similar buffer solution was prepared minus GSH that was used as the compound control sample. The samples were incubated at 37 °C, and an aliquot (50 μL) was taken at different time points (0, 10, 30, 60, and 120 min) and quenched with cold acetonitrile (400 μL) containing internal standards and ethyl maleimide solution (50 μL of a 20 mM solution in acetonitrile). The samples were vortexed, and the supernatants were subjected to LCMS analysis. The remaining percentage at each time point was determined using the peak area ratios from the extracted ion chromatograms. The remaining percentage at t min (%) = peak area ratio t min/peak area ratio 0 min × 100.
Metabolic Stability.
Liver microsomes were purchased from Sekisui XenoTech, LLC. (Kansas City, KS). The microsomes (0.2 mg protein/mL) and the compounds (1 μM) were mixed in phosphate buffer (pH 7.4). The reactions were initiated by adding an NADPH-generating system (a mixture of MgCl2, β-NADP+, glucose-6-phosphate, and glucose-6-phosphate dehydrogenase) to the mixtures before incubation. Incubations were conducted at 37 °C and terminated by adding acetonitrile. The zero-time incubations, which served as the controls, were terminated by adding acetonitrile before adding an NADPH-generating system. After the samples were mixed and centrifuged, the compound concentration in the supernatant fractions was measured by LC/MS/MS.
In Vivo Pharmacokinetic Analysis in Mice.
The free base of TDI-11861 (33) was dosed in adult, male CD-1 mice (n = 3 each arm). The IV dose in mice (2 mg/kg) was administered as a solution at 0.4 mg/mL in 20% HPBCD. The PO doses in mice (10 and 50 mpk) were administered as a solution at 2 and 10 mg/mL concentrations, respectively, in DMSO/PEG400/PBS (10:40:50). The dosing volume for all studies in mice was 5 mL/kg. Serial blood samples were collected after dosing at selected time points up to 24 h. After centrifugation to harvest plasma, the concentrations of TDI-11861 (33) were determined by liquid chromatography coupled with tandem mass spectrometry. The standard PK parameters in mice (Cl, T1/2, AUC, and F) were calculated using the 2 mpk IV and 10 mpk PO arms. The reported mean values were calculated from individual animals in each arm. All animal studies included as part of this manuscript were performed at Pharmaron in accordance with guidelines defined by “Institutional Animal Care and Use Committee (IACUC)”.
Crystal Structure Determination.
The protein construct comprising the catalytic domains of human sAC (hsAC-cat) was expressed, purified, and crystallized as described,49 with a micro-seeding step added to improve the reproducibility of high-quality crystal growth. In short, hsAC-cat with a C-terminal His-tag was expressed in insect cells; purified through affinity, ion exchange, and size exclusion chromatography; and crystallized in hanging drops with 0.1 M sodium acetate pH 5.0, 0.2 M trisodium citrate, 14% (w/v) PEG 4000, and 10% (v/v) glycerol as reservoir solution. Crystals were transferred to a drop containing 0.1 M sodium acetate pH 5.0, 0.2 M trisodium citrate, 15% (w/v) PEG 4000, 20% (v/v) glycerol, 10% DMSO (v/v), soaked with 5 mM ligand for 24 h, and flash-frozen in liquid nitrogen.
Diffraction data were collected at 100 K at BESSY beamline 14.1 operated by Helmholtz-Zentrum Berlin50 and processed with XDSapp.51 Phases were determined through Patterson searches with Phaser52 using hsAC apo (PDB ID 4CLL) as a search model. The crystallographic model was then rebuilt in Coot53 and refined with Phenix.54
EXPERIMENTAL SECTION
Synthetic Materials and Methods.
Reagents and solvents were obtained from commercial sources and used without further purification. 1H NMR spectra (400 MHz) were collected on a Varian spectrometer. 13C NMR spectra (126 MHz) were collected on a Bruker spectrometer. Chemical shifts are reported in ppm relative to residual solvent peak in the indicated solvent, and for 1H NMR spectra, multiplicities, coupling constants in hertz, and numbers of protons are indicated. Purities of all reported compounds were greater than 95% based on HPLC chromatograms obtained on an Agilent 1200 LCMS system. The Supporting Information contains details surrounding the preparation of 5–14, 16–18, and 21–32.
Preparation of 2. Ethyl 4-[hydroxyl(phenyl)methyl]-1-[(4-methoxyphenyl)methyl]pyrazole-3-carboxylate.
To a mixture of ethyl 4-iodo-1-[(4-methoxyphenyl)methyl]pyrazole-3-carboxylate (3.0 g, 7.8 mmol, 1 equiv) in THF (30 mL) was added i-PrMgCl-LiCl (1.3 M, 6.3 mL, 1.05 equiv) dropwise at −15 °C under N2. After stirring at −15 °C for 30 min, benzaldehyde (907 mg, 8.55 mmol, 864 μL, 1.1 equiv) was added to the mixture dropwise at −15 °C. The resulting reaction mixture was stirred at 15 °C for 12 h under N2. The reaction mixture was quenched with saturated aqueous NH4Cl solution (100 mL). The mixture was extracted with ethyl acetate (80 mL × 3). The combined organic layers were washed with brine (100 mL), dried over Na2SO4, and filtered. The filtrate was concentrated under reduced pressure. The residue was purified by flash chromatography (ISCO; 80 g SepaFlash Silica Flash Column, gradient elution of 0–25% ethyl acetate/petroleum ether @ 100 mL/min) which furnished the title compound (1.9 g, 5.19 mmol, 67%). 1H NMR: (400 MHz, chloroform-d) δ 7.39–7.17 (m, 5H), 7.09 (br d, J = 8.2 Hz, 2H), 6.91–6.69 (m, 3H), 5.92 (br d, J = 4.0 Hz, 1H), 5.17 (s, 2H), 4.64 (br d, J = 4.6 Hz, 1H), 4.48–4.28 (m, 2H), 3.73 (s, 3H), 1.35 (br t, J = 7.0 Hz, 3H).
Ethyl 4-benzyl-1-[(4-methoxyphenyl)methyl]pyrazole-3-carboxylate.
To a solution of NaI (4.66 g, 31.1 mmol, 6 equiv) in MeCN (20 mL) was added TMSCl (3.38 g, 31.1 mmol, 3.95 mL, 6 equiv) under N2. After stirring at 15 °C for 10 min, a solution of ethyl 4-[hydroxyl(phenyl)methyl]-1-[(4-methoxyphenyl)methyl]pyrazole-3-carboxylate (1.9 g, 5.2 mmol, 1 equiv) in MeCN (10 mL) was added. The mixture was stirred at 15 °C for 2 h under N2. The reaction mixture was quenched with saturated, aqueous Na2SO3 solution (150 mL). The mixture was extracted with ethyl acetate (80 mL × 3). The combined organic layers were washed with brine (80 mL), dried over Na2SO4, and filtered. The filtrate was concentrated under reduced pressure. The residue was purified by gradient flash chromatography (ISCO; 20 g SepaFlash Silica Flash Column, gradient elution of 0–20% ethyl acetate/petroleum ether @ 75 mL/min) which furnished the title compound (1.6 g, 4.6 mmol, 88%).
Ethyl 3-[4-benzyl-1-[(4-methoxyphenyl)methyl]pyrazol-3-yl]-3-oxo-propanoate.
A mixture of ethyl 4-benzyl-1-[(4-methoxyphenyl)-methyl]pyrazole-3-carboxylate (390 mg, 1.11 mmol, 1 equiv) in THF (10 mL) was cooled to 0 °C and then NaH (134 mg, 3.34 mmol, 60 wt % dispersion in oil, 3 equiv) was added. After stirring for 20 min, ethyl acetate (686 mg, 7.79 mmol, 0.76 mL, 7 equiv) was added dropwise at 0 °C. The mixture was stirred at 70 °C for 2 h under N2. The reaction mixture was quenched with saturated NH4Cl solution (80 mL). The aqueous phase was extracted with ethyl acetate (80 mL × 3). The combined organic layers were washed with brine (100 mL), dried over Na2SO4, and filtered. The filtrate was concentrated under reduced pressure. The residue was purified by flash silica gel chromatography (ISCO; 12 g SepaFlash Silica Flash Column, eluent of 0–15% ethyl acetate/petroleum ether gradient @ 45 mL/min) to furnish the title compound (250 mg, 57%) as a yellow oil. 1H NMR: (400 MHz, DMSO-d6) δ 7.64 (s, 1H), 7.31–7.13 (m, 7H), 6.96–6.85 (m, 2H), 5.27 (s, 2H), 4.06 (q, J = 7.1 Hz, 2H), 4.01 (s, 2H), 3.96 (s, 2H), 3.74–3.69 (m, 3H), 1.13 (t, J = 7.1 Hz, 3H).
2-Amino-4-[4-benzyl-1-[(4-methoxyphenyl)methyl]pyrazol-3-yl]-1H-pyrimidin-6-one.
To a solution of ethyl 3-[4-benzyl-1-[(4-methoxyphenyl)methyl]pyrazol-3-yl]-3-oxo-propanoate (250 mg, 0.637 mmol, 1 equiv) in EtOH (5 mL) was added guanidine carbonate (172 mg, 0.956 mmol, 1.5 equiv). The mixture was stirred for 24 h at 85 °C under N2. The reaction mixture was diluted with water (20 mL) and then concentrated under reduced pressure to remove the EtOH. The solution was adjusted to pH = 5 via addition of aqueous HCl (2 N). The mixture was filtered, and the residue was dried under reduced pressure to furnish the title compound (150 mg, 61%) as a white solid.
4-[4-Benzyl-1-[(4-methoxyphenyl)methyl]pyrazol-3-yl]-6-chloropyrimidin-2-amine.
To a solution of 2-amino-4-[4-benzyl-1-[(4-methoxyphenyl)methyl]pyrazol-3-yl]-1H-pyrimidin-6-one (150 mg, 0.387 mmol, 1 equiv) in dioxane (3 mL) was added POCl3 (890 mg, 5.81 mmol, 0.54 mL, 15 equiv) dropwise. The reaction mixture was stirred at 75 °C for 12 h under N2. The reaction mixture was quenched with saturated NaHCO3 solution (80 mL). The organic layer was separated, and aqueous phase was extracted with ethyl acetate (30 mL × 3). The combined organic layers were washed with brine (60 mL), dried over Na2SO4, and filtered. The filtrate was concentrated under reduced pressure. The residue was purified by flash silica gel chromatography (ISCO; 12 g SepaFlash Silica Flash Column, eluent of 0–25% ethyl acetate/petroleum ether gradient @ 45 mL/min) to furnish the title compound (40 mg, 0.099 mmol, 25% yield) as a yellow oil.
4-(4-Benzyl-1H-pyrazol-3-yl)-6-chloro-pyrimidin-2-amine (2).
To a solution of 4-[4-benzyl-1-[(4-methoxyphenyl)methyl]pyrazol-3-yl]-6-chloro-pyrimidin-2-amine (30 mg, 0.074 mmol, 1 equiv) in MeCN (1 mL) and H2O (1 mL) was added CAN (122 mg, 0.222 mmol, 3 equiv) at 0 °C. The reaction mixture was stirred at 0 °C for 0.5 h and then warmed to 20 °C. The reaction mixture was stirred at 20 °C for 12 h. The reaction mixture was quenched with saturated aqueous NaHCO3 solution (5 mL), diluted with H2O (20 mL), and extracted with ethyl acetate (30 mL × 3). The combined organic layers were washed with brine (60 mL), dried over Na2SO4, and filtered. The filtrate was concentrated under reduced pressure. The residue was purified by preparative-HPLC (column: Waters Xbridge 150 × 25 mm, 5 μm; mobile phase: [water (10 mM NH4HCO3)-ACN]; B%: 35–55%, 10 min) which furnished the title compound 2 as a white solid (4.4 mg, 21%). 1H NMR: (400 MHz, DMSO-d6) δ 7.55 (s, 1H), 7.31–7.18 (m, 4H), 7.16–7.06 (m, 3H), 7.04 (s, 1H), 4.28 (s, 2H); LCMS: (MH+) 286.1.
Preparation of 4. Ethyl 4-benzyl-1H-pyrazole-3-carboxylate.
Ethyl 4-benzyl-1-[(4-methoxyphenyl)methyl]pyrazole-3-carboxylate (1.53 g, 4.37 mmol, 1 equiv) was dissolved in TFA (20 mL). The mixture was stirred at 85 °C for 12 h. The reaction mixture was concentrated under reduced pressure to remove TFA. The reaction mixture was diluted with H2O (80 mL) and extracted with ethyl acetate (80 mL × 3). The combined organic layers were washed with brine (60 mL), dried over Na2SO4, and filtered. The filtrate was concentrated under reduced pressure. The residue was purified by flash chromatography (ISCO; 20 g SepaFlash Silica Flash Column, gradient elution of 0–20% ethyl acetate/petroleum ether @ 100 mL/min), which furnished the title compound (950 mg, 62%) as a white solid. 1H NMR: (400 MHz, DMSO-d6) δ 13.82–13.17 (m, 1H), 7.69–7.41 (m, 1H), 7.33–7.13 (m, 5H), 4.33–4.18 (m, 2H), 4.04 (br s, 2H), 1.33–1.22 (m, 3H).
4-Benzyl-1-(difluoromethyl)pyrazole-3-carboxylate.
A mixture of ethyl 4-benzyl-1H-pyrazole-3-carboxylate (830 mg, 3.60 mmol, 1 equiv) in DMF (10 mL) was cooled to 0 °C. Sodium hydride (433 mg, 10.8 mmol, 60 wt % dispersion in oil, 3 equiv) was added. After stirring for 20 min, ethyl 2-bromo-2,2-difluoro-acetate (878 mg, 4.33 mmol, 0.556 mL, 1.2 equiv) was added dropwise at 0 °C. The mixture was stirred at 15 °C for 2 h under N2. The reaction mixture was quenched with saturated, aqueous NH4Cl (80 mL). The mixture was extracted with ethyl acetate (60 mL × 3). The combined organic layers were washed with brine (50 mL), dried over Na2SO4, and filtered. The filtrate was concentrated under reduced pressure. The residue was purified by flash chromatography (ISCO; 12 g SepaFlash Silica Flash Column, gradient elution of 0–3% ethyl acetate/petroleum ether @ 36 mL/min) which furnished the title compound (220 mg, 0.78 mmol, 22%) as a yellow oil. 1H NMR: (400 MHz, DMSO-d6) δ 8.13 (s, 1H), 8.02–7.70 (m, 1H), 7.31–7.26 (m, 2H), 7.24–7.16 (m, 3H), 4.28 (q, J = 6.7 Hz, 2H), 4.08–4.04 (m, 2H), 1.28–1.23 (m, 3H).
Ethyl 3-[4-benzyl-1-(difluoromethyl)pyrazol-3-yl]-3-oxo-propanoate.
A mixture of ethyl acetate (396 mg, 4.50 mmol, 0.44 mL, 7 equiv) in THF (2 mL) was cooled to 0 °C. Sodium hydride (31 mg, 0.77 mmol, 60 wt % dispersion in oil, 1.2 equiv) was added. After stirring for 2 min, ethyl 4-benzyl-1-(difluoromethyl)pyrazole-3-carboxylate (180 mg, 0.64 mmol, 1 equiv) in THF (1 mL) was added dropwise at 0 °C. The mixture was stirred at 50 °C for 1 h under N2. The reaction mixture was quenched with saturated aqueous NH4Cl (20 mL). The mixture was extracted with ethyl acetate (30 mL × 3). The combined organic layers were washed with brine (50 mL), dried over Na2SO4, and filtered. The filtrate was concentrated under reduced pressure. The residue was purified by flash silica gel chromatography (ISCO; 4 g SepaFlash Silica Flash Column, eluent of 0–3% ethyl acetate/petroleum ether gradient at 35 mL/min) to furnish the title compound (100 mg, 49%) as a yellow oil.
2-Amino-4-[4-benzyl-1-(difluoromethyl)pyrazol-3-yl]-1H-pyrimidin-6-one.
To a solution of ethyl 3-[4-benzyl-1-(difluoromethyl)-pyrazol-3-yl]-3-oxo-propanoate (80 mg, 0.25 mmol, 1 equiv) in EtOH (2 mL) was added guanidine carbonate (134 mg, 0.745 mmol, 3 equiv). The mixture was stirred at 85 °C for 12 h under N2. The reaction mixture was diluted with H2O (10 mL) and concentrated under reduced pressure to remove EtOH. The mixture was extracted with ethyl acetate (15 mL × 3). The combined organic layers were washed with brine (30 mL), dried over Na2SO4, and filtered. The filtrate was concentrated under reduced pressure to furnish the title compound (80 mg, crude, ~100%) as a white solid.
4-[4-Benzyl-1-(difluoromethyl)pyrazol-3-yl]-6-chloro-pyrimidin-2-amine (4).
POCl3 (507 mg, 3.31 mmol, 0.31 mL, 15 equiv) was added into a solution of 2-amino-4-[4-benzyl-1-(difluoromethyl)-pyrazol-3-yl]-1H-pyrimidin-6-one (70 mg, 0.22 mmol, 1 equiv) in dioxane (2 mL) at 15 °C. Then the mixture was heated to 75 °C and stirred for 2 h under N2. The reaction mixture was adjusted to pH 7 by the addition of a NaOH solution (1 N). The reaction mixture was extracted with ethyl acetate (30 mL × 3). The combined organic layers were washed with brine (60 mL), dried over Na2SO4, and filtered. The filtrate was concentrated under reduced pressure. The residue was purified by prep-HPLC (column: Phenomenex Gemini-NX 150 × 30 mm × 5 μm; mobile phase: [water (10 mM NH4HCO3)-ACN]; B%: 38–68%, 6 min) to furnish the title compound 4 (14 mg, 0.041 mmol, 19% yield) as a white solid. 1H NMR: (400 MHz, DMSO-d6) δ 8.05 (s, 1H), 7.97–7.65 (m, 1H), 7.32–7.22 (m, 6H), 7.20–7.13 (m, 1H), 7.01 (s, 1H), 4.29 (s, 2H); LCMS: (MH+) 336.1.
Preparation of 19. 4-[Hydroxy-[2-(2-morpholinoethoxy)phenyl]-methyl]-1-methyl-pyrazole-3-carboxylate.
To a solution of methyl 4-iodo-1-methyl-pyrazole-3-carboxylate (1.35 g, 5.07 mmol, 1 equiv) in THF (15 mL) was added isopropropylmagnesium chloride–lithium chloride complex (1.3 M, 4.3 mL, 1.1 equiv) at −10 °C under N2. After stirring at −10 °C for 0.5 h, a solution of 2-(2-morpholinoethoxy)benzaldehyde (1.40 g, 5.95 mmol, 1.17 equiv) in THF (5 mL) was added to the mixture dropwise. After the addition, the mixture was allowed to warm slowly to 25 °C and stirred at 25 °C for 12 h. The reaction mixture was diluted with sat. NH4Cl solution (100 mL), and the mixture was extracted with ethyl acetate (50 mL × 3). The organic layer was washed with brine (70 mL), dried over Na2SO4, and filtered. The filtrate was concentrated under reduced pressure. The residue was purified by column chromatography (SiO2, petroleum ether/ethyl acetate = 1:1 to 1:0 followed by 5% MeOH as an additive) to furnish the title compound (800 mg, 2.13 mmol, 42% yield) as a yellow oil. 1H NMR: (chloroform-d, 400 MHz) δ 7.26 (s, 1H), 7.17 (dt, J = 1.7, 7.8 Hz, 1H), 7.08 (dd, J = 1.4, 7.8 Hz, 1H), 6.87–6.82 (m, 2H), 6.44 (s, 1H), 4.21–4.09 (m, 2H), 3.88 (s, 3H), 3.80 (s, 3H), 3.62 (s, 4H), 2.69–2.62 (m, 2H), 2.46–2.36 (m, 4H).
Methyl 1-methyl-4-[[2-(2-morpholinoethoxy)phenyl]methyl]-pyrazole-3-carboxylate.
To a solution of NaI (2.87 g, 19.2 mmol, 6 equiv) in MeCN (10 mL) was added TMSCl (2.08 g, 19.2 mmol, 2.43 mL, 6 equiv) under N2. After 10 min of stirring at 25 °C, a solution of methyl 4-[hydroxy-[2-(2-morpholinoethoxy)phenyl]-methyl]-1-methyl-pyrazole-3-carboxylate (1.20 g, 3.20 mmol, 1 equiv) in MeCN (6 mL) was added. The mixture was stirred at 25 °C under N2 for 1 h. The reaction mixture was diluted with sat. aq. Na2SO3 (70 mL). The solution was extracted with ethyl acetate (50 mL × 3). The combined organic layer was washed with brine (100 mL), dried over Na2SO4, and filtered. The filtrate was concentrated under reduced pressure to remove the solvent. The residue was purified by column chromatography (SiO2, petroleum ether/ethyl acetate = 1:1 to 1:0 and then 5% MeOH additive) to furnish the title compound (1.1 g, 3.1 mmol, 96% yield) as a yellow oil. 1H NMR: (chloroform-d, 400 MHz) δ 7.24–7.15 (m, 2H), 6.99 (s, 1H), 6.95–6.84 (m, 2H), 4.24 (br t, J = 4.8 Hz, 2H), 4.10 (s, 2H), 3.93 (s, 3H), 3.88 (s, 3H), 3.76 (br d, J = 4.0 Hz, 4H), 2.92 (br s, 2H), 2.69 (br s, 4H).
Ethyl 3-[1-methyl-4-[[2-(2-morpholinoethoxy)phenyl]methyl]-pyrazol-3-yl]-3-oxo-propanoate.
To a solution of methyl 1-methyl-4-[[2-(2-morpholinoethoxy)phenyl]methyl]pyrazole-3-carboxylate (990 mg, 2.75 mmol, 1 equiv) and ethyl acetate (1.70 g, 19.3 mmol, 1.89 mL, 7 equiv) in THF (10 mL) was added LiHMDS (1 M, 8.26 mL, 3 equiv) at −40 °C quickly in one portion. The mixture was stirred at −40 °C for 2 h under N2. The reaction mixture was diluted with sat. NH4Cl solution (100 mL), and the mixture was extracted with ethyl acetate (50 mL × 3). The organic layer was washed with brine (90 mL), dried over Na2SO4, and filtered. The filtrate was concentrated under reduced pressure. The residue was purified by column chromatography (SiO2, petroleum ether/ethyl acetate = 1:0) to furnish the title compound (900 mg, 79%) as a yellow oil. 1H NMR: (chloroform-d, 400 MHz). δ 7.20 (d, J = 7.5 Hz, 2H), 6.99 (s, 1H), 6.92–6.83 (m, 2H), 4.22 (d, J = 7.1 Hz, 2H), 4.15–4.10 (m, 4H), 4.04 (s, 2H), 3.84 (s, 3H), 3.71–3.65 (m, 4H), 2.78 (t, J = 5.6 Hz, 2H), 2.57–2.48 (m, 4H), 1.31–1.26 (m, 3H).
2-Amino-4-[1-methyl-4-[[2-(2-morpholinoethoxy)phenyl]-methyl]pyrazol-3-yl]-1H-pyrimidin-6-one.
Ethyl 3-[1-methyl-4-[[2-(2-morpholinoethoxy)phenyl]methyl]pyrazol-3-yl]-3-oxo-propanoate (800 mg, 1.93 mmol, 1 equiv) and guanidine carbonate (1.04 g, 5.78 mmol, 3 equiv) were taken up in anhydrous EtOH (16 mL). The mixture was stirred for 36 h at 85 °C under N2 during which time a white precipitate formed. The reaction mixture was diluted with water (10 mL) and then concentrated under the reduced pressure to remove the EtOH. The pH of the solution was adjusted to 5 by the addition of aqueous HCl (4 N). Then, the pH of the solution was adjusted to 9 by the addition of aqueous NH3. The solution was filtered, and the residue was dried under reduced pressure to afford the title compound (400 mg, 51%) as a light, yellow solid.
4-Chloro-6-[1-methyl-4-[[2-(2-morpholinoethoxy)phenyl]-methyl]pyrazol-3-yl]pyrimidin-2-amine (19).
POCl3 (2.24 g, 14.6 mmol, 1.36 mL, 15 equiv) was added into a solution of 2-amino-4-[1-methyl-4-[[2-(2-morpholinoethoxy)phenyl]methyl]pyrazol-3-yl]-1H-pyrimidin-6-one (400 mg, 0.975 mmol, 1 equiv) in dioxane (8 mL) at 25 °C. The mixture was heated to 75 °C for 16 h. The reaction mixture was added slowly to an aq. NaHCO3 solution (saturated, 110 mL) to quench the excess POCl3. The solution was extracted with ethyl acetate (60 mL × 5). The organic layer was washed with brine (70 mL), dried over Na2SO4, and filtered. The filtrate was concentrated under reduced pressure. The residue was purified by prep-HPLC (column: Phenomenex Synergi C18 150 × 25 × 10 μm; mobile phase: [water (0.2% formic acid)-ACN]; B%: 20–50%, 10 min) to furnish the title compound (157 mg, formic acid salt, 34%) 19 as an off-white solid. 1H NMR: (DMSO-d6, 400 MHz). δ 8.13 (s, 1H), 7.26 (s, 1H), 7.21 (dd, J = 1.6, 7.4 Hz, 1H), 7.19–7.12 (m, 1H), 7.06 (br s, 2H), 6.98 (s, 1H), 6.95 (d, J = 8.0 Hz, 1H), 6.84 (t, J = 7.3 Hz, 1H), 4.20 (s, 2H), 4.05 (t, J = 5.6 Hz, 2H), 3.80 (s, 3H), 3.53–3.48 (m, 4H), 2.63 (br t, J = 5.3 Hz, 2H), 2.40 (br s, 4H); 13C NMR (126 MHz, DMSO) δ 163.83, 163.68, 160.42, 158.42, 158.18, 144.21, 132.56, 130.70, 129.91, 127.91, 122.17, 121.01, 116.64, 112.18, 104.57, 66.43, 53.96, 40.59, 40.50, 40.43, 40.34, 40.26, 40.17, 40.09, 40.00, 39.92, 39.83, 39.67, 39.50, 39.44, 25.67; LCMS: (M + H+): 429.2. HRMS calcd for C21H26N6ClO2 (M + H+) 429.1801; found: 429.1800.
Preparation of 20. 4-[(2-Allyloxyphenyl)-hydroxy-methyl]-1-methyl-pyrazole-3-carboxylate.
To a solution of methyl 4-iodo-1-methyl-pyrazole-3-carboxylate (5.00 g, 18.8 mmol, 1 equiv) in THF (50 mL) was added i-PrMgCl-LiCl (1.3 M, 15.2 mL, 1.05 equiv) at −15 °C. The mixture was stirred at −15 °C for 0.5 h under N2. 2-Allyloxybenzaldehyde (3.05 g, 18.8 mmol, 1 equiv) in THF (5 mL) was added to the mixture at −15 °C. The whole mixture was stirred at 25 °C for another 11.5 h under N2. The reaction mixture was poured into a saturated ammonium chloride solution (150 mL). The mixture was extracted with ethyl acetate (50 mL × 3). The organic phase was washed with brine (80 mL), dried over anhydrous Na2SO4, and filtered. The filtrate was concentrated under reduced pressure. The residue was purified by flash silica gel chromatography (ISCO; 80 g SepaFlash Silica Flash Column, eluent of 0–50% ethyl acetate/petroleum ether gradient @ 100 mL/min) to furnish the title compound (3.80 g, 12.6 mmol, 67% yield) as a light-yellow oil. 1H NMR: (400 MHz, chloroform-d) δ 7.53 (dd, J = 1.2, 7.5 Hz, 1H), 7.26–7.22 (m, 1H), 7.00 (t, J = 7.5 Hz, 1H), 6.95 (s, 1H), 6.86 (d, J = 8.2 Hz, 1H), 6.39 (d, J = 3.9 Hz, 1H), 6.05–5.91 (m, 1H), 5.34–5.18 (m, 2H), 4.77 (br d, J = 4.6 Hz, 1H), 4.53 (d, J = 5.1 Hz, 2H), 3.96 (s, 3H), 3.85 (s, 3H).
Methyl 4-[(2-allyloxyphenyl)methyl]-1-methyl-pyrazole-3-carboxylate.
To a solution of NaI (10.1 g, 67.5 mmol, 6 equiv) in MeCN (15 mL) was added TMSCl (7.33 g, 67.5 mmol, 8.56 mL, 6 equiv) under N2. The mixture was stirred at 25 °C for 10 min. Methyl 4-[(2-allyloxyphenyl)-hydroxy-methyl]-1-methyl-pyrazole-3-carboxylate (3.4 g, 11.3 mmol, 1 equiv) was dissolved in MeCN (20 mL) and added to the mixture. The whole mixture was stirred at 25 °C for 1 h under N2. The reaction mixture was poured into saturated sodium sulfite solution (150 mL). The mixture was extracted with ethyl acetate (50 mL × 3). The organic phase was washed with brine (80 mL), dried over anhydrous Na2SO4, and filtered. The filtrate was concentrated under reduced pressure. The residue was purified by flash silica gel chromatography (ISCO; 40 g SepaFlash Silica Flash Column, eluent of 0–20% ethyl acetate/petroleum ether gradient @ 75 mL/min) to furnish the title compound (3.1 g, 96%) as a yellow oil. 1H NMR: (400 MHz, chloroform-d) δ 7.23–7.14 (m, 2H), 7.04 (s, 1H), 6.92–6.82 (m, 2H), 6.03 (tdd, J = 5.1, 10.4, 17.2 Hz, 1H), 5.37 (qd, J = 1.5, 17.3 Hz, 1H), 5.25 (dd, J = 1.4, 10.6 Hz, 1H), 4.55 (td, J = 1.5, 5.0 Hz, 2H), 4.14 (s, 2H), 3.92 (s, 3H), 3.86 (s, 3H).
Ethyl 3-[4-[(2-allyloxyphenyl)methyl]-1-methyl-pyrazol-3-yl]-3-oxo-propanoate.
To a solution of methyl 4-[(2-allyloxyphenyl)-methyl]-1-methyl-pyrazole-3-carboxylate (3.10 g, 10.8 mmol, 1 equiv) and EtOAc (6.68 g, 75.8 mmol, 7.42 mL, 7 equiv) in THF (30 mL) was added LiHMDS (1 M, 32.5 mL, 3 equiv) at −40 °C quickly in one portion. The mixture was stirred at −40 °C for 1 h under N2. The reaction mixture was poured into a saturated, aqueous ammonium chloride solution (200 mL). The mixture was extracted with ethyl acetate (50 mL × 3). The organic phase was washed with brine (80 mL), dried over anhydrous Na2SO4, and filtered. The filtrate was concentrated under reduced pressure. The residue was purified by flash silica gel chromatography (ISCO; 40 g SepaFlash Silica Flash Column, eluent of 0–15% ethyl acetate/petroleum ether gradient @ 100 mL/min) to furnish the title compound (3.5 g, 95%) as a yellow oil. 1H NMR: (400 MHz, chloroform-d). δ 7.24 (dd, J = 1.5, 7.4 Hz, 1H), 7.18 (dt, J = 1.8, 7.8 Hz, 1H), 7.01 (s, 1H), 6.91–6.83 (m, 2H), 6.04 (tdd, J = 5.1, 10.4, 17.2 Hz, 1H), 5.36 (qd, J = 1.6, 17.3 Hz, 1H), 5.25 (dd, J = 1.4, 10.6 Hz, 1H), 4.55 (td, J = 1.5, 5.0 Hz, 2H), 4.25–4.19 (m, 2H), 4.15 (s, 2H), 4.04 (s, 2H), 3.83 (s, 3H), 1.30–1.26 (m, 3H).
4-[4-[(2-Allyloxyphenyl)methyl]-1-methyl-pyrazol-3-yl]-2-amino-1H-pyrimidin-6-one.
To a solution of ethyl 3-[4-[(2-allyloxyphenyl)-methyl]-1-methyl-pyrazol-3-yl]-3-oxo-propanoate (3.50 g, 10.2 mmol, 1 equiv) in EtOH (30 mL) was added guanidine carbonate (5.53 g, 30.7 mmol, 3 equiv). The mixture was stirred at 85 °C for 12 h under N2. The reaction mixture was diluted with water (10 mL), and the mixture concentrated under reduced pressure to remove the EtOH. The yellow precipitate formed was collected while the pH of the remaining aqueous phase was adjusted to 5 by the addition of aq. HCl solution (1 N). The combined yellow precipitate was triturated with EtOAc/MeOH (5:1, 10 mL). The solid was collected by filtration and dried under reduced pressure to furnish the title compound (1.0 g, 29%) as a yellow solid. The material was used directly in the next step without further purification. LCMS: (M + H+): 338.1.
4-[4-[(2-Allyloxyphenyl)methyl]-1-methyl-pyrazol-3-yl]-6-chloropyrimidin-2-amine.
To a solution of 4-[4-[(2-allyloxyphenyl)-methyl]-1-methyl-pyrazol-3-yl]-2-amino-1H-pyrimidin-6-one (900 mg, 2.67 mmol, 1 equiv) in dioxane (10 mL) was added POCl3 (6.14 g, 40.0 mmol, 3.72 mL, 15 equiv). The mixture was stirred at 75 °C for 12 h under N2. The reaction mixture was concentrated under reduced pressure to remove POCl3. The mixture was dissolved in EtOAc (30 mL), and the resulting solution was added slowly to a saturated, aqueous sodium bicarbonate solution (150 mL). The mixture was extracted with ethyl acetate (50 mL × 3). The organic phase was washed with brine (80 mL), dried over anhydrous Na2SO4, and filtered. The filtrate was concentrated under reduced pressure. The residue was purified by flash silica gel chromatography (ISCO; 12 g SepaFlash Silica Flash Column, eluent of 0–30% ethyl acetate/petroleum ether gradient @ 75 mL/min) to furnish the title compound as (0.7 g, 74%) as a yellow solid. 1H NMR: (400 MHz, chloroform-d) δ 7.24 (s, 1H), 7.21–7.11 (m, 2H), 7.07 (s, 1H), 6.90–6.85 (m, 2H), 6.04 (tdd, J = 5.1, 10.4, 17.2 Hz, 1H), 5.38 (qd, J = 1.6, 17.3 Hz, 1H), 5.25 (qd, J = 1.4, 10.6 Hz, 1H), 5.12 (br s, 2H), 4.57 (td, J = 1.5, 5.0 Hz, 2H), 4.26 (s, 2H), 3.88 (s, 3H).
3-[2-[[3-(2-Amino-6-chloro-pyrimidin-4-yl)-1-methyl-pyrazol-4-yl]methyl]phenoxy]propane-1,2-diol.
To a solution of 4-[4-[(2-allyloxyphenyl)methyl]-1-methyl-pyrazol-3-yl]-6-chloro-pyrimidin-2-amine (0.75 g, 2.1 mmol, 1 equiv) in THF (48 mL) and H2O (6 mL) was added NMO (740 mg, 6.32 mmol, 3 equiv) and K2OsO4 dihydrate (78 mg, 0.21 mmol, 0.1 equiv) at 25 °C. The mixture was stirred at 25 °C for 12 h under N2. The reaction mixture was poured into a saturated, aqueous sodium sulfite solution (150 mL). The mixture was extracted with ethyl acetate (50 mL × 3). The organic phase was washed with brine (80 mL), dried over anhydrous Na2SO4, and filtered. The filtrate was concentrated under reduced pressure. The residue was purified by flash silica gel chromatography (ISCO; 12 g SepaFlash Silica Flash Column, eluent of 0–100% ethyl acetate/petroleum ether (additive 3% MeOH) gradient @ 70 mL/min) to furnish the title compound (700 mg, 86%) as a light-yellow solid. 1H NMR: (400 MHz, DMSO-d6) δ 7.53 (s, 1H), 7.18 (d, J = 7.5 Hz, 1H), 7.15–7.09 (m, 1H), 7.06 (s, 2H), 6.98 (s, 1H), 6.92 (d, J = 8.1 Hz, 1H), 6.78 (t, J = 7.4 Hz, 1H), 4.99 (d, J = 5.0 Hz, 1H), 4.66 (t, J = 5.7 Hz, 1H), 4.25 (s, 2H), 4.02–3.98 (m, 1H), 3.92–3.86 (m, 1H), 3.85–3.81 (m, 1H), 3.81 (s, 3H), 3.48–3.41 (m, 2H).
2-[2-[[3-(2-Amino-6-chloro-pyrimidin-4-yl)-1-methyl-pyrazol-4-yl]methyl]phenoxy]acetaldehyde.
To a solution of 3-[2-[[3-(2-amino-6-chloro-pyrimidin-4-yl)-1-methyl-pyrazol-4-yl]methyl]-phenoxy]propane-1,2-diol (650 mg, 1.67 mmol, 1 equiv) in dioxane (10 mL) and H2O (3 mL) was added NaIO4 (892 mg, 4.17 mmol, 0.231 mL, 2.5 equiv) at 0 °C. The mixture was stirred at 25 °C for 2 h under N2. The reaction mixture was poured into saturated, aq. Na2SO3 (150 mL). The mixture was extracted with ethyl acetate (50 mL × 3). The organic phase was washed with brine (80 mL), dried over anhydrous Na2SO4, and filtered. The filtrate was concentrated under reduced pressure to furnish the title compound (750 mg, crude) as a light-yellow oil. The material was used directly in next step without further purification.
4-Chloro-6-[1-methyl-4-[[2-[2-(6-oxa-3-azabicyclo[3.1.1]heptan-3-yl)ethoxy]phenyl]methyl]pyrazol-3-yl]pyrimidin-2-amine (20).
To a solution of 2-[2-[[3-(2-amino-6-chloro-pyrimidin-4-yl)-1-methyl-pyrazol-4-yl]methyl]phenoxy]acetaldehyde (0.11 g, 0.31 mmol, 1 equiv) in MeOH (1 mL) and THF (0.5 mL) was added TEA (37 mg, 0.37 mmol, 0.051 mL, 1.2 equiv) and 6-oxa-3-azabicyclo[3.1.1]-heptane (50 mg, 0.37 mmol, 1.2 equiv, HCl). The mixture was stirred at 25 °C for 2 h under N2. NaBH3CN (58 mg, 0.92 mmol, 3 equiv) was added to the mixture. The mixture was stirred at 25 °C for another 2 h under N2. The reaction mixture was poured into H2O (100 mL). The mixture was extracted with ethyl acetate (30 mL × 3). The organic phase was washed with brine (50 mL), dried over anhydrous Na2SO4, and filtered. The filtrate was concentrated under reduced pressure. The residue was purified by prep-TLC (EtOAc/MeOH = 20:1) to furnish the desired compound. The compound was further purified by prep-HPLC (column: Waters Xbridge BEH C18 100 × 30 mm × 10 μm; mobile phase: [water (10 mM NH4HCO3)-ACN]; B%: 25–55%, 8 min) to furnish the title compound as (14 mg, 10%) 20 as a white solid. 1H NMR: (400 MHz, DMSO-d6) δ 7.23–7.13 (m, 3H), 7.06 (br s, 2H), 7.00–6.96 (m, 2H), 6.84 (t, J = 7.3 Hz, 1H), 4.33 (d, J = 6.0 Hz, 2H), 4.21 (s, 2H), 4.09 (t, J = 5.6 Hz, 2H), 3.78 (s, 3H), 3.00 (d, J = 11.3 Hz, 2H), 2.87 (t, J = 5.6 Hz, 2H), 2.78 (q, J = 6.4 Hz, 1H), 2.67 (d, J = 11.3 Hz, 2H), 2.09 (d, J = 7.8 Hz, 1H); 13C NMR (126 MHz, DMSO) δ 163.8, 163.7, 160.4, 156.7, 144.3, 132.5, 130.6, 130.1, 127.9, 122.3, 120.8, 112.3, 104.6, 79.2, 66.4, 55.8, 55.5, 30.4, 25.6; LCMS: (M + H+): 441.2; HRMS calcd for C22H26N6ClO2 (M + H+) 441.1801; found: 441.1802.
Preparation of TDI-11861 (33). 4-Iodo-1-tetrahydropyran-2-ylpyrazole-3-carboxylate.
To a solution of ethyl 4-iodo-1H-pyrazole-3-carboxylate (20 g, 75 mmol, 1 equiv) in THF (75 mL) were added DHP (19.0 g, 226 mmol, 20.6 mL, 3 equiv) and PTSA (1.29 g, 7.52 mmol, 0.1 equiv). The reaction mixture was stirred at 80 °C for 8 h under N2. The reaction mixture was concentrated under reduced pressure. Then, the mixture was diluted with H2O (300 mL) and extracted with ethyl acetate (100 mL × 3). The combined organic layers were washed with brine (100 mL), dried over Na2SO4, and filtered. The filtrate was concentrated under reduced pressure. The residue was purified by flash silica gel chromatography (ISCO; 20 g SepaFlash Silica Flash Column, gradient eluent of 0–20% ethyl acetate/petroleum ether @ 100 mL/min), which furnished the title compound (24 g, 91%) as a colorless oil.
Ethyl 4-[(2-allyloxyphenyl)-hydroxy-methyl]-1-tetrahydropyran-2-yl-pyrazole-3-carboxylate.
To a solution of ethyl 4-iodo-1-tetrahydropyran-2-yl-pyrazole-3-carboxylate (13.3 g, 38.0 mmol, 1 equiv) in THF (150 mL) was added i-PrMgCl-LiCl (1.30 M, 30.7 mL, 1.05 equiv) at −15 °C under a nitrogen atmosphere. After stirring at −15 °C for 0.5 h, a solution of 2-allyloxybenzaldehyde (6.78 g, 41.8 mmol, 1.1 equiv) in THF (20 mL) was added to the mixture dropwise. After the addition, the mixture was stirred at 60 °C for 8 h and then at 25 °C for 8 h under N2. The reaction mixture was quenched with saturated, aqueous NH4Cl (300 mL). The mixture was extracted with ethyl acetate (100 mL × 3). The combined organic layer was washed with brine (100 mL), dried over Na2SO4, and filtered. The filtrate was concentrated under reduced pressure. The residue was purified by flash silica gel chromatography (ISCO; 20 g SepaFlash Silica Flash Column, eluent of 0–18% ethyl acetate/petroleum ether gradient @ 45 mL/min) to furnish the title compound as a yellow oil (7.7 g, 19.9 mmol, 52%).
Ethyl 4-[(2-allyloxyphenyl)methyl]-1H-pyrazole-3-carboxylate.
To a solution of NaI (16.8 g, 112 mmol, 6 equiv) in MeCN (150 mL) was added TMSCl (12.1 g, 112 mmol, 14.2 mL, 6 equiv) dropwise under N2. After 10 min of stirring at 25 °C, a solution of ethyl 4-[(2-allyloxyphenyl)-hydroxy-methyl]-1-tetrahydropyran-2-yl-pyrazole-3-carboxylate (7.2 g, 18.6 mmol, 1 equiv) in MeCN (20 mL) was added dropwise. The mixture was stirred at 25 °C under N2 for 2 h. The reaction mixture was quenched with saturated, aqueous Na2SO3 (300 mL). The mixture was extracted with ethyl acetate (100 mL × 3). The combined organic layer was washed with brine (100 mL), dried over Na2SO4, and filtered. The filtrate was concentrated under reduced pressure. The residue was purified by flash silica gel chromatography (ISCO; 80 g SepaFlash Silica Flash Column, eluent of 0–30% ethyl acetate/petroleum ether gradient @ 100 mL/min) to furnish the title compound as a colorless oil (4.6 g, 91%).
Ethyl 4-[(2-allyloxyphenyl)methyl]-1-(difluoromethyl)pyrazole-3-carboxylate.
To a solution of ethyl 4-[(2-allyloxyphenyl)methyl]-1H-pyrazole-3-carboxylate (4.6 g, 16.1 mmol, 1 equiv) in MeCN (80 mL) were added KF (2.3 g, 40 mmol, 0.94 mL, 2.5 equiv) and 1-[[bromo(difluoro)methyl]-ethoxy-phosphoryl]oxyethane (7.7 g, 29 mmol, 1.8 equiv). The mixture was stirred at 25 °C for 40 h under N2. The reaction mixture was quenched with saturated, aqueous NH4Cl (200 mL). The mixture was extracted with ethyl acetate (100 mL × 3). The combined organic layer was washed with brine (100 mL), dried over Na2SO4, and filtered. The filtrate was concentrated under reduced pressure. The residue was purified by flash silica gel chromatography (ISCO; 4 g SepaFlash Silica Flash Column, eluent of 0–5% ethyl acetate/petroleum ether gradient @ 45 mL/min) to furnish the title compound as a colorless oil (5.2 g, ~100%).
Ethyl 3-[4-[(2-allyloxyphenyl)methyl]-1-(difluoromethyl)pyrazol-3-yl]-3-oxo-propanoate.
To a solution of ethyl 4-[(2-allyloxyphenyl)methyl]-1-(difluoromethyl)pyrazole-3-carboxylate (3.0 g, 8.9 mmol, 1 equiv) and ethyl acetate (5.5 g, 62 mmol, 6.1 mL, 7 equiv) in THF (100 mL) was added LiHMDS (1.0 M, 27 mL, 3 equiv) at −40 °C under N2. The mixture was stirred at −40 °C for 1 h under N2. The reaction mixture was quenched with saturated, aqueous NH4Cl (200 mL). The mixture was extracted with ethyl acetate (100 mL × 3). The combined organic layer was washed with brine (100 mL), dried over Na2SO4, and filtered. The filtrate was concentrated under reduced pressure. The residue was purified by flash silica gel chromatography (ISCO; 40 g SepaFlash Silica Flash Column, eluent of 0–4% ethyl acetate/petroleum ether gradient @ 75 mL/min) to furnish as a colorless oil (2.5 g, 6.6 mmol, 74%). LCMS: (M + H+): 379.1.
4-[4-[(2-Allyloxyphenyl)methyl]-1-(difluoromethyl)pyrazol-3-yl]-2-amino-1H-pyrimidin-6-one.
To a solution of ethyl 3-[4-[(2-allyloxyphenyl)methyl]-1-(difluoromethyl)pyrazol-3-yl]-3-oxo-propanoate (2.5 g, 6.6 mmol, 1 equiv) in EtOH (50 mL) was added guanidine carbonate (3.6 g, 20 mmol, 3 equiv). The mixture was stirred at 85 °C for 12 h under N2. The reaction mixture was diluted with H2O (50 mL) and concentrated under reduced pressure to remove EtOH. The mixture was extracted with ethyl acetate (100 mL × 3). The combined organic layer was diluted with THF (100 mL), dried over Na2SO4, and filtered. The filtrate was concentrated under reduced pressure to furnish the title compound as a yellow solid (2.2 g, 89%). LCMS: (M + H+): 374.1.
4-[4-[(2-Allyloxyphenyl)methyl]-1-(difluoromethyl)pyrazol-3-yl]-6-chloro-pyrimidin-2-amine.
POCl3 (13.6 g, 88.4 mmol, 8.21 mL, 15 equiv) was added into a solution of 4-[4-[(2-allyloxyphenyl)methyl]-1-(difluoromethyl)pyrazol-3-yl]-2-amino-1H-pyrimidin-6-one (2.20 g, 5.89 mmol, 1 equiv) in dioxane (5 mL) at 25 °C. The mixture was heated to 75 °C for 12 h under N2. The reaction mixture was concentrated under reduced pressure to remove the solvent. The reaction mixture was quenched with saturated, aqueous NaHCO3 (200 mL). The mixture was extracted with ethyl acetate (100 mL × 3). The combined organic layer was washed with brine (100 mL), dried over Na2SO4, and filtered. The filtrate was concentrated under reduced pressure. The residue was purified by flash silica gel chromatography (ISCO; 20 g SepaFlash Silica Flash Column, eluent of 0–10% ethyl acetate/petroleum ether gradient @ 75 mL/min) to furnish the title compound as a yellow solid (1.10 g, 2.8 mmol, 48%).
3-[2-[[3-(2-Amino-6-chloro-pyrimidin-4-yl)-1-(difluoromethyl)-pyrazol-4-yl]methyl]phenoxy]propane-1,2-diol.
To a solution of 4-[4-[(2-allyloxyphenyl)methyl]-1-(difluoromethyl)pyrazol-3-yl]-6-chloro-pyrimidin-2-amine (550 mg, 1.40 mmol, 1 equiv) in THF (4 mL) and H2O (4 mL) were added NMO (460 mg, 3.93 mmol, 0.415 mL, 2.8 equiv) and OsO4 (71 mg, 0.28 mmol, 0.014 mL, 0.2 equiv) at 0 °C. The reaction mixture was stirred at 25 °C for 2 h under N2. The reaction mixture was quenched with saturated aqueous Na2SO3 (60 mL). The mixture was extracted with ethyl acetate (30 mL × 3). The combined organic layer was washed with brine (20 mL), dried over Na2SO4, and filtered. The filtrate was concentrated under reduced pressure. The residue was purified by flash silica gel chromatography (ISCO; 4 g SepaFlash Silica Flash Column, eluent of 0–60% ethyl acetate/petroleum ether gradient @ 50 mL/min) to furnish the title compound as a yellow solid (450 mg, 1.06 mmol, 75%).
2-[2-[[3-(2-Amino-6-chloro-pyrimidin-4-yl)-1-(difluoromethyl)-pyrazol-4-yl]methyl]phenoxy]acetaldehyde.
A mixture of 3-[2-[[3-(2-amino-6-chloro-pyrimidin-4-yl)-1-(difluoromethyl)pyrazol-4-yl]-methyl]phenoxy]propane-1,2-diol (450 mg, 1.06 mmol, 1 equiv) in dioxane (9 mL) and H2O (3 mL) was added NaIO4 (565 mg, 2.64 mmol, 0.146 mL, 2.5 equiv) at 0 °C. The reaction mixture was stirred at 20 °C for 2 h under N2. The reaction mixture was quenched with saturated, aqueous Na2SO3 (40 mL). The mixture was extracted with ethyl acetate (30 mL × 3). The combined organic layer was washed with brine (30 mL), dried over Na2SO4, and filtered. The filtrate was concentrated under reduced pressure to furnish the title compound as a yellow solid (417 mg, ~100%).
[(3R)-4-[2-[2-[[3-(2-Amino-6-chloro-pyrimidin-4-yl)-1-(difluoromethyl)pyrazol-4-yl]methyl]phenoxy]ethyl]morpholin-3-yl]methanol (33, TDI-11861).
To a solution of 2-[2-[[3-(2-amino-6-chloro-pyrimidin-4-yl)-1-(difluoromethyl)pyrazol-4-yl]methyl]-phenoxy]acetaldehyde (100 mg, 0.254 mmol, 1 equiv) in DCE (3 mL) were added TEA (26 mg, 0.25 mmol, 0.035 mL, 1 equiv) and [(3R)-morpholin-3-yl]methanol (43 mg, 0.28 mmol, HCl salt). After stirring at 20 °C for 3 h, NaBH(OAc)3 (215 mg, 1.02 mmol, 4 equiv) was added to the mixture. The mixture was stirred at 20 °C for 12 h under N2. The reaction mixture was quenched with saturated aqueous NaHCO3 (40 mL). The mixture was extracted with ethyl acetate (20 mL × 3). The combined organic layer was washed with brine (30 mL), dried over Na2SO4, and filtered. The filtrate was concentrated under reduced pressure. The residue was purified by prep-HPLC (column: Waters Xbridge BEH C18 100 × 30 mm × 10 μm; mobile phase: [water (10 mM NH4HCO3)-ACN]; B%: 25–55%) to furnish the title compound as a white solid (36 mg, 29%). 1H NMR: (400 MHz, chloroform-d) δ 7.43 (s, 1H), 7.32–7.27 (m, 1H), 7.25–7.19 (m, 1H), 7.16 (s, 0.5H), 7.11–7.06 (m, 1H), 7.01 (s, 0.3H), 6.95–6.88 (m, 2H), 5.53–5.37 (m, 2H), 4.35–4.20 (m, 2H), 4.19–4.11 (m, 1H), 4.10–4.02 (m, 1H), 3.85 (dd, J = 4.1, 11.6 Hz, 1H), 3.80–3.70 (m, 2H), 3.62–3.46 (m, 2H), 3.45–3.36 (m, 1H), 3.29–3.20 (m, 1H), 2.90 (td, J = 2.7, 11.8 Hz, 1H), 2.79–2.66 (m, 2H), 2.59–2.48 (m, 2H); 13C NMR (126 MHz, DMSO-d6) δ 162.2, 160.7, 159.3, 155.1, 146.6, 128.9, 128.4, 127.2, 126.6, 122.3, 119.2, 110.9, 110.5, 109.0, 107.0, 103.7, 67.4, 65.0, 64.9, 59.9, 57.8, 51.6, 50.4, 23.8; LCMS: (M + H+): 495.2. HRMS calcd for C22H26N6ClF2O3 (M + H+) 495.1718; found: 495.1715.
Supplementary Material
ACKNOWLEDGMENTS
The authors thank the BESSY beamline staff for excellent technical support and Dr. Tanweer Khan for generating 13C NMR data. They acknowledge the MSKCC supercomputing resources (https://www.mskcc.org/research/ski/core-facilities/high-performance-computing-group) made available for conducting the research reported in this paper. The authors gratefully acknowledge the support to the project generously provided by the Tri-Institutional Therapeutics Discovery Institute (TDI), a 501(c)(3) organization. TDI receives financial support from Takeda Pharmaceutical Company, TDI’s parent institutes (Memorial Sloan Kettering Cancer Center, The Rockefeller University and Weill Cornell Medicine) and a generous contribution from Lewis Sanders and other philanthropic sources.
Funding
This work was supported by NIH Grants P50 HD100549 and R01 HD088571 (to J.B. and L.R.L.), F31 AG069501 (T.R.), F31 HD105363 (J.F.), and by generous funding from the Male Contraceptive Initiative (MCI) (to J.B. and L.R.L.).
ABBREVIATIONS
- AC
adenylyl cyclase
- IP
intraperitoneal
- PO
per os
- sAC
soluble adenylyl cyclase
- tmAC
transmembrane adenylyl cyclase
- PK
pharmacokinetic
Footnotes
Supporting Information
The Supporting Information is available free of charge at https://pubs.acs.org/doi/10.1021/acs.jmedchem.2c01133.
Inhibitory activities of TDI-11861 against human tmACs (Figure S1); HPLC traces for 2, 4, 9, 19, 20, 22, 23, 26, 28, and 33; synthesis and characterization for 5–14, 16–18, and 21–32; diffraction data and refinement statistics (Table S1); kinase activity data for TDI-11861 (33) (Table S2); drug target safety panel data for TDI-11861 (33) (Table S3); and analytical method for mouse plasma samples (PDF)
Smiles data (CSV)
Complete contact information is available at: https://pubs.acs.org/10.1021/acs.jmedchem.2c01133
The authors declare the following competing financial interest(s): Drs. Meinke, Levin, Buck, and Steegborn are co-founders of Sacyl Pharmaceuticals, Inc. established to develop sAC inhibitors into on-demand contraceptives.
Contributor Information
Michael Miller, Tri-Institutional Therapeutics Discovery Institute, New York, New York 10021, United States.
Thomas Rossetti, Department of Pharmacology, Weill Cornell Medicine, New York, New York 10021, United States.
Jacob Ferreira, Department of Pharmacology, Weill Cornell Medicine, New York, New York 10021, United States.
Lubna Ghanem, Department of Pharmacology, Weill Cornell Medicine, New York, New York 10021, United States.
Melanie Balbach, Department of Pharmacology, Weill Cornell Medicine, New York, New York 10021, United States.
Navpreet Kaur, Department of Pharmacology, Weill Cornell Medicine, New York, New York 10021, United States.
Lonny R. Levin, Department of Pharmacology, Weill Cornell Medicine, New York, New York 10021, United States
Jochen Buck, Department of Pharmacology, Weill Cornell Medicine, New York, New York 10021, United States.
Maria Kehr, Department of Biochemistry, University of Bayreuth, 95440 Bayreuth, Germany.
Sandrine Coquille, Department of Biochemistry, University of Bayreuth, 95440 Bayreuth, Germany.
Joop van den Heuvel, Helmholtz Centre for Infection Research, Recombinant Protein Expression, 38124 Braunschweig, Germany.
Clemens Steegborn, Department of Biochemistry, University of Bayreuth, 95440 Bayreuth, Germany.
Makoto Fushimi, Tri-Institutional Therapeutics Discovery Institute, New York, New York 10021, United States.
Efrat Finkin-Groner, Tri-Institutional Therapeutics Discovery Institute, New York, New York 10021, United States.
Robert W. Myers, Tri-Institutional Therapeutics Discovery Institute, New York, New York 10021, United States
Stacia Kargman, Tri-Institutional Therapeutics Discovery Institute, New York, New York 10021, United States.
Nigel J. Liverton, Tri-Institutional Therapeutics Discovery Institute, New York, New York 10021, United States
David J. Huggins, Tri-Institutional Therapeutics Discovery Institute, New York, New York 10021, United States; Department of Physiology and Biophysics, Weill Cornell Medicine, New York, New York 10021, United States.
Peter T. Meinke, Tri-Institutional Therapeutics Discovery Institute, New York, New York 10021, United States; Department of Pharmacology, Weill Cornell Medicine, New York, New York 10021, United States.
Data Availability Statement
Diffraction data and refined model for the sAC/TDI-11861 complex have been deposited with the worldwide PDB (wwPDB) under accession number 8B75.
REFERENCES
- (1).Ostrom KF; LaVigne JE; Brust TF; Seifert R; Dessauer CW; Watts VJ; Ostrom RS Physiological Roles of Mammalian Transmembrane Adenylyl Cyclase Isoforms. Physiol. Rev 2022, 102, 815–857. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (2).Wiggins SV; Steegborn C; Levin LR; Buck J Pharmacological Modulation of the Co2/Hco3(−)/Ph-, Calcium-, and Atp-Sensing Soluble Adenylyl Cyclase. Pharmacol. Ther 2018, 190, 173–186. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (3).Kamenetsky M; Middelhaufe S; Bank EM; Levin LR; Buck J; Steegborn C Molecular Details of Camp Generation in Mammalian Cells: A Tale of Two Systems. J. Mol. Biol 2006, 362, 623–639. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (4).Rahman N; Ramos-Espiritu L; Milner TA; Buck J; Levin LR Soluble Adenylyl Cyclase Is Essential for Proper Lysosomal Acidification. J. Gen. Physiol 2016, 148, 325–339. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (5).Valsecchi F; Konrad C; Manfredi G Role of Soluble Adenylyl Cyclase in Mitochondria. Biochim. Biophys. Acta, Mol. Basis Dis 2014, 1842, 2555–2560. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (6).Zippin JH; Farrell J; Huron D; Kamenetsky M; Hess KC; Fischman DA; Levin LR; Buck J Bicarbonate-Responsive ″Soluble″ Adenylyl Cyclase Defines a Nuclear Camp Microdomain. J. Cell Biol 2004, 164, 527–534. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (7).Zhou D; Ota K; Nardin C; Feldman M; Widman A; Wind O; Simon A; Reilly M; Levin LR; Buck J; Wakamatsu K; Ito S; Zippin JH Mammalian Pigmentation Is Regulated by a Distinct Camp-Dependent Mechanism That Controls Melanosome Ph. Sci. Signaling 2018, 11, No. eaau7987. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (8).Rossetti T; Jackvony S; Buck J; Levin LR Bicarbonate, Carbon Dioxide and Ph Sensing Via Mammalian Bicarbonate-Regulated Soluble Adenylyl Cyclase. Interface Focus 2021, 11, No. 20200034. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (9).Zippin JH; Chen Y; Straub SG; Hess KC; Diaz A; Lee D; Tso P; Holz GG; Sharp GW; Levin LR; Buck J Co2/Hco3(−)- and Calcium-Regulated Soluble Adenylyl Cyclase as a Physiological Atp Sensor. J. Biol. Chem 2013, 288, 33283–33291. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (10).Lee YS; Tresguerres M; Hess K; Marmorstein LY; Levin LR; Buck J; Marmorstein AD Regulation of Anterior Chamber Drainage by Bicarbonate-Sensitive Soluble Adenylyl Cyclase in the Ciliary Body. J. Biol. Chem 2011, 286, 41353–41358. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (11).Gandhi JK; Roy Chowdhury U; Manzar Z; Buck J; Levin LR; Fautsch MP; Marmorstein AD Differential Intraocular Pressure Measurements by Tonometry and Direct Cannulation after Treatment with Soluble Adenylyl Cyclase Inhibitors. J. Ocul. Pharmacol. Ther 2017, 33, 574–581. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (12).Hess KC; Jones BH; Marquez B; Chen Y; Ord TS; Kamenetsky M; Miyamoto C; Zippin JH; Kopf GS; Suarez SS; Levin LR; Williams CJ; Buck J; Moss SB The ″Soluble″ Adenylyl Cyclase in Sperm Mediates Multiple Signaling Events Required for Fertilization. Dev. Cell 2005, 9, 249–259. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (13).Xie F; Garcia MA; Carlson AE; Schuh SM; Babcock DF; Jaiswal BS; Gossen JA; Esposito G; van Duin M; Conti M Soluble Adenylyl Cyclase (Sac) Is Indispensable for Sperm Function and Fertilization. Dev. Biol 2006, 296, 353–362. [DOI] [PubMed] [Google Scholar]
- (14).Balbach M; Fushimi M; Huggins DJ; Steegborn C; Meinke PT; Levin LR; Buck J Optimization of Lead Compounds into on-Demand, Nonhormonal Contraceptives: Leveraging a Public-Private Drug Discovery Institute Collaborationdagger. Biol. Reprod 2020, 103, 176–182. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (15).Balbach M; Ghanem L; Rossetti T; Kaur N; Ritagliati C; Ferreira J; Krapf D; Puga Molina LC; Santi CM; Hansen JN; Wachten D; Fushimi M; Meinke PT; Buck J; Levin LR Soluble Adenylyl Cyclase Inhibition Prevents Human Sperm Functions Essential for Fertilization. Mol. Hum. Reprod 2021, 27, No. gaab054. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (16).Matzuk MM; Georg GI; Yan W A Special Issue on Contraceptive Development: Past, Present, and Future; Oxford University Press, 2020; Vol. 103, pp 145–146. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (17).Balbach M; Rossetti T; Ferreira J; Ghanem L; Ritagliati C; Myers RW; Meinke PT; Buck J; Levin LR On Demand Male Contraception Via Acute Systemic Inhibition of Soluble Adenylyl Cyclase. Nat. Commun 2022. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (18).Balbach M; Beckert V; Hansen JN; Wachten D Shedding Light on the Role of Camp in Mammalian Sperm Physiology. Mol. Cell. Endocrinol 2018, 468, 111–120. [DOI] [PubMed] [Google Scholar]
- (19).Wennemuth G; Carlson AE; Harper AJ; Babcock DF Bicarbonate Actions on Flagellar and Ca2+ -Channel Responses: Initial Events in Sperm Activation. Development 2003, 130, 1317–1326. [DOI] [PubMed] [Google Scholar]
- (20).Buffone MG; Wertheimer EV; Visconti PE; Krapf D Central Role of Soluble Adenylyl Cyclase and Camp in Sperm Physiology. Biochim. Biophys. Acta, Mol. Basis Dis 2014, 1842, 2610–2620. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (21).Chen Y; Cann MJ; Litvin TN; Iourgenko V; Sinclair ML; Levin LR; Buck J Soluble Adenylyl Cyclase as an Evolutionarily Conserved Bicarbonate Sensor. Science 2000, 289, 625–628. [DOI] [PubMed] [Google Scholar]
- (22).Esposito G; Jaiswal BS; Xie F; Krajnc-Franken MA; Robben TJ; Strik AM; Kuil C; Philipsen RL; van Duin M; Conti M; Gossen JA Mice Deficient for Soluble Adenylyl Cyclase Are Infertile Because of a Severe Sperm-Motility Defect. Proc. Natl. Acad. Sci. U.S.A 2004, 101, 2993–2998. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (23).Akbari A; Pipitone GB; Anvar Z; Jaafarinia M; Ferrari M; Carrera P; Totonchi M Adcy10 Frameshift Variant Leading to Severe Recessive Asthenozoospermia and Segregating with Absorptive Hypercalciuria. Hum. Reprod 2019, 34, 1155–1164. [DOI] [PubMed] [Google Scholar]
- (24).Fushimi M; Buck H; Balbach M; Gorovyy A; Ferreira J; Rossetti T; Kaur N; Levin LR; Buck J; Quast J; van den Heuvel J; Steegborn C; Finkin-Groner E; Kargman S; Michino M; Foley MA; Miller M; Liverton NJ; Huggins DJ; Meinke PT Discovery of Tdi-10229: A Potent and Orally Bioavailable Inhibitor of Soluble Adenylyl Cyclase (Sac, Adcy10. ACS Med. Chem. Lett 2021, 12, 1283–1287. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (25).Ramos-Espiritu L; Kleinboelting S; Navarrete FA; Alvau A; Visconti PE; Valsecchi F; Starkov A; Manfredi G; Buck H; Adura C; Zippin JH; van den Heuvel J; Glickman JF; Steegborn C; Levin LR; Buck J Discovery of Lre1 as a Specific and Allosteric Inhibitor of Soluble Adenylyl Cyclase. Nat. Chem. Biol 2016, 12, 838–844. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (26).Kleinboelting S; Diaz A; Moniot S; van den Heuvel J; Weyand M; Levin LR; Buck J; Steegborn C Crystal Structures of Human Soluble Adenylyl Cyclase Reveal Mechanisms of Catalysis and of Its Activation through Bicarbonate. Proc. Natl. Acad. Sci. U.S.A 2014, 111, 3727–3732. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (27).Steegborn C Structure, Mechanism, and Regulation of Soluble Adenylyl Cyclases - Similarities and Differences to Transmembrane Adenylyl Cyclases. Biochim. Biophys. Acta, Mol. Basis Dis 2014, 1842, 2535–2547. [DOI] [PubMed] [Google Scholar]
- (28).Schuetz DA; de Witte WEA; Wong YC; Knasmueller B; Richter L; Kokh DB; Sadiq SK; Bosma R; Nederpelt I; Heitman LH; Segala E; Amaral M; Guo D; Andres D; Georgi V; Stoddart LA; Hill S; Cooke RM; De Graaf C; Leurs R; Frech M; Wade RC; de Lange ECM; Ijzerman AP; Müller-Fahrnow A; Ecker GF Kinetics for Drug Discovery: An Industry-Driven Effort to Target Drug Residence Time. Drug Discovery Today 2017, 22, 896–911. [DOI] [PubMed] [Google Scholar]
- (29).Wang L; Wu Y; Deng Y; Kim B; Pierce L; Krilov G; Lupyan D; Robinson S; Dahlgren MK; Greenwood J; et al. Accurate and Reliable Prediction of Relative Ligand Binding Potency in Prospective Drug Discovery by Way of a Modern Free-Energy Calculation Protocol and Force Field. J. Am. Chem. Soc 2015, 137, 2695–2703. [DOI] [PubMed] [Google Scholar]
- (30).Wade AD; Rizzi A; Wang Y; Huggins DJ Computational Fluorine Scanning Using Free-Energy Perturbation. J. Chem. Inf. Model 2019, 59, 2776–2784. [DOI] [PubMed] [Google Scholar]
- (31).Zhang Z; Tang W Drug Metabolism in Drug Discovery and Development. Acta Pharm. Sin. B 2018, 8, 721–732. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (32).Issa NT; Wathieu H; Ojo A; Byers SW; Dakshanamurthy S Drug Metabolism in Preclinical Drug Development: A Survey of the Discovery Process, Toxicology, and Computational Tools. Curr. Drug Metab 2017, 18, 556–565. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (33).Johnson BM; Shu YZ; Zhuo X; Meanwell NA Metabolic and Pharmaceutical Aspects of Fluorinated Compounds. J. Med. Chem 2020, 63, 6315–6386. [DOI] [PubMed] [Google Scholar]
- (34).Myszka DG; Rich RL Implementing Surface Plasmon Resonance Biosensors in Drug Discovery. Pharm. Sci. Technol. Today 2000, 3, 310–317. [DOI] [PubMed] [Google Scholar]
- (35).Copeland RA; Basavapathruni A; Moyer M; Scott MP Impact of Enzyme Concentration and Residence Time on Apparent Activity Recovery in Jump Dilution Analysis. Anal. Biochem 2011, 416, 206–210. [DOI] [PubMed] [Google Scholar]
- (36).Hirozane Y; Toyofuku M; Yogo T; Tanaka Y; Sameshima T; Miyahisa I; Yoshikawa M Structure-Based Rational Design of Staurosporine-Based Fluorescent Probe with Broad-Ranging Kinase Affinity for Kinase Panel Application. Bioorg. Med. Chem. Lett 2019, 29, No. 126641. [DOI] [PubMed] [Google Scholar]
- (37).Sameshima T; Miyahisa I; Yamasaki S; Gotou M; Kobayashi T; Sakamoto J High-Throughput Quantitative Intrinsic Thiol Reactivity Evaluation Using a Fluorescence-Based Competitive Endpoint Assay. SLAS Discovery 2017, 22, 1168–1174. [DOI] [PubMed] [Google Scholar]
- (38).Buck J; Levin LR The Role of Soluble Adenylyl Cyclase in Health and Disease. Biochim. Biophys. Acta, Mol. Basis Dis 2014, 1842, 2533–2534. [DOI] [PubMed] [Google Scholar]
- (39).Copeland RA Evolution of the Drug-Target Residence Time Model. Expert Opin. Drug Discovery 2021, 16, 1441–1451. [DOI] [PubMed] [Google Scholar]
- (40).Lee KSS; Yang J; Niu J; Ng CJ; Wagner KM; Dong H; Kodani SD; Wan D; Morisseau C; Hammock BD Drug-Target Residence Time Affects in Vivo Target Occupancy through Multiple Pathways. ACS Cent. Sci 2019, 5, 1614–1624. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (41).Walkup GK; You Z; Ross PL; Allen EK; Daryaee F; Hale MR; O’Donnell J; Ehmann DE; Schuck VJ; Buurman ET; Choy AL; Hajec L; Murphy-Benenato K; Marone V; Patey SA; Grosser LA; Johnstone M; Walker SG; Tonge PJ; Fisher SL Translating Slow-Binding Inhibition Kinetics into Cellular and in Vivo Effects. Nat. Chem. Biol 2015, 11, 416–423. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (42).Khanna A; Cote A; Arora S; Moine L; Gehling VS; Brenneman J; Cantone N; Stuckey JI; Apte S; Ramakrishnan A; Bruderek K; Bradley WD; Audia JE; Cummings RT; Sims RJ 3rd; Trojer P; Levell JR Design, Synthesis, and Pharmacological Evaluation of Second Generation Ezh2 Inhibitors with Long Residence Time. ACS Med. Chem. Lett 2020, 11, 1205–1212. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (43).Bradshaw JM; McFarland JM; Paavilainen VO; Bisconte A; Tam D; Phan VT; Romanov S; Finkle D; Shu J; Patel V; Ton T; Li X; Loughhead DG; Nunn PA; Karr DE; Gerritsen ME; Funk JO; Owens TD; Verner E; Brameld KA; Hill RJ; Goldstein DM; Taunton J Prolonged and Tunable Residence Time Using Reversible Covalent Kinase Inhibitors. Nat. Chem. Biol 2015, 11, 525–531. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (44).Friesner RA; Banks JL; Murphy RB; Halgren TA; Klicic JJ; Mainz DT; Repasky MP; Knoll EH; Shelley M; Perry JK; Shaw DE; Francis P; Shenkin PS Glide: A New Approach for Rapid, Accurate Docking and Scoring. 1. Method and Assessment of Docking Accuracy. J. Med. Chem 2004, 47, 1739–1749. [DOI] [PubMed] [Google Scholar]
- (45).Litvin TN; Kamenetsky M; Zarifyan A; Buck J; Levin LR Kinetic Properties of “Soluble” Adenylyl Cyclase: Synergism between Calcium and Bicarbonate. J. Biol. Chem 2003, 278, 15922–15926. [DOI] [PubMed] [Google Scholar]
- (46).Salomon Y Adenylate Cyclase Assay. Adv. Cyclic Nucleotide Res 1979, 10, 35–55. [PubMed] [Google Scholar]
- (47).Nakashima S; Yamamoto K; Arai Y; Ikeda Y Impact of Physicochemical Profiling for Rational Approach on Drug Discovery. Chem. Pharm. Bull 2013, 61, 1228–1238. [DOI] [PubMed] [Google Scholar]
- (48).Masumoto K; Takeyasu A; Oizumi K; Kobayashi T Studies of Novel 1, 4-Dihydropyridine Ca Antagonist Cs-905. I. Measurement of Partition Coefficient (Log P) by High Performance Liquid Chromatography (Hplc). Yakugaku Zasshi 1995, 115, 213–220. [DOI] [PubMed] [Google Scholar]
- (49).Kleinboelting S; van den Heuvel J; Kambach C; Weyand M; Leipelt M; Steegborn C Expression, Purification, Crystallization and Preliminary X-Ray Diffraction Analysis of a Mammalian Type 10 Adenylyl Cyclase. Acta Crystallogr., Sect. F: Struct. Biol. Commun 2014, 70, 467–469. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (50).Mueller U; Darowski N; Fuchs MR; Forster R; Hellmig M; Paithankar KS; Puhringer S; Steffien M; Zocher G; Weiss MS Facilities for Macromolecular Crystallography at the Helmholtz-Zentrum Berlin. J. Synchrotron Radiat 2012, 19, 442–449. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (51).Sparta KM; Krug M; Heinemann U; Mueller U; Weiss MS Xdsapp2. 0. J. Appl. Crystallogr 2016, 49, 1085–1092. [Google Scholar]
- (52).McCoy AJ; Grosse-Kunstleve RW; Adams PD; Winn MD; Storoni LC; Read RJ Phaser Crystallographic Software. J. Appl. Crystallogr 2007, 40, 658–674. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (53).Emsley P; Lohkamp B; Scott WG; Cowtan K Features and Development of Coot. Acta Crystallogr., Sect. D: Biol. Crystallogr 2010, 66, 486–501. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (54).Liebschner D; Afonine PV; Baker ML; Bunkoczi G; Chen VB; Croll TI; Hintze B; Hung LW; Jain S; McCoy AJ; Moriarty NW; Oeffner RD; Poon BK; Prisant MG; Read RJ; Richardson JS; Richardson DC; Sammito MD; Sobolev OV; Stockwell DH; Terwilliger TC; Urzhumtsev AG; Videau LL; Williams CJ; Adams PD Macromolecular Structure Determination Using X-Rays, Neutrons and Electrons: Recent Developments in Phenix. Acta Crystallogr., Sect. D: Struct. Biol 2019, 75, 861–877. [DOI] [PMC free article] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.
Supplementary Materials
Data Availability Statement
Diffraction data and refined model for the sAC/TDI-11861 complex have been deposited with the worldwide PDB (wwPDB) under accession number 8B75.
