Abstract
The sperm-specific Ca2+ channel CatSper (cation channel of sperm) controls the intracellular Ca2+ concentration and, thereby, the swimming behavior of sperm from many species. The steroidal ethylenediamine RU1968 (1) represents a well-characterized, potent, and fairly selective cross-species inhibitor of CatSper. Due to its two additional centers of chirality in the amine-bearing side chain, RU1968 is a mixture of diastereomeric pairs of enantiomers and, thus, difficult to synthesize. This has hampered the use of this commercially not available inhibitor as a powerful tool for research. Here, simplifying both structure and synthesis, we introduced novel stereochemically less complex and enantiomerically pure aminomethyl RU1968 analogues lacking the C-21 CH3 moiety. Starting from (+)-estrone, a five-step synthesis was developed comprising a Wittig reaction as the key step, leading to a diastereomerically pure 17β-configured aldehyde. Subsequent reductive amination yielded diastereomerically and enantiomerically pure amines. Compared to RU1968, the novel ethylenediamine 2d and homologous trimethylenediamine derivative 2e inhibited CatSper with similar and even twofold enhanced potency, respectively. Considering that these aminomethyl analogues are enantiomerically pure and much easier to synthesize than RU1968, we envisage their common use in future studies investigating the physiology of CatSper in sperm.
Keywords: sperm; CatSper; RU1968, steroids; Ca2+ flux; electrophysiology
The function of sperm is controlled by changes in the intracellular Ca2+ concentration ([Ca2+]i).1−4 Although the make-up of Ca2+-signaling pathways in sperm is quite diverse,5,6 the sperm-specific Ca2+ channel CatSper channel is a common component, controlling [Ca2+]i in sperm from many species including mammals.4,7,8 Across species, CatSper is activated by depolarization of the membrane potential and alkalization of the intracellular medium.9−12 Moreover, in human sperm, CatSper is also promiscuously activated by steroids and prostaglandins contained in reproductive fluids.13−18 It is unequivocal that in mice and humans, CatSper is required for fertilization: loss of CatSper function affects the swimming behavior of sperm and, thereby, causes male infertility.4,7,19−21 However, in humans, the precise role of CatSper and its control by steroids and prostaglandins during fertilization is still only ill-defined, and except for sea urchins,12 its function in species other than mice and humans is largely unknown. To address these questions, pharmacological tools, e.g., inhibitors, are required that allow interfering with the function of the channel. Of note, inhibitors that selectively target CatSper in sperm might not only serve as a tool for basic research but also to scrutinize CatSper as a target for novel contraceptives.
In the past decade, several compounds that inhibit CatSper have been discovered and employed to gain insights into the function of the channel, including for example NNC-0396 (NNC),16,18 Mibefradil,18 MDL12330A (MDL),22 HC-056456 (HC),23 and RU1968.24 Among these compounds, the steroidal sigma receptor ligand RU196825 represents the most thoroughly characterized and best-performing CatSper inhibitor and, thus, a promising lead structure.24 Of note, the action of RU1968 on sperm does not involve activation of sigma receptors.24,26 In brief, RU1968 rather directly inhibits CatSper across species with a half-maximal inhibitory concentration (IC50) of a few μM, lacks toxic side effects, suppresses CatSper-mediated motility responses, and mimics loss of CatSper function.24 Nevertheless, RU1968 also suffers from some shortcomings. In particular, the compound is not commercially available, and the synthesis reported by Rennhack et al.24 started from racemic estrone leading to racemic products. During synthesis, four diastereomeric pairs of enantiomers were formed and RU1968 had to be isolated from a complex mixture of stereoisomers. This demanding procedure has hampered the common (re-)synthesis and use of the drug: thus far, RU1968 has been utilized only in a few studies11,17,27,28 other than that by Rennhack et al.24 to investigate the function of CatSper.
Here, we report the design, synthesis, and functional characterization of RU1968 analogues that are easy to synthesize and do not require separation of stereoisomers. The novel enantiomerically pure CatSper inhibitors were derived by removal of the methyl moiety in the side chain and, thus, differ from RU1968 by one center of chirality (Figure 1), reducing the number of possible stereoisomers. We show that these compounds inhibit CatSper in human sperm with a similar efficacy and, depending on the modification, twofold enhanced potency compared to their congener RU1968.
Figure 1.
Design of novel CatSper inhibitors 2 derived from prototypical CatSper inhibitor RU1968 (1). In 2, the C-21 CH3 moiety (red circle) leading to a chiral center in the 20-position of RU1968 (1) is removed. * Compound 1 is a racemic mixture.
Results
Synthesis of RU1968 Analogues
The synthesis of aminomethyl derivatives 2a–c (Scheme 1) started with methylation of enantiomerically pure (+)-estrone (3) with CH3I under phase-transfer catalysis to obtain the methyl ether 4 in 90% yield. A Wittig reaction of ketone 4 with methoxymethyltriphenylphosphonium chloride afforded the enol ether 5, which was hydrolyzed with HCl to give the aldehyde 6 in 38% yield over two steps. The aldehyde 6 lacking the additional methyl moiety of RU1968 was isolated as a mixture of 17β and 17α diastereomers in the ratio 92:8. Reductive amination of the aldehyde 6 with pyrrolidine, 1-methylpiperazine, or 1-phenylpiperazine and NaBH(OAc)3 led to amines 7a–c in 41–74% yield. Removal of the methyl ether from 7a–c with concentrated HBr provided RU1968 analogues 2a–c in 7–56% yield (Scheme 1). The aminophenols 2a–c were isolated as mixtures of diastereomers in the ratio 91:9 to 98:2 (17β:17α). Unfortunately, it was not possible to separate the diastereomers at the stage of the aldehyde 6, the methyl ethers 7, or the phenols 2. Moreover, the amine 2d containing the RU1968 substituent was also prepared via this route, but the resulting diastereomeric amines could not be separated either.
Scheme 1. Synthesis of Enantiomerically Pure Estrol Derivatives 2a–c with an Aminomethyl Substituent in the 17-Positiona.
Reagents and reaction conditions: (a) CH3I, TBAI, CH2Cl2, NaOH, 70 °C, 5 h, 90%. (b) H3COCH2PPh3 Cl, KOtBu, THF, −20 °C to rt, 24 h. (c) HCl, THF/H2O, reflux, 40 min, 38% (over two steps). (d) R2NH, NaBH(OAc)3, HOAc, THF, Na2SO4, rt, 24 h, 74% (7a), 41% (7b) 42% (7c). (e) HBr, 100 °C, 6 h, 7% (2a), 52% (2b) 56% (2c).
To improve purification of the final products, the protective group of the phenol was modified. Instead of a methyl moiety, a benzyl moiety was introduced by alkylation of phenol 3 with benzyl bromide. Wittig reaction of benzylated ketone 8 and subsequent hydrolysis led to the aldehyde 10 (Scheme 2). Although a diastereomeric mixture of aldehyde 10 was formed, recrystallization provided pure 17β-configured aldehyde 10 in 48% yield. The configuration of aldehyde 10 was confirmed by X-ray crystal structure analysis (Figure 2). Amines 11d and 11e were prepared by reductive amination of aldehyde 10 with (N,N-dimethylamino)alkanamines and NaBH(OAc)3 in 68 and 77% yield, respectively. The final removal of the O-benzyl group with H2 and Pd/C catalysts produced single diastereomers of aminophenols 2d and 2e in 43 and 53% yield, respectively (Scheme 2).
Scheme 2. Synthesis of Enantiomerically Pure Aminomethyl Estranol Derivatives 2d and 2ea.
Reagents and reaction conditions: (a) BnBr, CH2Cl2, NaOH, 70 °C, 5 h, 90%. (b) H3COCH2PPh3 Cl, KOtBu, THF, −20 °C to rt, 24 h, 99%. (c) HCl, THF, reflux, 40 min, 48%. (d) R2NH, NaBH(OAc)3, HOAc, THF, Na2SO4, rt, 24 h, 68% (11d), 77% (11e). (e) H2, Pd/C, THF, rt, 3 h, 43% (2d), 53% (2e).
Figure 2.

X-ray crystal structure of aldehyde 10. Thermal ellipsoids are set at 30% probability. The structure clearly shows the β-orientation of the formyl moiety in the 17-position; i.e., 10 reveals (17S) configuration.
Action of RU1968 Analogues on CatSper in Human Sperm and on Human Slo3
We studied the action of compounds 2a–e in populations of human sperm loaded with a fluorescent Ca2+ indicator, using a fluorescence plate reader.15,18,24,29 The intracellular Ca2+ concentration ([Ca2+]i) was monitored before and after application of various concentrations of the respective compound and subsequent application of progesterone (2 μM) or PGE1 (2 μM) to evoke Ca2+ influx via CatSper.
At concentrations ≥4 μM, compound 2a evoked a small transient Ca2+ increase (Figure 3A,B), whose dose dependence, amplitude, and time course were reminiscent of the Ca2+ transient evoked by its congener RU1968 (see the study of Rennhack et al.24). The mechanism underlying these drug-evoked changes in [Ca2+]i is unknown but does not involve a toxic action, considering that, like RU1968,24 compound 2a as well as compounds 2b–e did not affect the viability of sperm (Supplementary Figure S1). Furthermore, compound 2a slowed down and completely suppressed in a dose-dependent fashion CatSper-mediated Ca2+ signals evoked by progesterone (Figure 3C) or prostaglandin E1 (Supplementary Figure S2A).
Figure 3.
Action of compounds 2a–e on the intracellular Ca2+ concentration and CatSper-mediated Ca2+ signals in human sperm. (A) Representative Ca2+ signals in human sperm loaded with a fluorescent Ca2+ indicator evoked by compound 2a and subsequent stimulation by progesterone (2 μM). ΔF/F (%) indicates the percentage change in fluorescence (ΔF) with respect to the mean basal fluorescence (F) before application of 2a. (B) Ca2+ signals evoked by 2a shown in (A) depicted on an extended time scale. (C) Progesterone-induced Ca2+ signals evoked in the presence of 2a shown in (A), depicted relative to the mean basal fluorescence before the application of progesterone. (D) Dose–response relation of the maximal Ca2+-signal amplitudes evoked by progesterone in the presence of 2a shown in (C), and dose–response relation of the PGE1-evoked Ca2+ signal in the presence of compound 2a. To increase comparability between experiments, Ca2+ signal amplitude is normalized to the control signal amplitude evoked in the absence of the compound (set to 1). (E) Representative Ca2+ signals evoked by compound 2b and subsequent application of progesterone. (F) Ca2+ signal evoked by 2b shown on an extended time scale. (G) Progesterone-induced Ca2+ signals in the presence of 2b. (H) Dose–response relation of normalized maximal signal amplitudes evoked by progesterone or PGE1 in the presence of 2b. (I) Representative Ca2+ signals evoked by compound 2c and subsequent application of progesterone. (J) Ca2+ signals evoked by 2c shown on an extended time scale. (K) Progesterone-induced Ca2+ signals in the presence of 2c. (L) Representative Ca2+ signals evoked by compound 2d and subsequent application of progesterone. (M) Ca2+ signal evoked by 2d shown on an extended time scale. (N) Progesterone-induced Ca2+ signals in the presence of 2d. (O) Dose–response relation of normalized maximal signal amplitudes evoked by progesterone or PGE1 in the presence of 2d. (P) Representative Ca2+ signals evoked by compound 2e and subsequent application of progesterone. (Q) Ca2+ signals evoked by 2e shown on an extended time scale. (R) Progesterone-induced Ca2+ signals in the presence of 2e. (S) Dose–response relation of normalized maximum signal amplitudes evoked by progesterone or PGE1 in the presence of 2e.
To quantify the inhibitory action of 2a on CatSper, we depicted and analyzed the progesterone- or PGE1-evoked increases in fluorescence relative to the baseline right before application of the ligand, correcting for 2a-induced changes in fluorescence (compare Figure 3A,C). Analysis of the dose–response relations yielded IC50 values of 7.6 ± 1.8 and 8.2 ± 1.8 μM (mean ± standard deviation, n = 3) for the inhibition of progesterone- and PGE1-evoked Ca2+ signals, respectively (Figure 3D; Table 1), i.e., about twofold larger than those reported for RU196824 (Table 1). Thus, compared to RU1968, compound 2a inhibits CatSper with similar efficacy but slightly reduced potency.
Table 1. IC50 Values of Compounds 2a–e for the Inhibition of Progesterone- and PGE1-Induced Ca2+ Signalsa.
n.d., IC50 values could not be determined; P4, progesterone; n = 3.
Like compound 2a, also compounds 2b (Figure 3E,F), 2c (Figure 3I,J), 2d (Figure 3L,M), and 2e (Figure 3P,Q) evoked small Ca2+ increases on their own. Yet, compared to 2a, the 2b- and 2c-evoked Ca2+ increase was longer lasting and rather brief, respectively, whereas for 2d and 2e, its amplitude was significantly smaller. Furthermore, compounds 2b (Figure 3E,G; Supplementary Figure S2B), 2d (Figure 3L,N; Supplementary Figure S2D), and 2e (Figure 3P,R; Supplementary Figure S2E) also suppressed in dose-dependent fashion Ca2+ signals evoked by progesterone or PGE1. RU1968 and 2d, which bear the same substituent, exhibit similar IC50 values, i.e., 3.5–4.0 μM (Figure 3O, Supplementary Figure S2D, and Table 1). Therefore, we conclude that the C-21 methyl group in RU1968 (see Figure 1, red circle) is neither essential for inhibition of CatSper nor does it affect the potency. For 2b, which structurally differs from 2d’s (dimethylamino)ethylamino moiety only by cyclization, the potency was reduced compared to 2d (and RU19681) and rather similar to that of 2a, i.e., ∼8 μM (Figure 3H,O,D; Supplementary Figure S2A, and Table 1). Thus, neither a methyl piperazine (2b) nor a pyrrolidine (2a) enhances the potency, indicating that cyclic motifs and loss of the second amine in the side chain are tolerated but not favorable. By contrast, the trimethylenediamine 2e (Figure 3R,S; Supplementary Figure S2E) featured IC50 values of ∼1.5 μM, representing an at least twofold gain in potency compared to 2d and RU1968, rendering it the most potent inhibitor of our new set (Table 1). Finally, the phenylpiperazine 2c did not inhibit Ca2+ signals evoked by progesterone (Figure 3I,K) or PGE1 (Supplementary Figure S2C), suggesting spatial limitations at CatSper’s inhibitory binding site.
Furthermore, the sperm-specific Slo3 channel represents the principal K+ channel in mouse30,31 and human sperm.32 RU1968 does not affect mouse Slo3 but inhibits human Slo3, yet, with about tenfold lower potency than CatSper in electrophysiological recordings.24 Therefore, we studied the action of 2d—as a representative of the series of RU1968 analogues—on human Slo3. We also studied the action of compound 2c, considering that it did not inhibit human CatSper.
To this end, we transiently expressed human Slo3 together with its auxiliary subunit LRRC52 in CHO cells and recorded K+ currents carried by Slo3 by whole-cell patch clamp recordings.30 Compound 2d suppressed Slo3 currents in a dose-dependent fashion with an IC50 of 10 ± 2 μM (n = 5, Figure 4A,B, Table 1), indicating that the RU1968 analogues also inhibit Slo3. Yet, the slight, statistically insignificant increase in the IC50 value of 2d versus RU1968 (7 ± 6 μM)24 might point toward a small gain in selectivity for CatSper over Slo3. Remarkably, compound 2c not only failed to inhibit CatSper but also Slo3 (Figure 4C,D), highlighting the overlapping pharmacology of the inhibitory binding sites of the channels in humans.
Figure 4.

Action of compounds 2d and 2c on heterologous human Slo3. (A) Representative current voltage relation recorded from a CHO cell transiently expressing human Slo3 in the absence and presence of different concentrations of 2d. (B) Dose–response relation of the suppression of mean Slo3 current amplitudes by 2d (n = 4); amplitudes are depicted relative to that recorded in the absence of 2d (set to 1). (C) Representative current voltage relation recorded from a CHO cell transiently expressing human Slo3 channels in the absence (control) and presence of 2d. (D) Mean current amplitude recorded in the presence of 2c (100 μM) relative to that in its absence (set to 1) (n = 4).
Discussion
Starting from enantiomerically pure (+)-estrone, we established a straightforward synthesis and isolation of diastereomerically and enantiomerically pure analogues of RU1968. Compared to RU1968 (1), the ethylenediamine 2d (IC50 = 3.8 μM) and homologous trimethylenediamine 2e (IC50 = 1.7 μM) inhibited CatSper with similar and twofold enhanced potency, respectively. Considering that 2d (TS116) and 2e (TS150) are much easier to synthesize and isolate, we envisage their common use instead of RU1986 in future studies investigating the role of CatSper.
Of note, future studies are also required to further improve the properties of this class of CatSper inhibitors. For example, by an unknown mechanism, RU1968 and its analogues 2 evoke small Ca2+ signals on their own. This reinforces that the pharmacology of CatSper is complex. The channel seems to harbor several activator- and inhibitor-binding sites, which might be allosterically coupled. Compounds of the RU1968 class might engage an inhibitory site and, at the same time, also represent weakly efficacious agonists, causing small elevations of [Ca2+]i on their own. However, their precise mechanism and site of action on CatSper seems difficult to elucidate by structure–function analysis or site-directed mutagenesis, considering that CatSper resists functional expression.
Yet, the cryo-electron microscopy (cryo-EM) structure of the CatSper complex isolated from mouse sperm was recently achieved.33 The cryo-EM analysis of CatSper in the presence of small-molecule modulators such as RU1968 might identify their binding sites and, thus, facilitate the synthesis of novel analogues based on structure-based drug design.34
Moreover, like RU1968, also its analogues are not perfectly selective for CatSper. In human sperm, the compounds also inhibit Slo3. To further improve the inhibitor’s selectivity for CatSper, a structure–activity analysis is required to identify RU1968 analogues that do not act on human Slo3. The fact that human Slo3 can be functionally expressed in cultured cells facilitates this endeavor. Furthermore, RU196825 and most likely also its new analogues act on sigma receptors expressed in somatic cells. This action would need to be abolished prior to an in vivo use of the compounds.
Finally, only recently, a high-throughput screening (HTS) approach and ensuing comprehensive structure–activity and selectivity analysis identified seven new classes of CatSper inhibitors.35 One of these classes of compounds inhibited CatSper at low micromolar concentrations, whereas at 10 μM, the drug did not affect human Slo3. If the drug might inhibit human Slo3 at >10 μM remains to be determined by studying its action on Slo3 in a dose-dependent fashion. Nevertheless, this HTS-based approach yielded another promising lead compound to develop potent and highly selective CatSper inhibitors for basic research and potential application as contraceptives.
Materials and Methods
Chemical Synthesis
Unless otherwise noted, moisture-sensitive reactions were conducted under dry nitrogen. Thin-layer chromatography (tlc): silica gel 60 F254 plates (Merck). Flash chromatography (fc): silica gel 60, 40–64 μm (Merck); parentheses include the diameter of the column (d), fraction size (v), eluent, and Rf value. Melting point: melting point apparatus MP50 (Mettler Toledo), uncorrected. MS: MAT GCQ (Thermo-Finnigan): EI, MAT LCQ (Thermo Finnigan): ESI. 1H NMR (400 MHz), 13C NMR (100 MHz): Agilent DD2 400 and 600 MHz spectrometers.; δ in ppm related to tetramethylsilane; coupling constants are given with 0.5 Hz resolution; the assignments of 13C and 1H NMR signals were supported by 2D NMR techniques. IR: FT/IR IRAffinity-1 IR spectrometer (Shimadzu) using an ATR technique. Optical rotation: Polarimeter UniPoL 1000 (Schmidt Haensch); 1.0 dm tube; concentration c in g/100 mL; T = 20 °C; wavelength 589 nm (D-line of Na light); the unit of the specific rotation ([α]DT ° mL dm–1 g–1) is omitted for clarity.
HPLC Analysis of Compound Purities
HPLC: Merck Hitachi Equipment; UV detector: L-7400; autosampler: L-7200; pump: L-7100; degasser: L-7614; column: LiChrospher 60 RP-select B (5 μm); LiChroCART 250–4 mm cartridge; flow rate: 1.0 mL/min; injection volume: 5.0 or 10 μL; detection at λ = 210 nm; solvents: A: water with 0.05% (v/v) trifluoroacetic acid; B: acetonitrile with 0.05% (v/v) trifluoroacetic acid: gradient elution: (A %): 0–4 min: 90%, 4–29 min: 90 → 0%, 29–31 min: 0%, 31–31.5 min: 0 → 90%, 31.5–40 min: 90%. Data acquisition: HSM software; manual integration. The purity of all test compounds was >95%.
Synthetic Procedures
3-Methoxyestra-1,3,5(10)-trien-17-one (4)
A suspension of estrone (3, 1.0 g, 3.70 mmol, 1.0 equiv), TBAI (0.07 g, 0.19 mmol, 0.05 equiv), an aqueous solution of NaOH (10%w/w, 20 mL), and CH3I (0.87 mL, 14.1 mmol, 3.8 equiv) were added in CH2Cl2. After heating to 70 °C for 5 h, the mixture was cooled down to room temperature. Layers were separated, and the aqueous phase was extracted with CH2Cl2 (3 × 70 mL). The organic layers were combined, dried (Na2SO4), and filtered, and the solvent was evaporated under reduced pressure. The product was not further purified. (TLC: cyclohexane/ethyl acetate 9:1, Rf = 0.28). C19H24O2 (284.4 g/mol). Colorless solid, mp 170–171 °C, yield 1.04 g (90%). 1H NMR (400 MHz, CDCl3): δ [ppm] = 0.91 (s, 3H, 18-CH3), 1.37–1.71 (m, 6H, 7-CH2 (1H), 8-CH, 11-CH2 (1H), 12-CH2 (1H), 14-CH, 15-CH2 (1H)), 1.88–2.31 (m, 5H, 7-CH2 (1H), 9-CH, 12-CH2 (1H), 15-CH2 (1H), 16-CH2 (1H)), 2.35–2.56 (m, 2H, 11-CH2 (1H), 16- CH2 (1H)), 2.86–2.95 (m, 2H, 6-CH2), 3.78 (s, 3H, OCH3), 6.62–6.68 (d, J = 2.7 Hz, 1H, 4-CH), 6.72 (dd, J = 8.6/2.8 Hz, 1H, 2-CH), 7.21 (dd, J = 8.7/1.1 Hz, 1H, 1-CH). 13C NMR (101 MHz, CDCl3): δ [ppm] = 13.8 (1C, C-18), 21.6 (1C, C-15), 25.8 (1C, C-11), 26.4 (1C, C-7), 29.5 (1C, C-6), 31.4 (1C, C-12), 35.9 (1C, C-16), 38.2 (1C, C-8), 43.8 (1C, C- 9), 47.9 (1C, C-13), 50.3 (1C, C-14), 55.0 (1C, OCH3), 111.4 (1C, C-2), 113.7 (1C, C-4), 126.2 (1C, C-1), 131.9 (1C, C-10), 137.6 (1C, C-5), 157.4 (1C, C-3), 220.7 (1C, 17-C=O). IR (neat): ν̃ [cm–1] = 2955 (C–Haliph), 2916 (C–Haliph), 2874(C–Haliph), 1730 (C=O), 1504 (C=Carom), 1454 (C=Carom). Exact mass (APCI): (m/z) = 285.1863 (calcd. 285.1849 for C19H25O2 [M + H]+). Purity (HPLC): tR = 22.4 min, purity 97.8%.
3-Methoxyestra-1,3,5(10)-triene-17-carbaldehyde (6)
Under N2, a suspension of Wittig reagent CH3OCH2PPh3Cl (1.80 g, 5.25 mmol, 1.50 equiv) in tetrahydrofuran (THF) (4.20 mL) was cooled to −15 °C. Then, KOtBu (0.59 g, 5.25 mmol, 1.50 equiv) was added to the suspension. The mixture was stirred for 15 min. Ketone 4 (1.00 g, 3.35 mmol, 1.0 equiv) was added, and the cooling was removed after 1 h. After stirring overnight, water was added followed by the extraction with cyclohexane (3 × 30 mL). The organic layers were combined, dried (Na2SO4), and filtered, and the solvent was removed in vacuo. A yellow solid was obtained. The residue was purified by flash chromatography (Ø = 5 cm, h = 20 cm, V = 20 mL, cyclohexane/ethyl acetate = 99:1 → 98.5:1.5 → 97.5:2.5 → 97:3 → 95:5). A transparent waxy solid (enol ether 5) was obtained. The crude product was dissolved in THF (60 mL). Then, 20 mL of a 5 %V/V HCl (20 mL) was added. The mixture was heated to 73 °C for 40 min. The mixture was extracted with cyclohexane (3 × 30 mL). The combined organic layers were dried (Na2SO4), filtered, and concentrated in vacuo, and the residue was purified using a flash column (Ø = 5 cm, h = 18 cm, V = 20 mL, cyclohexane/ethyl acetate = Ø = 5 cm, h = 18 cm, V = 20 mL, cyclohexane/ethyl acetate = 99:1 → 98.5:1.5 → 97.5:2.5 → 97:3 → 95:5, cyclohexane/ethyl acetate 9:1 Rf = 0.50). Colorless solid, mp 122–123 °C, yield 400 mg (38%). C20H26O2 (298.4 g/mol). 1H NMR (600 MHz, CDCl3): δ [ppm] = 0.80 (s, 3 × 0.92 H, 18–CH3), 0.96 (s, 3 × 0.08 H, 18–CH3), 1.32–1.56 (m, 5H, 7-CH2 (1H), 8-CH, 11-CH2 (1H), 14-CH, 16-CH2 (1H)), 1.65 (td, J = 12.9/4.0 Hz, 1H, 12-CH2 (1H)), 1.73–1.96 (m, 3H, 7-CH2 (1H), 11-CH2 (1H), 15-CH2 (1H)), 2.10–2.22 (m, 2H, 12-CH2 (1H), 15-CH2 (1H)), 2.24–2.30 (m, 2H, 9-CH), 2.34 (dtd, J = 13.4/4.1/2.8 Hz, 1H, 16–CH2), 2.39 (td, J = 9.2/2.1 Hz, 1H, 17-CH), 2.81–2.93 (m, 2H, 6-CH2), 3.78 (s, 3H, OCH3), 6.59–6.65 (d, J = 2.8 Hz, 1H, 4-CH), 6.7 (dd, J = 8.6/2.9 Hz, 1H, 2-CH), 7.17–7.22 (m, 1H, 1-CH), 9.82 (s, 1 H, HC=O). 13C NMR (151 MHz, CDCl3): δ [ppm] = 14.3 (1C, C-18), 21.5 (1C, C-15), 24.9 (1C, C-11), 26.5 (1C, C-16), 28.4 (1C, C-7), 29.9 (1C, C-6), 38.4 (1C, C-12), 38.8 (1C, C-8), 44.2 (1C, C- 9), 45.4 (1C, C-13), 55.6 (2C, C-14, OCH3), 63.3 (1C, C-17), 111.9 (1C, C-2), 114.2 (1C, C-4), 126.6 (1C, C-1), 132.7 (1C, C-10), 138.2 (1C, C-5), 157.9 (1C, C-3), 205.2 (1C, C=O). IR (neat): ν̃ [cm–1] = 2934 (C–Haliph), 2913 (C–Haliph), 2870 (C–Haliph), 1734 (C=O), 1472 (C=Carom), 1447 (C=Carom). Exact mass (APCI): (m/z) = 299.1987 (calcd. 299.2006 for C20H27O2 ([M + H]+). Purity (HPLC): tR = 23.5 min, purity 95.5%.
1-[(3-Methoxyestra-1,3,5(10)-trien-17-yl)methyl]pyrrolidine (7a)
Aldehyde 6 (194 mg, 0.65 mmol, 1.0 equiv), pyrrolidine (0.267 mL, 3.25 mmol, 5.0 equiv), CH3COOH (0.112 mL, 1.95 mmol, 3.0 equiv), and NaBH(OAc)3 (0.83 g, 3.90 mmol, 6.0 equiv) were suspended in THF (3 mL). The mixture was stirred at room temperature for 24 h. H2O (20 mL) was added, and the mixture was extracted with CH2Cl2 at a basic pH value of 12 (3 × 20 mL). The organic layers were combined, dried (Na2SO4), and filtered. The solvent was removed in vacuo, and the residue was purified by flash chromatography (Ø = 2 cm, h = 18 cm, V = 10 mL, CH2Cl2/CH3OH = 9/1, Rf = 0.75).
Colorless solid, 143–144 °C, yield 170 mg (74%). C24H35NO (353.3 g/mol). 1H NMR (600 MHz, CDCl3): δ [ppm] = 0.63 (s, 3 × 0.81 H, 18–CH3), 0.82 (s, 3 × 0.19 H, 18-CH3), 1.19–1.58 (m, 7H, 7- CH2 (1H), 8-CH, 11-CH2 (1H), 12-CH2 (1H), 14-CH, 15-CH2 (1H), 16-CH2 (1H)), 1.74–2.08 (m, 8H, 7-CH2 (1H), 12-CH2 (1H), 15-CH2 (1H), 16-CH2 (1H), N(CH2CH2)2 (4H)), 2.16–2.31 (m, 2H, 9-CH, 11-CH2 (1H)), 2.39 (m, 1H, R2CH2N), 2.55–2.67 (m, 5H, N(CH2CH2)2 (4H), 17-CHCH2N (1H)), 2.79–2.91 (m, 2H, 6-CH2), 3.77 (s, 3H, OCH3), 6.62 (d, J = 2.7 Hz, 1H, 4-CH), 6.70 (dd, J = 8.6/2.8 Hz, 1H, 2-CH), 7.20 (dd, J = 8.6/1.1 Hz, 1H, 1-CH). 13C NMR (151 MHz, CDCl3): δ [ppm] = 12.5 (1C, 18-CH3), 23.6 (2C, N(CH2CH2)2, 24.7 (1C, C-15), 26.5 (1C, C-11), 28.0 (1C, C-7), 28.7 (1C, C- 16), 30.1 (1C, C-6), 38.0 (1C, C-12), 38.8 (1C, C-8), 44.2 (1C, C-9), 45.7 (1C, C-13), 50.3 (1C, C-17), 54.9 (2C, N(CH2CH2)2), 55.3 (1C, C-14), 58.3 (1C, 17-CHCH2N), 111.6 (1C, C-2), 113.9 (1C, C-4), 126.4 (1C, C-1), 133.1 (1C, C-10), 138.2 (1C, C- 5), 157.5 (1C, C-3). IR (neat): ν̃ [cm–1] = 2932 (C–Haliph), 2866 (C–Haliph), 2778 (C–Haliph), 1501 (C=Carom), 1447 (C=Carom). Exact mass (APCI): (m/z) = 354.2791 (calcd. 354.2791 for C24H36 NO [M + H]+). Purity (HPLC): tR = 20.8 min, purity 95.1%.
1-[(3-Methoxyestra-1,3,5(10)-trien-17-yl)methyl]-4-methylpiperazine (7b)
Aldehyde 6 (170 mg, 0.61 mmol, 1.0 equiv), 1-methylpiperazine (0.34 mL, 3.00 mmol, 5.0 equiv), CH3COOH (0.105 mL, 1.80 mmol, 3.0 equiv), and NaBH(OAc)3 (0.776 g, 3.66 mmol, 6.0 equiv) were suspended in THF (3 mL). The mixture was stirred at room temperature for 24 h. Water was added, and the mixture was extracted with CH2Cl2 at a basic pH value of 12 (3 × 25 mL). The organic layers were combined, dried (Na2SO4), and filtered. The solvent was removed in vacuo, and the residue was purified by flash chromatography (Ø = 2 cm, h = 18 cm, V = 10 mL, CH2Cl2/CH3OH = 9/1, Rf = 0.43).
Colorless solid, mp 118–119 °C, yield 90 mg (41%). C25H38N2O (382.3 g/mol). 1H NMR (400 MHz, CDCl3): δ [ppm] = 0.64 (s, 3 × 0.88 H, 18–CH3), 0.82 (s, 3 × 0.12 H, 18–CH3), 1.17–1.56 (m, 7H, 7- CH2 (1H), 8-CH, 11-CH2 (1H), 12-CH2 (1H), 14-CH, 15-CH2 (1H), 16-CH2 (1H)), 1.60–1.81 (m, 2H, 15-CH2 (1H), 17-CH), 1.81–1.97 (m, 2H, 7-CH2 (1H), 16-CH2 (1H)), 1.99–2.10 (m, 1H, 12-CH2), 2.14–2.28 (m, 3H, 9-CH, 11-CH2 (1H), R2CH2N(1H)), 2.31 (s, 3H, NCH3), 2.40–2.58 (m, 9H, R2CH2N(1H), N(CH2CH2)2N (8H)), 2.77–2.94 (m, 2H, 6-CH2), 3.77 (s, 3H, OCH3), 6.60–6.64 (d, J = 2.8, 1H, 4-CH), 6.70 (dd, J = 8.6/2.8 Hz, 1H, 2-CH) 7.20 (dd, J = 8.7/1.1 Hz, 1H, 1-CH). 13C NMR (101 MHz, CDCl3): δ [ppm] = 12.1 (1C, 18-CH3), 24.4 (1C, C-15), 26.3 (1C, C-11), 27.7-28.2 (2C, C-7, C-16), 29.7 (1C, C-6), 37.9 (1C, C-12), 38.5 (1C, C-8), 42.4 (1C, C-13), 44.0 (1C, C-9), 45.7 (1C, NCH3), 47.0 (1C, C-17), 53.254.7 (4C, N(CH2CH2)2N), 54.9 (1C, C-14), 55.0 (1C, OCH3), 60.1 (1C, R2CH2N), 111.2 (1C, C-2), 113.6 (1C, C-4), 126.1 (1C, C-1), 132.8 (1C, C-10), 137.9 (1C, C-5), 157.2 (1C, C-3). IR (neat): ν̃ [cm–1] = 2932 (C–Haliph), 2866 (C–Haliph), 2843 (C–Haliph), 1504 (C=Carom), 1447 (C=Carom). Exact mass (APCI): (m/z) = 383.3050 (calcd. 383.3057 for C22H32 N2O [M + H]+). Purity (HPLC): tR = 17.0 min, purity 97.1%.
1-[(3-Methoxyestra-1,3,5(10)-trien-17-yl)methyl]-4-phenylpiperazine (7c)
Aldehyde 6 (150 mg, 0.50 mmol, 1.0 equiv), 1-phenylpiperazine (0.28 mL, 2.50 mmol, 5.0 equiv), CH3COOH (0.0859 mL, 1.5 mmol, 3.0 equiv), and NaBH(OAc)3 (0.636 g, 3.00 mmol, 6.0 equiv) were suspended in THF (3 mL). The mixture was stirred at room temperature for 24 h. Water was added, and the mixture was extracted with CH2Cl2 at a basic pH value of 12 (3 × 25 mL). The organic layers were combined, dried (Na2SO4), and filtered. The solvent was removed in vacuo, and the residue was purified by flash chromatography (Ø = 2 cm, h = 18 cm, V = 10 mL, CH2Cl2/CH3OH = 9/1, Rf = 0.78). Colorless solid, yield 94 mg (42%). C30H40N2O (444.7 g/mol). 1H NMR (400 MHz, CDCl3): δ [ppm] = 0.67 (s, 3 × 0.90 H, 18-CH3), 0.84 (s, 3× 0.10 H, 18-CH3), 1.18–1.57 (m, 7H, 7- CH2 (1H), 8-CH, 11-CH2 (1H), 12-CH2 (1H), 14-CH, 15-CH2 (1H), 16-CH2 (1H)), 1.68–1.83 (m, 2H, 15-CH2 (1H), 17-CH), 1.85–2.00 (m, 2H, 7-CH2 (1H), 16-CH2 (1H)), 2.08 (dt, J = 12.6/3.1 Hz, 1H, 12-CH2), 2.15–2.32 (m, 3H, 9-CH, 11-CH2 (1H), 17-CHCH2NR2 (1H)), 2.43–2.69 (m, 5H, 17-CHCH2NR2 (1H), N(CH2CH2)2N (4H)), 2.78–2.95 (m, 2H, 6-CH2), 3.20 (t, J = 5.0 Hz, 4H, N(CH2CH2)2N (4H)), 3.78 (s, 3H, OCH3), 6.63 (d, J = 2.7 Hz, 1H, 4-CH), 6.71 (dd, J = 8.6/2.8 Hz, 1H, 2-CH), 6.85 (tt, J = 7.3, 1.1 Hz, 1H, C-4ph), 6.90–6.98 (m, 2H, Carom), 7.16–7.23 (dd, J = 9.2/1.2, 1H, 1-CH), 7.24–7.30 (m, 2H, Carom). 13C NMR (101 MHz, CDCl3): δ [ppm] = 12.1 (1C, 18-CH3), 24.4 (1C, C-15), 26.3 (1C, C-11), 27.7 (1C, C-16), 28.1 (1C, C-7), 29.7 (1C, C-6), 38.1 (1C, C-12), 38.5 (1C, C-8), 42.4 (1C, C-13), 44.0 (1C, C-9), 47.1 (1C, C-17), 49.0 (2C, N(CH2CH2)2N), 53.6 (2C, N(CH2CH2)2N), 54.79 (1C, OCH3), 55.0 (1C, C-14), 60.2 (1C, 17-CHCH2NR2), 111.2 (1C, C-2), 113.6 (1C, C-4), 115.8 (2C, Carom), 119.3 (1C, Carom), 126.1 (1C, C-1), 128.9 (2C, Carom), 132.9 (1C, C- 10), 137.9 (1C, C-5), 151.3 (1C, Carom), 157.2 (1C, C-3). IR (neat): ν̃ [cm–1] = 2936 (C–Haliph), 2816 (C–Haliph), 1497 (C=Carom). Exact mass (APCI): (m/z) = 445.3198 (calcd. 445.3213 for C22H32 N2O [M + H]+). Purity (HPLC): tR = 22.7 min, purity 97.2%.
17-[(Pyrrolidin-1-yl)methyl]estra-1,3,5(10)-trien-3-ol (2a)
Amine 7a (150 mg, 0.34 mmol) was suspended in an excess of HBr 48%. The mixture was heated to 100 °C in a sealed tube for 5 h. After cooling to room temperature, the mixture was neutralized to pH 7–8 with a saturated solution of Na2CO3 and extracted with ethyl acetate (3 × 25 mL). The organic layers were combined, dried (Na2SO4), and filtered. The solvent was removed in vacuo, and the residue was recrystallized from CH3OH (TLC: CH2Cl2/CH3OH = 9/1 + 2 drops of ammonia, Rf = 0.54). Colorless solid, mp 141–142 °C (decomposition), yield 9.0 mg (7%). C23H33NO (339.5 g/mol). 1H NMR (400 MHz, DMSO-D6): δ [ppm] = 0.60 (s, 3 × 0.91 H, 18–CH3), 0.78 (s, 3 × 0.09 H, 18-CH3), 1.12–1.40 (m, 7H, 7-CH2, 8-CH, 11-CH2, 12-CH2, 14-CH, 15-CH2, 16-CH2), 1.46–1.69 (m, 6H, 15-CH2, 17-CH, N(CH2CH2)2), 1.82 (ddd, J = 18.6, 12.5, 8.0 Hz, 2H, 7-CH2, 16-CH2), 1.92–2.01 (m, 1H, 12-CH2), 2.09 (d, J = 7.1 Hz, 1H, 9-CH), 2.16–2.31 (m, 2H, 11-CH2, 17-CHCH2N), 2.34–2.42 (m, 4H, N(CH2CH2)2), 2.42–2.47 (m, 1H, 17-CHCH2N), 2.67–2.80 (m, 2H, 6-CH2), 6.42 (d, J = 2.6 Hz, 1H, 4-CH), 6.49 (dd, J = 8.4/2.7 Hz, 1H, 2-CH), 6.99–7.06 (dd, J = 8.4/1.1 1H, 1-CH), 8.96 (s, 1H, OH). 13C NMR (151 MHz, DMSO-D6): δ [ppm] = 12.1 (1C, C-18), 23.1 (1C, N(CH2CH2)2) 24.0 (1C, C-15), 26.2 (1C, C-11), 27.5 (1C, C-16), (1C, C-7), 29.2 (1C, C-6), 37.7 (1C, C-12), 38.4 (1C, C-8), 42.0 (1C, C-13), 43.7 (1C, C-9), 49.3 (1C, C-17), 53.8 (2C, N(CH2CH2)2), 54.4 (1C, C-14), 57.5 (1C, 17-CHCH2N), 112.7 (1C, C-2), 114.9 (1C, C-4), 126.0 (1C, C-1), 130.5 (1C, C-10), 137.1 (1C, C-5), 154.9 (1C, C-3). IR (neat): ν̃ [cm–1] = 2928 (C–Haliph), 1582 (C=Carom), 1470 (C=Carom). Exact mass (APCI): (m/z) = [M + H]+ calculated for C23H34NO, 340.2635; found 340.2668. Purity (HPLC): tR1 = 17.5 min, diastereomer 1, intensity 91.8%, tR2 = 17.1 min, diastereomer 2, intensity 2.3%.
17-[(4-Methylpiperazin-1-yl)methyl]estra-1,3,5(10)-trien-3-ol (2b)
Amine 7b (130 mg, 0.43 mmol) was suspended in an excess of HBr 48%. The mixture was heated to 100 °C in a sealed tube for 5 h. After cooling down to room temperature, the mixture was neutralized to pH 7–8 with a saturated solution of Na2CO3 and extracted with ethyl acetate (3 × 30 mL). The organic layers were combined, dried (Na2SO4), and filtered. The solvent was removed in vacuo. (TLC: CH2Cl2/CH3OH = 9/1, Rf = 0.58). Colorless solid, mp 200–201 °C (decomposition), yield 65 mg (52%).
C24H36N2O (368.6 g/mol). 1H NMR (600 MHz, DMSO-D6): δ [ppm] = 0.59 (s, 3 × 0.93 H, 18–CH3), 0.78 (s, 3 × 0.08 H, 18–CH3), 1.10–1.36 (m, 7H, 7-CH2, 8-CH, 11-CH2, 12-CH2, 14-CH, 15-CH2, 16-CH2), 1.60–1.71 (m, 2H, 15–CH2, 17-CH), 1.75 (m, 2H, 7-CH, 11-CH2), 1.94–2.12 (m, 3H, 17-CHCH2N, 9-CH, 12-CH2), 2.14 (s, 3H, NCH3), 2.15–2.46 (m, H, 16-CH2, N(CH2CH2)2N (8H), 17-CHCH2N), 2.66–2.75 (m, 2H, 6-CH2), 6.40 (d, J = 2.6 Hz, 1H, 4-CH), 6.47 (dd, J = 8.4/2.6 Hz, 1H, 2-CH), 7.01 (d, J = 8.5 Hz, 1H, 1-CH), 8.95 (s, 1H, OH). 13C NMR (151 MHz, DMSO-D6): δ [ppm] = 12.0 (1C, 18-CH3), 24.0 (1C, C-15), 26.2 (1C, C-11), 27.5 (2C, C-7, C-16), 29.2 (1C, C-6), 37.8 (1C, C-12), 38.4 (1C, C-13), 40.0 (1C, NCH3), 42.0 (1C, C-8), 43.7 (1C, C-9), 45.63, 46.4 (1C, C-17), 53.1 (2C, N(CH2CH2)2N), 54.5 (1C, C-14), 54.8 (2C, N(CH2CH2)2N), 59.8 (1C, 17-CHCH2N), 112.7 (1C, C-2), 114.9 (1C, C-4), 126.0 (1C, C-1), 130.5 (1C, C-10), 137.1 (1C, C-5), 154.9 (1C, C-3). IR (neat): ν̃ [cm–1] = 2928 (O–H), 2851 (C–Haliph), 2820 (C–Haliph), 1462 (C=Carom), 1442 (C=Carom). Exact mass (APCI): (m/z) = 369.2933 (calcd. 369.2900 for C22H32 N2O [M + H]+). Purity (HPLC): tR1 = 14.1 min, diastereomer 1, intensity 93.1%; tR2 = 17.1 min, diastereomer 2, intensity 5.8%.
17-[(4-Phenylpiperazin-1-yl)methyl]estra-1,3,5(10)-trien-3-ol (2c)
Amine 7c (94 mg, 0.33 mmol) was suspended in an excess of HBr 48%. The mixture was heated to 100 °C in a sealed tube for 5 h. After cooling to room temperature, the mixture was neutralized to pH 7–8 with a saturated solution of Na2CO3 and extracted with ethyl acetate (3 × 30 mL). The organic layers were combined, dried (Na2SO4), and filtered. The solvent was removed in vacuo. (TLC: CH2Cl2/CH3OH = 9/1, Rf = 0.65). Colorless solid, mp 210–211 °C (decomposition), yield 51 mg (56%). C29H38N2O (430.6 g/mol). 1H NMR (600 MHz, DMSO-D6): δ [ppm] = 0.64 (s, 3 × 0.98 H, 18-CH3), 0.80 (s, 3 × 0.02 H, 18-CH3), 1.19–1.37 (m, 7H, 7-CH2, 8-CH, 11-CH2, 12-CH2, 14-CH, 15-CH2, 16-CH2), 1.65–1.75 (m, 2H, 15-CH2, 17-CH), 1.76–1.85 (m, 2H, 7-CH2, 16-CH2), 2.07–2.14 (m, 2H, 9-CH, 12-CH2), 2.15–2.23 (m, 2H, 11-CH2, 17-CHCH2N), 2.40–2.56 (m, 5H, 17-CHCH2N, N(CH2CH2)2N), 2.65–2.77 (m, 2H, 6-CH2), 3.11 (t, J = 5.1 Hz, 4H, N(CH2CH2)2N), 6.43 (d, J = 2.6 Hz, 1H, 4-CH), 6.49 (dd, J = 8.4/2.6 Hz, 1H, 2-CH), 6.76 (t, J = 7.2 Hz, 1H, 4-CHph), 6.92 (d, J = 8.2 Hz, 2H, 2-CHph, 6-CHph), 7.03 (d, J = 8.4 Hz, 1H, 1-CH), 7.13–7.23 (m, 2H, 3-CHph, 5-CHph), 9.87 (s, 1H, OH). 13C NMR (151 MHz, DMSO-D6): δ [ppm] = 11.79 (1C, C-18), 23.78 (1C, C-15), 25.87 (1C, C-11), 27.24 (2C, C-7, C-16), 28.92 (1C, C-6), 37.50 (1C, C-12) 38.16 (1C, C-8), 41.86 (1C, C-13), 43.39 (1C, C-9), 46.16 (1C, C-17) 48.01 (2C, N(CH2CH2)2N), 53.01 (2C, N(CH2CH2)2N), 54.16 (1C, C-14), 59.57 (1C, 17-CHCH2N) 112.41 (1C, C-2), 114.63 (1C, C-4), 115.02 (1C, C-1), 118.42 (1C, Cph), 125.71 (2C, Cph), 128.62 (2C, Cph), 130.20 (1C, C-10) 136.85 (1C, C-5), 145.9 (1C, Cph), 154.60 (1C, C-3). IR (neat): ν̃ [cm–1] = 3209 (O–H), 2924 (C–Haliph), 2831 (C–Haliph), 1497 (C=Carom), 1450 (C=Carom). Exact mass (APCI): (m/z) = 431.3049 (calcd. 431.3057 for C22H32 N2O [M + H]+). Purity (HPLC): tR1 = 20.0, min, diastereomer 1, intensity 84.9%, tR2 = 22.6 min, diastereomer 2, intensity 11.9%.
3-Benzyloxyestra-1,3,5(10)-trien-17-one (8)
Tetrabutylammonium bromide (0.596 g, 1.85 mmol, 0.1 equiv), benzyl bromide (4.40 mL, 6.33 g, 37 mmol, 2 equiv), and 10% aqueous NaOH solution (90 mL) were added to a suspension of estrone (3, 5.00 g, 18.5 mmol, 1.0 equiv) in CH2Cl2 (90 mL). The mixture was heated to reflux for 6 h. After cooling to room temperature (rt), layers were separated, and the aqueous phase was extracted with CH2Cl2 (3 × 50 mL). The organic phases were combined, washed with brine (50 mL), dried (Na2SO4), and filtered, and the solvent was removed in vacuo. The residue was recrystallized from 96% ethanol. Colorless crystalline solid, mp 128–130 °C, yield 6.00 g (90%). Rf (cHex/EtOAc 7:3) = 0.34. C25H28O2 (360.5). 1H NMR (600 MHz, CDCl3): δ (ppm) = 0.91 (s, 3H, CH3), 1.39–1.69 (m, 6H, 7-CH2 (1H), 8-CH (1H), 11-CH2 (1H), 12-CH2 (1H), 14-CH (1H), 15-CH2 (1H)), 1.92–1.98 (m, 1H, 12-CH2), 2.01 (ddt, J = 12.8/5.7/2.9 Hz, 1H, 7-CH2), 2.06 (dddd, J = 11.9/8.8/5.5/1.0 Hz, 1H, 15-CH2), 2.15 (dt, J = 19.0/9.0 Hz, 1H, 16-CH2), 2.26 (td, J = 10.8/4.4 Hz, 1H, 9-CH), 2.36–2.43 (m, 1H, 11-CH2), 2.51 (ddd, J = 19.1/8.8/1.0 Hz, 1H, 16-CH2), 2.85–2.96 (m, 2H, 6-CH2), 5.04 (s, 2H, OCH2), 6.74 (d, J = 2.8 Hz, 1H, 4-CH), 6.80 (dd, J = 8.6/2.8 Hz, 1H, 2-CH), 7.21 (d, J = 8.7 Hz, 1H, 1-CH), 7.30–7.35 (m, 1H, 4-Hbenzyl), 7.35–7.41 (m, 2H, 3-Hbenzyl, 5-Hbenzyl), 7.41–7.46 (m, 2H, 2-Hbenzyl, 6-Hbenzyl). 13C NMR (151 MHz, CDCl3): δ (ppm) = 14.0 (1C, C-18), 21.7 (1C, C-15), 26.1 (1C, C-11), 26.7 (1C, C-7), 29.8 (1C, C-6), 31.7 (1C, C-12), 36.0 (1C, C-16), 38.5 (1C, C-8), 44.1 (1C, C-9), 48.2 (1C, C-13), 50.6 (1C, C-14), 70.1 (1C, PhCH2O), 112.5 (1C, C-2), 115.0 (1C, C-4), 126.0 (1C, C-1), 127.6 (2C, C-2benzyl, C-6benzyl), 128.0 (1C, C-4benzyl), 128.7 (2C, C-3benzyl, C-5benzyl), 132.5 (1C, C-10), 137.4 (1C, C-1benzyl), 137.9 (1C, C-5), 157.0 (1C, C-3), 221.1 (1C, C-17). Exact mass (ESI): (m/z) = 361.2191 (calcd. 361.2162 for C25H29O2 [M + H+]). IR (neat): ν̃ [cm–1] = 2920 (C–Haliph), 2870 (C–Haliph), 1732 (C=O), 1601 (C=Carom). Purity (HPLC): 99.6% (tR = 24.4 min). Specific rotation: [α]20D = 130.1 (c = 0.229 g/100 mL, CH2Cl2).
3-Benzyloxy-17-(methoxymethylen)estra-1,3,5(10)-triene (9)
Under N2, KOtBu (0.467 g, 4.16 mmol, 1.5 equiv) was added to a −20 °C stirred suspension of (methoxymethyl)triphenylphosphonium chloride (1.43 g, 4.16 mmol, 1.5 equiv) in dry THF (3.3 mL). After 30 min, ketone 8 (1.00 g, 2.77 mmol, 1.0 equiv) was added, and after stirring for 1 h at −20 °C, the cooling bath was removed. After 3 h, the mixture was cooled to −20 °C again and a suspension of (methoxymethyl)triphenylphosphonium chloride (0.475 g, 1.39 mmol, 0.5 equiv) and KOtBu (0.156 g, 1.39 mmol, 0.5 equiv) in THF (1.1 mL), which had been prepared separately and stirred at −20 °C for 30 min, was added. After 30 min, the cooling bath was removed and the transformation was terminated after another 2.5 h by addition of H2O (2 mL). The mixture was transferred to a separation funnel and diluted with H2O (25 mL), and the mixture was extracted with cHex (3 × 25 mL). The organic phases were combined and dried (Na2SO4), and the solvent was evaporated in vacuo. The residue was purified by flash chromatography (Ø = 4.5 cm, l = 16 cm, V = 30 mL, eluent: cHex/EtOAc 99:1). Colorless resin, yield 1.08 g (99%). Rf (cHex/EtOAc 9:1) = 0.34. C27H32O2 (374.5 g/mol). 1H NMR (600 MHz, CDCl3): δ (ppm) = 0.84 (s, 1.2H, CH3), 0.91 (s, 1.8H, CH3), 1.20–1.69 (m, 6H), 1.73–1.98 (m, 2.4H), 2.14–2.40 (m, 4H), 2.45–2.52 (m, 0.6H), 2.79–2.94 (m, 2H), 3.49 (s, 1.8H, OCH3), 3.58 (s, 1.2H, OCH3), 5.04 (s, 2H, PhCH2O), 5.71–5.76 (m, 1H, C=CHOCH3), 6.70–6.74 (m, 1H, 4-H), 6.78 (dd, J = 8.5/2.8 Hz, 1H, 2-H), 7.22 (dt, J = 8.9/1.9 Hz, 1H, 1-H), 7.28–7.35 (m, 1H, 4-Hbenzyl), 7.35–7.41 (m, 2H, 3-Hbenzyl, 5-Hbenzyl), 7.41–7.45 (m, 2H, 2-Hbenzyl, 6-Hbenzyl). The ratio of (E)/(Z)-configured diastereomers is 60:40. Exact mass (APCI): (m/z) = 389.2478 (calcd. 389.2475 for C27H33O2 [M + H+]). IR (neat): ν̃ [cm–1] = 2924 (C–Haliph), 2874 (C–Haliph), 1605 (C=Carom). Purity: >95%, estimated by 1H NMR spectroscopy.
3-Benzyloxyestra-1,3,5(10)-triene-17β-carbaldehyde (10)
Enol ether 9 (1.07 g, 2.75 mmol) was dissolved in THF (60 mL) and 5% aqueous HCl (20 mL). After heating to reflux for 1 h followed by cooling to rt, the solution was extracted with CHCl3 (3 × 25 mL). The organic layers were combined, dried (Na2SO4), and concentrated in vacuo. The residue was purified by flash chromatography (Ø = 4.5 cm, l = 19 cm, V = 30 mL, eluent: cHex/EtOAc 9:1), and the product was recrystallized from EtOH (abs.). Colorless solid, mp 124–125 °C, yield 0.497 g (48%). C26H30O2 (374.5 g/mol). Rf (cHex/EtOAc 7:3) = 0.49. 1H NMR (600 MHz, CDCl3): δ (ppm) = 0.80 (s, 3H, CH3), 1.32–1.48 (m, 4H, 7-CH2 (1H), 8-CH (1H), 12-CH2 (1H), 14-CH (1H)), 1.52 (tdd, J = 13.3/11.9/3.8 Hz, 1H, 11-CH2), 1.65 (td, J = 12.9/4.0 Hz, 1H, 15-CH2), 1.75–1.94 (m, 3H, 7-CH2 (1H), 12-CH2 (1H), 16-CH2 (1H)), 2.12–2.21 (m, 2H, 15-CH2 (1H), 16-CH2 (1H)), 2.27 (ddd, J = 14.2/10.1/4.3 Hz, 1H, 9-CH), 2.34 (dtd, J = 13.5/4.2/2.8 Hz, 1H, 11-CH2), 2.39 (td, J = 9.2/2.2 Hz, 1H, 17-CH), 2.81–2.93 (m, 2H, 6-CH2), 5.04 (s, 2H, PhCH2O), 6.73 (d, J = 2.8 Hz, 1H, 4-CH), 6.79 (dd, J = 8.6/2.8 Hz, 1H, 2-CH), 7.20 (d, J = 8.7 Hz, 1H, 1-CH), 7.29–7.35 (m, 1H, 4-Hbenzyl), 7.35–7.41 (m, 2H, 3-Hbenzyl, 5-Hbenzyl), 7.41–7.45 (m, 2H, 2-Hbenzyl, 6-Hbenzyl), 9.82 (d, J = 2.1 Hz, 1H, CH=O). 13C NMR (151 MHz, CDCl3): δ (ppm) = 14.1 (1C, C-18), 21.3 (1C, C-16), 24.7 (1C, C-12), 26.3 (1C, C-11), 27.9 (1C, C-7), 29.9 (1C, C-6), 38.4 (1C, C-8), 38.6 (1C, C-15), 44.0 (1C, C-9), 45.2 (1C, C-13), 55.4 (1C, C-14), 63.1 (1C, C-17), 70.1 (1C, PhCH2O), 112.5 (1C, C-2), 115.0 (1C, C-4), 126.4 (1C, C-1), 127.6 (2C, C-2benzyl, C-6benzyl), 128.0 (1C, C-4benzyl), 128.7 (2C, C-3benzyl, C-5benzyl), 132.8 (1C, C-10), 137.4 (1C, C-1benzyl), 138.1 (1C, C-5), 157.0 (1C, C-3), 205.0 (1C, CH=O). Exact mass (APCI): (m/z) = 375.2353, calcd. 375.2319 for C26H31O2 [M + H+]). IR (neat): ν̃ [cm–1] = 2924 (C–Haliph), 2874 (C–Haliph), 2712 (C–Haldehyde), 1717 (C=Oaldehyde), 1605 (C=Carom). Purity (HPLC): 98.7% (tR = 25.8 min). Specific rotation: [α]20D = 90.0 (c = 0.183 g/100 mL, CH2Cl2).
N1-[(3-Benzyloxyestra-1,3,5(10)-trien-17-yl)methyl]-N2,N2-dimethylethane-1,2-diamine (11d)
N,N-Dimethylethane-1,2-diamine (58.3 μL, 47.1 mg, 534 μmol, 2.0 equiv), HOAc (36.3 μL, 38.5 mg, 641 μmol), NaBH(OAc)3 (80%, 212 mg, 801 μmol, 3 equiv), and molecular sieves 4 Å (∼0.5 g) were added to a solution of aldehyde 10 (100 mg, 267 μmol, 1.0 equiv) in dry THF (5.0 mL). The mixture was stirred at rt for 2 d, filtered through a Celite plug, and rinsed thoroughly with EtOAc. The collected filtrate was washed once with 1 M NaOH (15 mL), dried (Na2SO4), and concentrated in vacuo. The residue was purified by flash chromatography (l = 20 cm, Ø = 2 cm, V = 12 mL, eluent CH2Cl2/MeOH/7 M NH3 in MeOH = 90:8:2). Colorless solid, mp 85–88 °C, yield 94 mg (77%). Rf (CH2Cl2/MeOH 9:1 + 1 drop conc. NH3) = 0.51. C30H42N2O (446.7 g/mol). 1H NMR (400 MHz, CDCl3): δ (ppm) = 0.65 (s, 3H, CH3), 1.17–1.56 (m, 7H, 7-CH2 (1H), 8-CH, 11-CH2 (1H), 12-CH2 (1H), 14-CH, 15-CH2 (1H), 16-CH2 (1H)), 1.61–1.73 (m, 1H, 17-CH), 1.73–1.81 (m, 1H, 15-CH2), 1.82–2.04 (m, 3H, 7-CH2 (1H), 12-CH2 (1H), 16-CH2 (1H)), 2.10–2.35 (m, 2H, 9-CH, 12-CH2 (1H), 2.24 (s, 6H, N(CH3)2), 2.41–2.53 (m, 3H, CHCH2NH (1H), CH2N(CH3)2), 2.68–2.93 (m, 5H, 6-CH2, CHCH2NH (1H), NCH2CH2,), 5.03 (s, 2H, PhCH2O), 6.71 (d, J = 2.5 Hz, 1H, 4-CH), 6.77 (dd, J = 8.6/2.8 Hz, 1H, 2-CH), 7.20 (dd, J = 8.3/0.8 Hz, 1H, 1-CH), 7.28–7.34 (m, 1H, 4-CHbenzyl), 7.34–7.40 (m, 2H, 3-CHbenzyl, 5-CHbenzyl), 7.40–7.45 (m, 2H, 2-CHbenzyl, 6-CHbenzyl). A signal for the NH-proton is not seen in the spectrum. 13C NMR (101 MHz, CDCl3): δ (ppm) = 12.8 (1C, CH3), 24.6 (1C, C-15), 26.6 (1C, C-11), 27.7 (1C, C-16), 28.0 (1C, C-7), 30.0 (1C, C-6), 38.4 (1C, C-12), 38.7 (1C, C-8), 42.4 (1C, C-13), 44.2 (1C, C-9), 45.7 (2C, N(CH3)2), 48.0 (1C, NHCH2CH2NMe2), 50.7 (1C, C-17), 51.8 (1C, NHCH2CH), 55.0 (1C, C-14), 59.0 (1C, CH2N(CH3)2), 70.1 (1C, PhCH2O), 112.4 (1C, C-2), 115.0 (1C, C-4), 126.4 (1C, C-1), 127.6 (1C, C-4benzyl), 128.0 (2C, C-2benzyl, C-6benzyl), 128.7 (1C, C-3benzyl, C-5benzyl), 133.4 (1C, C-10), 137.5 (1C, C-1benzyl), 138.3 (1C, C-5), 156.8 (1C, C-3). Exact mass (ESI): (m/z) = 447.3395, calcd. 447.3370 for C30H43N2O [M + H+]. IR (neat): ν̃ [cm–1] = 2978 (C–Haliph), 2931 (C–Haliph), 1613 (C=Carom), 1497 (C=Carom). Purity (HPLC): 99.0% (tR = 19.3 min). Specific rotation: [α]20D = +52.6 (c = 0.270 g/100 mL, CH2Cl2).
N1-[(3-Benzyloxyestra-1,3,5(10)-trien-17-yl)methyl]-N2,N2-dimethylpropane-1,3-diamine (11e)
N,N-Dimethylpropane-1,3-diamine (36.7 μL, 30 mg, 293 μmol, 1.1 equiv), HOAc (10 μL, 10.6 mg, 176 μmol), and NaBH(OAc)3 (80%, 113 mg, 427 μmol, 1.6 equiv). were added to a solution of aldehyde 10 (100 mg, 267 μmol, 1.0 equiv) in dry THF (1.0 mL). The mixture was stirred at rt for 24 h. H2O (15 mL) was added, and the mixture was extracted with EtOAc (3 × 10 mL). The organic layers were combined, dried (Na2SO4), filtered, and concentrated in vacuo. The residue was purified by flash chromatography (l = 13 cm, Ø = 1.5 cm, V = 12 mL, eluent CH2Cl2/MeOH/7 M NH3 in MeOH = 90:9:1). Yellow waxy solid, mp 115–126 °C, yield 68 mg (68%). Rf (CH2Cl2/MeOH 9:1 + 1 drop conc. NH3) = 0.21. C31H44N2O (460.7 g/mol). 1H NMR (400 MHz, CDCl3): δ (ppm) = 0.65 (s, 3H, C-18), 1.20–1.53 (m, 7H, 7-CH2 (1H), 8-CH, 11-CH2 (1H), 12-CH2 (1H), 14-CH, 15-CH2 (1H), 16-CH2 (1H)), 1.66–1.82 (m, 4H, 15-CH2 (1H), 17-CH, NCH2CH2CH2N), 1.83–2.01 (m, 3H, 7-CH2 (1H), 12-CH2 (1H), 16-CH2 (1H)), 2.14–2.34 (m, 2H, 9-CH, 11-CH2 (1H)), 2.25 (s, 6H, N(CH3)2 ), 2.37 (t, J = 6.9 Hz, 2H, CH2NMe2), 2.51 (dd, J = 11.3/9.1 Hz, 1H, CHCH2NH), 2.64–2.96 (m, 5H, 6-CH2, CHCH2NH(1H), NHCH2CH2), 5.03 (s, 2H, PhCH2O), 6.71 (d, J = 2.7 Hz, 1H, 4-CH), 6.77 (dd, J = 8.6/2.8 Hz, 1H, 2-CH), 7.19 (d, J = 8.6 Hz, 1H, 1-CH), 7.29–7.33 (m, 1H, 4-CHbenzyl), 7.34–7.40 (m, 2H, 3-CHbenzyl, 5-CHbenzyl), 7.40–7.45 (m, 2H, 2-CHbenzyl, 6-CHbenzyl). A signal for the NH proton is not seen in the spectrum. 13C NMR (101 MHz, CDCl3): δ (ppm) = 12.8 (1C, C-18), 24.5 (1C, C-15), 26.5 (1C, C-11), 27.0 (1C, CH2CH2N(CH3)2, 27.5 (1C, C-16), 27.9 (1C, C-7), 30.0 (1C, C-6), 38.2 (1C, C-12), 38.7 (1C, C-8), 42.4 (1C, C-13), 44.2 (1C, C-9), 45.7 (2C, N(CH3)2, 49.5 (1C, NHCH2CH2), 50.2 (1C, C-17), 51.4 (1C, CHCH2NH), 54.9 (1C, C-14), 58.7 (1C, CH2N(CH3)2), 70.1 (1C, PhCH2O), 112.4 (1C, C-2), 114.9 (1C, C-4), 126.4 (1C, C-1), 127.6 (2C, C-2benzyl, C-6benzyl), 127.9 (1C, C-4benzyl), 128.7 (2C, C-3benzyl, C-5benzyl), 133.3 (1C, C-10), 137.5 (1C, 1-Cbenzyl), 138.2 (1C, C-5), 156.8 (1C, C-3). Exact mass (ESI): (m/z) = 461.3525 (calcd. 461.3526 for C31H45N2O [M + H+]). IR (neat): ν̃ [cm–1] = 2978 (C–Haliph), 2881 (C–Haliph), 1612 (C=Carom), 1574 (C=Carom), 1501 (C=Carom). Purity (HPLC): 98.1% (tR = 19.4 min). Specific rotation: [α]20D = +42.1 (c = 0.240 mg/100 mL, CH2Cl2).
17-{[2-(Dimethylamino)ethylamino]methyl}estra-1,3,5(10)-trien-3-ol (2d)
Pd/C 10% (6 mg, 5.8 μmol, 0.05 eq.) was added to a solution of amine 11d (60 mg, 134 μmol, 1.0 equiv) in THF (10.0 mL) and EtOH (5.0 mL). The mixture was stirred at rt under a H2 atmosphere (balloon) for 20 h. Then, it was filtered through a Celite plug and rinsed thoroughly with EtOAc. After concentrating the filtrate in vacuo, the residue was purified by flash chromatography twice (fc1: l = 16.5 cm, Ø = 1.5 cm, V = 10 mL, eluent CH2Cl2/MeOH/7 M NH3 in MeOH = 99:9:1; fc2: l = 19.5 cm, Ø = 1 cm, V = 3 mL, CH2Cl2/MeOH/7 M NH3 in MeOH = 99:9:1). Yellow waxy solid, mp 41–51 °C, yield 20 mg (43%). Rf (CH2Cl2/MeOH 9:1 + 1 drop conc. NH3) = 0.29. C23H36N2O (356.6 g/mol). 1H NMR (600 MHz, DMSO-d6): δ (ppm) = 0.60 (s, 3H, CH3), 1.12–1.36 (m, 7H, 7-CH2 (1H), 8-CH, 11-CH2 (1H), 12-CH2 (1H), 14-CH, 15-CH2 (1H), 16-CH2 (1H)), 1.50–1.59 (m, 1H, 17-CH), 1.63–1.71 (m, 1H, 15-CH2), 1.74–1.81 (m, 1H, 7-CH2), 1.81–1.88 (m, 1H, 16-CH2), 1.88–1.93 (m, 1H, 12-CH2), 2.07–2.15 (m, 1H, 9-CH), 2.13 (s, 6H, N(CH3)2), 2.18–2.26 (m, 1H, 11-CH2), 2.31 (t, J = 6.3 Hz, 2H, CH2CH2N(CH3)2), 2.36–2.43 (m, 1H, CHCH2NH), 2.59 (t, J = 6.4 Hz, 2H, NHCH2CH2), 2.66 (dd, J = 11.3/5.4 Hz, 1H, CHCH2NH), 2.69–2.77 (m, 2H, 6-CH2), 6.42 (d, J = 2.6 Hz, 1H, 4-CH), 6.50 (dd, J = 8.4/2.7 Hz, 1H, 2-CH), 7.03 (d, J = 8.5 Hz, 1H, 1-CH), 8.97 (s, 1H, OH). A signal for the NH-proton is not seen in the spectrum. 13C NMR (101 MHz, DMSO-d6): δ (ppm) = 12.3 (1C, CH3), 24.0 (1C, C-15), 26.1 (1C, C-11), 27.2 (1C, C-16), 27.5 (1C, C-7), 29.2 (1C, C-6), 37.9 (1C, C-12), 38.4 (1C, C-8), 41.8 (1C, C-13), 43.6 (1C, C-9), 45.3 (2C, N(CH3)2), 47.3 (1C, NHCH2CH2), 50.0 (1C, C-17), 51.0 (1C, CHCH2NH, 54.4 (1C, C-14), 58.4 (1C, CH2N(CH3)2), 112.7 (1C, C-2), 114.9 (1C, C-4), 126.0 (1C, C-1), 130.5 (1C, C-10), 137.1 (1C, C-5), 154.9 (1C, C-3). Exact mass (ESI): (m/z) = 357.2929, calcd. 357.2900 for C23H37N2O [M + H+]. IR (neat): ν̃ [cm–1] = 2978 (C–Haliph), 1608 (C=Carom), 1581 (C=Carom), 1458 (C=Carom). Purity (HPLC): 97.4% (tR = 14.3 min). Specific rotation: [α]20D = +56.7 (c = 0.177 mg/100 mL, MeOH).
17-{[3-(Dimethylamino)propylamino]methyl}estra-1,3,5(10)-trien-3-ol (2e)
Pd/C 10% (13 mg, 5.8 μmol, 0.1 equiv) was added to a solution of amine 11e (51 mg, 111 μmol, 1.0 equiv) in THF (5.0 mL) and EtOH (1.0 mL). The mixture was stirred at rt under a H2 atmosphere (balloon) for 11 h. Then, it was filtered through a Celite plug twice and rinsed thoroughly with EtOAc. After concentrating the filtrate in vacuo, the residue was purified by flash chromatography twice (fc1: l = 15 cm, Ø = 1.5 cm, V = 10 mL, eluent CH2Cl2/MeOH/7 M NH3 in the MeOH gradient from 95:4:1 to 80:15:1). Colorless solid, mp 122–127 °C, yield 22 mg (53%). Rf (CH2Cl2/MeOH 9:1 + 1 drop conc. NH3) = 0.13. C24H38N2O (370.6 g/mol). 1H NMR (600 MHz, CDCl3): δ (ppm) = 0.62 (s, 3H, C-18H3), 1.14–1.51 (m, 7H, 7-CH2 (1H), 8-CH, 11-CH2 (1H), 12-CH2 (1H), 14-CH, 15-CH2 (1H), 16-CH2 (1H)), 1.59–1.78 (m, 4H, 15-CH2 (1H), 17-CH, NHCH2CH2), 1.79–2.00 (m, 3H, 7-CH2 (1H), 12-CH2 (1H), 16-CH2 (1H)), 2.09–2.31 (m, 2H, 9-CH, 12-CH2 (1H)), 2.25 (s, 6H, N(CH3)2), 2.38 (t, J = 7.2 Hz, 2H, CH2N(CH3)2), 2.46 (dd, J = 11.4/8.9 Hz, 1H, CHCH2NH), 2.61–2.89 (m, 5H, 6-CH2, CHCH2NH (1H), NHCH2CH2), 6.52 (d, J = 2.6 Hz, 1H, 4-CH), 6.58 (dd, J = 8.4/2.8 Hz, 1H, 2-CH), 7.10 (d, J = 8.3 Hz, 1H, 1-CH). Signals for the NH proton and the phenol proton are not seen in the spectrum. 13C NMR (101 MHz, DMSO-d6): δ (ppm) = 12.8 (1C, C-18), 24.5 (1C, C-15), 26.6 (1C, C-11), 27.0 (1C, CH2CH2N(CH3)2), 27.5 (1C, C-16), 28.0 (1C, C-7), 29.9 (1C, C-6), 38.3 (1C, C-12), 38.8 (1C, C-8), 42.4 (1C, C-13), 44.1 (1C, C-9), 45.5 (2C, N(CH3)2, 49.1 (1C, NHCH2CH2), 50.3 (1C, C-17), 51.3 (1C, CHCH2NH), 54.9 (1C, C-14), 58.3 (1C, CH2N(CH3)2, 113.1 (1C, C-2), 115.8 (1C, C-4), 126.4 (1C, C-1), 131.7 (1C, C-10), 138.1 (1C, C-5), 154.9 (1C, C-3). Exact mass (ESI): (m/z) = 371.3083, calcd. 371.3057 for C24H39N2O [M + H+]. IR (neat): ν̃ [cm–1] = 2978 (C–Haliph), 1612 (C=Carom), 1582 (C=Carom), 1462 (C=Carom). Purity (HPLC): 95.3% (tR = 14.2 min). Specific rotation: [α]20D = +54.2 (c = 0.205 g/100 mL, MeOH).
X-ray Diffraction Measurement
Data sets for compound 10 were collected with a Bruker D8 Venture Photon III diffractometer. Programs used: data collection: APEX3 V2019.1–0,36 cell refinement: SAINT V8.40A;37 data reduction: SAINT V8.40A;37 absorption correction, SADABS V2016/2;38 structure solution SHELXT-2015;39 structure refinement SHELXL-2015(40) and graphics XP.(41)R-values are given for observed reflections, and wR2 values are given for all reflections.
X-ray crystal structure analysis of 10: A colorless needle-like specimen of C26H30O2, approximate dimensions 0.040 mm × 0.082 mm × 0.272 mm, was used for the X-ray crystallographic analysis. The X-ray intensity data were measured using a Bruker D8 Venture Bruker D8 Venture Photon III diffractometer system equipped with a micro focus tube CuKα (CuKα, λ = 1.54178 Å) and a MX mirror monochromator. A total of 1810 frames were collected. The total exposure time was 23.88 h. The frames were integrated with the Bruker SAINT software package using a wide-frame algorithm. The integration of the data using a triclinic unit cell yielded a total of 13,467 reflections to a maximum θ angle of 66.66° (0.84 Å resolution), of which 3461 were independent (average redundancy 3.891, completeness = 99.7%, Rint = 7.48%, and Rsig = 5.95%) and 2882 (83.27%) were greater than 2σ(F2). The final cell constants of a = 6.4778(2) Å, b = 8.1459(2) Å, c = 10.1467(3) Å, α = 71.796(2)°, β = 86.666(2)°, γ = 82.824(2)°, and volume = 504.53(3) Å3 are based upon the refinement of the XYZ-centroids of 5436 reflections above 20 σ(I) with 9.176° < 2θ < 132.7°. Data were corrected for absorption effects using the Multi-Scan method (SADABS). The ratio of minimum to maximum apparent transmission was 0.877. The calculated minimum and maximum transmission coefficients (based on the crystal size) are 0.8570 and 0.9770, respectively. The structure was solved and refined using the Bruker SHELXTL software package, using the space group P1, with Z = 1 for the formula unit, C26H30O2. The final anisotropic full-matrix least-squares refinement on F2 with 254 variables converged at R1 = 4.87% for the observed data and wR2 = 12.44% for all data. The goodness-of-fit was 1.071. The largest peak in the final difference electron density synthesis was 0.145 e–/Å3, and the largest hole was −0.176 e–/Å3 with an RMS deviation of 0.039 e–/Å3. On the basis of the final model, the calculated density was 1.233 g/cm3 and F(000), 202 e–.
CCDC-2181677 (compound 10) contains the supplementary crystallographic data for this paper. These data can be obtained free of charge from The Cambridge Crystallographic Data Centre via www.ccdc.cam.ac.uk/data_request/cif.
Functional Analysis of Drug Actions
Isolation of Motile Sperm
Experiments on human sperm were conducted in agreement with the standards set by the Declaration of Helsinki. Healthy volunteers provided human ejaculate samples with prior written consent and under approval of the institutional ethics committees of the medical association Westfalen-Lippe and the Medical Faculty of the University of Münster (4INie). Motile sperm used for Ca2+ fluorimetric experiments were purified by the swim-up procedure as described previously.18,24 In brief, in 50 mL falcon tubes angled at 45°, 4 mL of human tubular fluid (HTF) medium, containing (in mM) 93.8 NaCl, 4.69 KCl, 0.2 MgSO4, 0.37 KH2PO4, 2.04 CaCl2, 0.33 Na-pyruvate, 21.4 lactic acid, 2.78 glucose, 21 HEPES, and 4 NaHCO3 (pH adjusted to 7.35 with NaOH) was underlaid with 0.7 to 1.2 mL of ejaculate. After incubation for 1 h at 37 °C, supernatants were collected, and sperm were washed twice with HTF by centrifugation at 700 × g for 20 min. After the second centrifugation, sperm were resuspended to 107 cells·mL–1 in HTF containing 3 mg mL–1 human serum albumin (HSA, Irvine Scientific). Before experiments, sperm were incubated at 37 °C.
Measurement of Changes of the Intracellular Ca2+ Concentration
Changes of [Ca2+]i in motile human sperm were measured in population using a plate reader (Fluostar Omega, BMG Labtech) with 384-well plates and fluorescence readout as described previously.18,24 Sperm were loaded with the fluorescent Ca2+ indicator Fluo4 by incubation with the prodrug ester Fluo4-AM (5 μM, ThermoFisher) in the presence of Pluronic F-127 (0.05% w/v) for 20 min. Centrifugation at 700 × g for 5 min and resuspension to 5 × 106 cells·mL–1 in HTF removed excess dye. For the experiments, wells were filled with 50 μL of the dye-loaded sperm suspension. Fluorescence was excited at 480 nm, and emission was recorded at 520 nm. After recording a baseline, 25 μL of inhibitor solutions was added and fluorescence response recorded. After 4 min, 7.5 μL of stimulus (progesterone or PGE1; 2 μM final) was added and ensuing changes in fluorescence were recorded. Solutions were added using an electric multichannel Eppendorf pipette. Changes in Fluo-4 fluorescence, i.e., [Ca2+]i, are depicted as ΔF/F (%), i.e., the change in fluorescence (ΔF) relative to the mean basal fluorescence (F) before application of buffer or compounds/ligands to correct for intra- and inter-experimental variations in basal fluorescence among individual wells or to correct for compound-evoked changes in fluorescence (see below). For examination of compound-evoked Ca2+ signal, basal fluorescence (F) was determined before addition of compounds. For determination of inhibitory effects of compounds on ligand-evoked Ca2+ signals, basal fluorescence (F) was determined before addition of the respective ligand. IC50 values were calculated by determining the maximal signal amplitudes evoked by the ligand in the absence and presence of various compound concentrations. For some analyses, ΔF/F was normalized to the maximal signal amplitude evoked in the absence of the compound to ease comparisons between experiments. Data from fluorescence recordings were analyzed in GraphPad Prism version 9.4.0.
Electrophysiological Recordings of Slo3 Currents
CHO cells were co-transfected with a pcDNA3.1(+) vector containing the full length coding sequence of human Slo3 modified with a carboxy-terminal hemagglutinin tag (HA-tag) and in which the sequence coding for the neomycin resistance gene was replaced by the coding sequence for citrine and a pcDNA3.1(+) vector containing a sequence encoding hLRRC52-mCherry.32 Cells were co-transfected with 2–5 ng/μL of the respective vector for 6–8 h using Lipofectamine 2000 in Opti-MEM (Gibco). Afterward, cells were cultivated in the F-12 medium; after another 24 h, cells were transferred onto poly-l-lysine-coated cover slides and incubated in 5 mM Na-butyrate at least 12 h before the experiment.
Electrophysiological recordings were performed in the whole-cell configuration using an Axopatch 200B patch clamp amplifier (Molecular Devices, Sunnyvale, CA, USA) controlled by Clampex 10.7 (Molecular Devices). Signals were low-pass filtered at 10 kHz with a four-pole Bessel filter and digitized with a Digidata 1440A data acquisition system (Molecular Devices). Series resistance and cell capacitance were compensated. A step protocol with steps from −100 to +150 mV followed by a step to 50 mV from a holding potential of −80 mV was used. The intracellular solution contained 130 mM K-aspartate, 10 mM NaCl, 1 mM EGTA, 5 mM HEPES, and 15 mM glucose, pH 7.3 with KOH, and the pipette resistance was 4–6 MΩ. Cells were perfused with extracellular solution (140 mM NaCl, 5.4 mM KCl, 1 mM MgCl2,1.8 mM CaCl2, 5 mM HEPES, and 10 mM glucose, pH 7.4 with NaOH). All substances were diluted in extracellular solution and applied to the cells using a gravity-driven perfusion system. Experiments were performed at rt.
Flow-Cytometry Analysis of Cell Viability
Sperm were incubated in parallel for 20 min at 37 ° C in HTF in the absence (control) and presence of the respective compound at a concentration of 60 μM. For the last 10 min, propidium iodide (PI) was added at a concentration of 3 μM. The fluorescence of PI in individual sperm was determined by an excitation wavelength of 488 nm and a detection filter of 690/50 BP at a gain of 250 in a CytoFlexS flow cytometer (BD Biosciences). At least 10,000 events were analyzed per condition, and the fraction of viable sperm was calculated based on the intensity of the PI fluorescence.
Acknowledgments
This work was supported by the Research Training Group “Chemical biology of ion channels (Chembion)” (T.S., T.S., and B.W.) and the clinical research unit CRU236 (T.S. and C.B.) funded by the German Research Foundation (DFG) as well as the Center for Clinical Research (IZKF; Str/014/21 to T.S.) and the MedK program (C.E.) of the Medical Faculty of the University of Münster.
Supporting Information Available
The Supporting Information is available free of charge at https://pubs.acs.org/doi/10.1021/acsptsci.2c00188.
(Figures S1 and S2) Action of compounds 2a–e on the viability of human sperm and on PGE1-evoked Ca2+ signals in human sperm and HPLC chromatograms and 1H and 13C NMR spectra (PDF)
Author Contributions
T.S. and B.W. contributed equally to this work. All persons designated as authors qualify for authorship, and all those who qualify for authorship are listed. B.W. and T.S. conceived and designed the study and coordinated the experiments. T.S., B.W., and T.S. wrote the manuscript. T.S., B.T., C.E., S.R., C.G.D., and C.B. acquired, analyzed, and/or interpreted data and revised the manuscript critically for important intellectual content. All authors approved the manuscript.
The authors declare no competing financial interest.
Supplementary Material
References
- Alvarez L.; Friedrich B. M.; Gompper G.; Kaupp U. B. The computational sperm cell. Trends Cell Biol. 2014, 24, 198–207. 10.1016/j.tcb.2013.10.004. [DOI] [PubMed] [Google Scholar]
- Mata-Martínez E.; Sánchez-Cárdenas C.; Chávez J. C.; Guerrero A.; Treviño C. L.; Corkidi G.; Montoya F.; Hernandez-Herrera P.; Buffone M. G.; Balestrini P. A.; Darszon A. Role of calcium oscillations in sperm physiology. Biosystems 2021, 209, 104524 10.1016/j.biosystems.2021.104524. [DOI] [PubMed] [Google Scholar]
- Publicover S. J.; Giojalas L. C.; Teves M. E.; de Oliveira G. S. M. M.; Garcia A. A. M.; Barratt C. L. R.; Harper C. V. Ca2+ signalling in the control of motility and guidance in mammalian sperm. Front. Biosci. 2008, 13, 5623–5637. 10.2741/3105. [DOI] [PubMed] [Google Scholar]
- Wang H. M. G.; McGoldrick L. L.; Chung J.-J. Sperm ion channels and transporters in male fertility and infertility. Nat. Rev. Urol. 2021, 18, 46–66. 10.1038/s41585-020-00390-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kaupp U. B.; Strünker T. Signaling in Sperm: More Different than Similar. Trends Cell Biol. 2017, 27, 101–109. 10.1016/j.tcb.2016.10.002. [DOI] [PubMed] [Google Scholar]
- Saggiorato G.; Alvarez L.; Jikeli J. F.; Kaupp U. B.; Gompper G.; Elgeti J. Human sperm steer with second harmonics of the flagellar beat. Nat. Commun. 2017, 8, 1415. 10.1038/s41467-017-01462-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Brown S. G.; Publicover S. J.; Barratt C. L. R.; Da Martins Silva S. J. Human sperm ion channel (dys)function: implications for fertilization. Hum. Reprod. Update 2019, 25, 758–776. 10.1093/humupd/dmz032. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Rahban R.; Nef S. CatSper: The complex main gate of calcium entry in mammalian spermatozoa. Mol. Cell. Endocrinol. 2020, 518, 110951 10.1016/j.mce.2020.110951. [DOI] [PubMed] [Google Scholar]
- Kirichok Y.; Navarro B.; Clapham D. E. Whole-cell patch-clamp measurements of spermatozoa reveal an alkaline-activated Ca2+ channel. Nature 2006, 439, 737–740. 10.1038/nature04417. [DOI] [PubMed] [Google Scholar]
- Lishko P. V.; Kirichok Y. The role of Hv1 and CatSper channels in sperm activation. J. Physiol. 2010, 588, 4667–4672. 10.1113/jphysiol.2010.194142. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Loyo-Celis V.; Orta G.; Beltrán C.; Darszon A. CatSper channels in sea urchin sperm. Cell Calcium 2021, 99, 102466 10.1016/j.ceca.2021.102466. [DOI] [PubMed] [Google Scholar]
- Seifert R.; Flick M.; Bönigk W.; Alvarez L.; Trötschel C.; Poetsch A.; Müller A.; Goodwin N.; Pelzer P.; Kashikar N. D.; Kremmer E.; Jikeli J.; Timmermann B.; Kuhl H.; Fridman D.; Windler F.; Kaupp U. B.; Strünker T. The CatSper channel controls chemosensation in sea urchin sperm. EMBO J. 2015, 34, 379–392. 10.15252/embj.201489376. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Brenker C.; Schiffer C.; Wagner I. V.; Tüttelmann F.; Röpke A.; Rennhack A.; Kaupp U. B.; Strünker T. Action of steroids and plant triterpenoids on CatSper Ca2+ channels in human sperm. Proc. Natl. Acad. Sci. U. S. A. 2018, 115, E344–E346. 10.1073/pnas.1717929115. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Brown S. G.; Costello S.; Kelly M. C.; Ramalingam M.; Drew E.; Publicover S. J.; Barratt C. L. R.; Da Silva S. M. Complex CatSper-dependent and independent Ca2+i signalling in human spermatozoa induced by follicular fluid. Hum. Reprod. 2017, 32, 1995–2006. 10.1093/humrep/dex269. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Jeschke J. K.; Biagioni C.; Schierling T.; Wagner I. V.; Börgel F.; Schepmann D.; Schüring A.; Kulle A. E.; Holterhus P. M.; von Wolff M.; Wünsch B.; Nordhoff V.; Strünker T.; Brenker C. The Action of Reproductive Fluids and Contained Steroids, Prostaglandins, and Zn2+ on CatSper Ca2+ Channels in Human Sperm. Front. Cell Dev. Biol. 2021, 9, 699554 10.3389/fcell.2021.699554. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lishko P. V.; Botchkina I. L.; Kirichok Y. Progesterone activates the principal Ca2+ channel of human sperm. Nature 2011, 471, 387–391. 10.1038/nature09767. [DOI] [PubMed] [Google Scholar]
- Rehfeld A. Revisiting the action of steroids and triterpenoids on the human sperm Ca2+ channel CatSper. Mol. Hum. Reprod. 2020, 26, 816–824. 10.1093/molehr/gaaa062. [DOI] [PubMed] [Google Scholar]
- Strünker T.; Goodwin N.; Brenker C.; Kashikar N. D.; Weyand I.; Seifert R.; Kaupp U. B. The CatSper channel mediates progesterone-induced Ca2+ influx in human sperm. Nature 2011, 471, 382–386. 10.1038/nature09769. [DOI] [PubMed] [Google Scholar]
- Luo T.; Chen H.-Y.; Zou Q.-X.; Wang T.; Cheng Y.-M.; Wang H.-F.; Wang F.; Jin Z.-L.; Chen Y.; Weng S.-Q.; Zeng X.-H. A novel copy number variation in CATSPER2 causes idiopathic male infertility with normal semen parameters. Hum. Reprod. 2019, 34, 414–423. 10.1093/humrep/dey377. [DOI] [PubMed] [Google Scholar]
- Ren D.; Navarro B.; Perez G.; Jackson A. C.; Hsu S.; Shi Q.; Tilly J. L.; Clapham D. E. A sperm ion channel required for sperm motility and male fertility. Nature 2001, 413, 603–609. 10.1038/35098027. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Williams H. L.; Mansell S.; Alasmari W.; Brown S. G.; Wilson S. M.; Sutton K. A.; Miller M. R.; Lishko P. V.; Barratt C. L. R.; Publicover S. J.; Da Martins Silva S. Specific loss of CatSper function is sufficient to compromise fertilizing capacity of human spermatozoa. Hum. Reprod. 2015, 30, 2737–2746. 10.1093/humrep/dev243. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Brenker C.; Goodwin N.; Weyand I.; Kashikar N. D.; Naruse M.; Krähling M.; Müller A.; Kaupp U. B.; Strünker T. The CatSper channel: a polymodal chemosensor in human sperm. EMBO J. 2012, 31, 1654–1665. 10.1038/emboj.2012.30. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Carlson A. E.; Burnett L. A.; del Camino D.; Quill T. A.; Hille B.; Chong J. A.; Moran M. M.; Babcock D. F. Pharmacological targeting of native CatSper channels reveals a required role in maintenance of sperm hyperactivation. PLoS One 2009, 4, e6844 10.1371/journal.pone.0006844. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Rennhack A.; Schiffer C.; Brenker C.; Fridman D.; Nitao E. T.; Cheng Y.-M.; Tamburrino L.; Balbach M.; Stölting G.; Berger T. K.; Kierzek M.; Alvarez L.; Wachten D.; Zeng X.-H.; Baldi E.; Publicover S. J.; Kaupp U. B.; Strünker T. A novel cross-species inhibitor to study the function of CatSper Ca2+ channels in sperm. Br. J. Pharmacol. 2018, 175, 3144–3161. 10.1111/bph.14355.25. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bonfils A.; Humbert J.; Philibert D. RU 3117 a steroidal compound with high affinity for sigma sites in rat testis membranes. J. Steroid Biochem. Mol. Biol. 1996, 59, 49–54. 10.1016/S0960-0760(96)00086-6. [DOI] [PubMed] [Google Scholar]
- Schaefer M.; Habenicht U. F.; Bräutigam M.; Gudermann T. Steroidal sigma receptor ligands affect signaling pathways in human spermatozoa. Biol. Reprod. 2000, 63, 57–63. 10.1095/biolreprod63.1.57. [DOI] [PubMed] [Google Scholar]
- Birch M. R.; Johansen M.; Skakkebæk N. E.; Andersson A.-M.; Rehfeld A. In vitro investigation of endocrine disrupting effects of pesticides on Ca2+–signaling in human sperm cells through actions on the sperm-specific and steroid-activated CatSper Ca2+–channel. Environ. Int. 2022, 167, 107399 10.1016/j.envint.2022.107399. [DOI] [PubMed] [Google Scholar]
- Torrezan-Nitao E.; Brown S. G.; Mata-Martínez E.; Treviño C. L.; Barratt C.; Publicover S. Ca2+i oscillations in human sperm are triggered in the flagellum by membrane potential-sensitive activity of CatSper. Hum. Reprod. 2021, 36, 293–304. 10.1093/humrep/deaa302. [DOI] [PubMed] [Google Scholar]
- Schiffer C.; Müller A.; Egeberg D. L.; Alvarez L.; Brenker C.; Rehfeld A.; Frederiksen H.; Wäschle B.; Kaupp U. B.; Balbach M.; Wachten D.; Skakkebaek N. E.; Almstrup K.; Strünker T. Direct action of endocrine disrupting chemicals on human sperm. EMBO Rep. 2014, 15, 758–765. 10.15252/embr.201438869. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Santi C. M.; Martínez-López P.; de La Vega-Beltrán J. L.; Butler A.; Alisio A.; Darszon A.; Salkoff L. The SLO3 sperm-specific potassium channel plays a vital role in male fertility. FEBS Lett. 2010, 584, 1041–1046. 10.1016/j.febslet.2010.02.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zeng X.-H.; Yang C.; Kim S. T.; Lingle C. J.; Xia X.-M. Deletion of the Slo3 gene abolishes alkalization-activated K+ current in mouse spermatozoa. Proc. Natl. Acad. Sci. U. S. A. 2011, 108, 5879–5884. 10.1073/pnas.1100240108. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Brenker C.; Zhou Y.; Müller A.; Echeverry F. A.; Trötschel C.; Poetsch A.; Xia X.-M.; Bönigk W.; Lingle C. J.; Kaupp U. B.; Strünker T. The Ca2+–activated K+ current of human sperm is mediated by Slo3. Elife 2014, 3, e01438 10.7554/eLife.01438. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lin S.; Ke M.; Zhang Y.; Yan Z.; Wu J. Structure of a mammalian sperm cation channel complex. Nature 2021, 595, 746–750. 10.1038/s41586-021-03742-6. [DOI] [PubMed] [Google Scholar]
- Lees J. A.; Dias J. M.; Han S. Applications of Cryo-EM in small molecule and biologics drug design. Biochem. Soc. Trans. 2021, 49, 2627–2638. 10.1042/BST20210444. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Carlson E. J.; Francis R.; Liu Y.; Li P.; Lyon M.; Santi C. M.; Hook D. J.; Hawkinson J. E.; Georg G. I. Discovery and Characterization of Multiple Classes of Human CatSper Blockers. ChemMedChem 2022, 17, e202000499 10.1002/cmdc.202000499. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bruker AXS Inc. , APEX3; Bruker AXS Inc.: Madison, Wisconsin, USA. [Google Scholar]
- Bruker AXS Inc. , SAINT; Bruker AXS Inc.: Madison, Wisconsin, USA. [Google Scholar]
- Bruker AXS Inc. , SADABS: Bruker AXS area detector scaling and absorption correction; Bruker AXS Inc.: Madison, Wisconsin, USA. [Google Scholar]
- Sheldrick G. M. SHELXT - integrated space-group and crystal-structure determination. Acta Crystallogr. A Found. Adv. 2015, 71, 3–8. 10.1107/S2053273314026370. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sheldrick G. M. Crystal structure refinement with SHELXL. Acta Crystallogr. C Struct. Chem. 2015, 71, 3–8. 10.1107/S2053229614024218. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bruker AXS Inc. , XP: Interactive molecular graphics; Bruker AXS Inc.: Madison, Wisconsin, USA. [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.







