Europe PMC

This website requires cookies, and the limited processing of your personal data in order to function. By using the site you are agreeing to this as outlined in our privacy notice and cookie policy.

Abstract 


Three new (1-3) and two known (4 and 5) cytotoxic cardiac glycosides were isolated and characterized from a medicinal plant, Streblus asper Lour. (Moraceae), collected in Vietnam, with six new analogues and one known derivative (5a-g) synthesized from (+)-strebloside (5). A preliminary structure-activity relationship study indicated that the C-10 formyl and C-5 and C-14 hydroxy groups and C-3 sugar unit play important roles in the mediation of the cytotoxicity of (+)-strebloside (5) against HT-29 human colon cancer cells. When evaluated in NCr nu/nu mice implanted intraperitoneally with hollow fibers facilitated with either MDA-MB-231 human breast or OVCAR3 human ovarian cancer cells, (+)-strebloside (5) showed significant cell growth inhibitory activity in both cases, in the dose range 5-30 mg/kg.

Free full text 


Logo of nihpaAbout Author manuscriptsSubmit a manuscriptHHS Public Access; Author Manuscript; Accepted for publication in peer reviewed journal;
J Nat Prod. Author manuscript; available in PMC 2018 Mar 24.
Published in final edited form as:
PMCID: PMC5365359
NIHMSID: NIHMS836224
PMID: 27983842

Cardiac Glycoside Constituents of Streblus asper with Potential Antineoplastic Activity

Abstract

Three new (1–3) and two known (4 and 5) cytotoxic cardiac glycosides were isolated and characterized from a medicinal plant, Streblus asper, collected in Vietnam, with six new and one known derivatives (5a–5g) synthesized from (+)-strebloside (5). A preliminary structure-activity relationship study indicated that the C-19 formyl and C-5 and C-14 hydroxy groups and C-3 sugar unit play important roles in the mediation of the cytotoxicity of (+)-strebloside (5) against HT-29 human colon cancer cells. When evaluated in NCr nu/nu mice implanted intraperitoneally with hollow fibers facilitated with either MDA-MB-231 human breast or OVCAR3 human ovarian cancer cells, (+)-strebloside (5) showed significant cell growth inhibitory activity in both cases, in the dose range 5–30 mg/kg.

Moraceae (the mulberry or fig family) is a group of flowering plants comprising about 40 genera that have over 1000 species distributed in tropical and subtropical regions, of which the genus Streblus contains about 25 species.1 One of these is Streblus asper Lour., known as the “toothbrush tree”, with various parts having been used in the Ayurvedic medicinal system.2 When Swiss albino mice inoculated by Dalton’s ascitic lymphoma cells were treated with an ethyl acetate extract of the bark of S. asper (i.p., 200 or 400 mg/kg for nine consecutive days), the tumor volume was found to be decreased, with the survival parameters of mice increased.2 Several cardiac glycosides have been characterized from S. asper,36 and (+)-strebloside (5), occurring in high abundance, has been characterized as the main cytotoxic component.5

Many cardiac glycosides have been reported to show potent cytotoxicity toward different human cancer cell lines.7,8 For example, digoxin, isolated originally from Digitalis lanata Ehrh. (Plantaginaceae)9,10 and used traditionally to treat congestive heart failure,11 was found to exhibit anticancer potential.12 It inhibited neuroblastoma growth in vitro and in vivo13 and has emerged as a potential anticancer lead for the treatment of prostate cancer.14 Recently, a phase II clinical trial for the treatment of recurrent prostate cancer has been completed for digoxin.15 Oleandrin, the cardiac glycoside main component of a standardized Nerium oleander plant-based botanical drug, Anvirzel™, has been developed as a potential cancer chemotherapeutic agent.7,16

In a continuing search for anticancer agents from higher plants,17 a chloroform-soluble extract of the stem bark of S. asper collected in Vietnam was found to be highly cytotoxic toward the HT-29 human colon cancer cell line. Fractionation guided by bioassay against this cell line yielded three new (1–3) and two known (4 and 5) cardiac glycosides from this plant. Also, six new analogues and a known derivative were synthesized from (+)-strebloside (5). All compounds were tested for their cytotoxicity toward the HT-29 cell line, and 5 was further evaluated for its growth inhibitory activity against a small panel of human cancer and normal human cell lines, and for its in vivo antitumor efficacy, using a hollow fiber test system.

RESULTS AND DISCUSSION

A sample of the stem bark of S. asper collected in Vietnam was extracted with MeOH, and the extract was partitioned with n-hexane and CHCl3. When the cytotoxic CHCl3 partition was subjected to chromatographic separation guided by inhibitory activity against the HT-29 cell line, three new cardiac glycosides, (+)-19-hydroxykamaloside (1), (+)-5-hydroxyasperoside (2), and (+)-3'-de-O-methylkamaloside (3), and two known analogues, (+)-3-O-β-d-fucopyranosylperiplogenin (4)18 and (+)-strebloside (5)5 were isolated.

Compound 1 was purified as an amorphous colorless powder with a molecular formula of C31H48O10, as shown by a sodiated molecular ion peak at m/z 603.3153 (calcd 603.3140) observed in the HRESIMS, in conjunction with the 1H and 13C NMR spectroscopic data (Tables 1 and and2).2). The UV (λmax 216 nm) and IR (νmax 3459, 1743 and 1621 cm−1) spectra showed the absorptions of hydroxy groups and an unsaturated lactone unit.19,20 The 1H and 13C NMR data of 1 (Tables 1 and and22)21 exhibited the characteristic resonances for a cardiac glycoside, including two methyl groups, a sugar moiety, and a lactone unit.19,20 Comparison of the 13C NMR data of 1 with those of (+)-strebloside (5)5 showed that the resonance for C-19 of 5 at δC 208.3 was replaced by that at δC 65.0 in 1 (Table 2). The resonances for C-5 and C-10 of 5 were different from those for these carbons of 1, but they were closely comparable for other carbons of both compounds. Thus, compound 1 could be defined as 19-nor-10-hydroxymethylstrebloside, an analogue of digoxin.

Table 1

1H NMR Spectroscopic Data of 13 and 5a

position1b2c3d5c
1α 1.20 m
β 2.02 m
α 1.85 m
β 1.80 m
α 1.73 m
β 1.31 m
α 1.24 m
β 2.02 me
21.62e
1.94e
1.64 m
1.80 m
1.75 m
1.91 m
1.96 me
2.02 me
3α 4.25 mα 4.19 mα 4.14 mα 4.22 br s
41.54e
2.05e
α 1.74 m
β 2.07 m
α 1.59 m
β 2.17 m
1.68e
1.96 me
62.02e
1.66e
1.74 m
1.44 me
1.71 m
1.25
2.02 me
1.68e
7α 1.39 m
β 2.59 dt
(14.6, 3.2)
α 1.89 m
β 1.17 m
α 1.98 m
β 1.29
α 1.72 m
β 2.23 m
8β 1.54eβ 1.56eβ 1.52 mβ 1.90 me
9α 1.57eα 1.51eα 1.50 mα 2.06 m
111.51e
1.32 m
1.48 m
1.44 me
α 1.46 m
β 1.33 m
1.51e
121.39e
1.54e
1.51e
1.56e
1.67e
1.81 m
1.38 m
1.51e
152.02e
1.71 m
α 2.03 m
β 1.74 m
α 2.14 m
β 1.77 m
1.68e
1.96 me
16α 2.19 m
β 1.87 m
α 2.18 m
β 1.89 m
α 2.11 m
β 1.93 m
2.16 m
1.90 me
17α 2.79 dd
(9.4, 5.4)
α 2.81 dd
(9.2, 4.6)
α 2.85eα 2.77 dd
(8.8, 5.2)
18β 0.86 sβ 0.88 sβ 0.91 sβ 0.86 s
194.33e
3.45 t (10.1)
β 0.96 sβ 0.92 sβ 10.02 s
215.00 dd
(18.1, 1.4)
4.82 dd
(18.1, 1.6)
5.00 d (17.2)
4.83 d (17.7)
5.03 d (17.9)
4.87 d (18.2)
4.81 d (18.0)
4.96 d (18.0)
225.88 br s5.89 br s5.87 br s5.87 br s
1'α 4.33e
α 4.38 d (7.1)f
α 4.46 d (8.0)α 4.73 d (7.8)α 4.34 d (6.4)
2'β 3.26e
β 3.21 mf
β 3.03 t (9.0)β 3.02 dd
(7.8, 2.7)
β 3.23e
3'α 3.25e
α 3.23 mf
α 3.19 t (9.2)α 4.26 mα 3.21e
4'α 3.82 m
α 3.83 mf
β 3.51 mα 3.14 mα 3.81 br s
5'α 3.54e
α 3.63 mf
α 3.36 mα 3.68 dd
(9.2, 6.3)
α 3.51e
6'β 1.35 d (6.5)
β 1.26 d (6.5)f
3.79 m
3.88 m
β 1.19 d (6.2)β 1.35 d (6.4)
OH-5β 4.76 br sβ 3.49 mβ 3.98 mβ 4.37 br s
OH-194.50 br d (9.1)
OH-4'β 2.39 br sα 2.48 m
OH-6'β 1.95 m
OCH3-2'α 3.56 sα 3.61 sα 3.38 sα 3.55 s
OCH3-3'β 3.49 sβ 3.64 sβ 3.48 s
aAssignments of chemical shifts are based on the analysis of 1D- and 2D-NMR spectra. The overlapped signals were assigned from 1H–1H COSY, HSQC, and HMBC spectra without designating multiplicity.
bData (δ) measured in CDCl3 at 400.13 MHz and referenced to the internal standard TMS residual peak at δ 0.00.
cData (δ) measured in CDCl3 at 400.13 MHz and referenced to the solvent residual peak at δ 7.26.21
dData (δ) measured in acetone-d6 at 400.13 MHz and referenced to the internal standard TMS residual peak at δ 0.00.
eThe overlapped signals.
fData (δ) measured in acetone-d6 at 400.13 MHz and referenced to the solvent residual peak at δ 2.05.21

Table 2

13C NMR Spectroscopic Data of 13 and 5a

position1b2b3c5d
124.0 CH226.1 CH226.4 CH224.3 CH2
225.6 CH225.3 CH226.8 CH225.3 CH2
373.2 CH75.6 CH75.2 CH72.9 CH
434.9 CH234.2 CH234.7 CH234.3 CH2
577.1 C73.0 C73.2 C73.2 C
633.8 CH234.5 CH236.2 CH236.6 CH2
719.0 CH223.8 CH224.6 CH218.1 CH2
840.4 CH40.9 CH41.5 CH41.8 CH
939.1 CH39.2 CH39.6 CH39.5 CH
1042.4 C40.8 C41.6 C54.7 C
1121.1 CH221.5 CH222.4 CH222.1 CH2
1240.1 CH240.1 CH240.5 CH240.0 CH2
1349.4 C49.4 C50.3 C49.6 C
1485.4 C85.6 C85.5 C85.4 C
1532.5 CH233.1 CH233.5 CH232.1 CH2
1626.8 CH226.8 CH227.5 CH227.0 CH2
1750.6 CH50.7 CH51.6 CH50.6 CH
1815.7 CH315.7 CH316.1 CH315.7 CH3
1965.0 CH216.8 CH317.2 CH3208.3 CH
20174.4 C174.4 C176.3 C174.3 C
2173.5 CH273.4 CH273.9 CH273.6 CH2
22117.8 CH117.9 CH117.8 CH118.0 CH
23174.5 C174.2 C176.4 C174.5 C
1'99.4 CH101.0 CH98.1 CH100.0 CH
2'80.0 CH83.8 CH81.6 CH80.1 CH
3'83.9 CH86.2 CH68.6 CH84.0 CH
4'68.4 CH70.7 CH73.8 CH68.5 CH
5'70.5 CH75.1 CH70.4 CH70.5 CH
6'16.4 CH362.8 CH218.3 CH316.5 CH3
OCH3-2'61.2 CH360.9 CH357.1 CH361.4 CH3
OCH3-3'57.6CH361.0 CH357.8 CH3
aAssignments of chemical shifts are based on the analysis of 1D- and 2D-NMR spectra. CH3, CH2, CH, and C multiplicities were determined by DEPT 90, DEPT 135, and HSQC experiments.
bData (δ) measured in CDCl3 at 100.61 MHz and referenced to the internal standard TMS residual peak at δ 0.00.
cData (δ) acetone-d6 at 100.61 MHz and referenced to the internal standard TMS residual peak at δ 0.00.
dData (δ) measured in CDCl3 at 100.61 MHz and referenced to the solvent residual peak at δ 77.16.21

The conformation of the cardenolide ring system of digoxin has been proposed previously by investigation of its NMR spectroscopic parameters,22 and this was confirmed by that generated by a Mercury program,23 based on the reported crystal structure of digoxin (Figure 1).10 Using Snatzke’s method, the ECD spectrum induced by [Mo2(OAc)4] of digoxin showed a positive Cotton effect (CE) at 310 nm and a positive O-C-C-O dihedral angle (Figure 2), which indicated a (3'cS, 4'cS) absolute configuration.24 Thus, a (3S, 5R, 8R, 9S, 10S, 12R, 13S, 14S, 17R, 1'aR, 3'aS, 4'aS, 5'aR, 1'bS, 3'bS, 4'bS, 5'bR, 1'cS, 3'cS, 4'cS, 5'cR) absolute configuration was confirmed for digoxin from its induced ECD data and previously published crystal structure. A positive CE at 239 nm observed in the ECD spectrum of digoxin (Figure 3) correlated to a 17R configuration. This same configuration could be assigned for 1 from its ECD spectrum, which was consistent with that of digoxin (Figure 3). This determination was also supported by the resonances for H-17 and C-18 of 1, which were consistent with those reported for a cardiac glycoside comprising a 17β-lactone unit but different from those published for a 17-epi-cardiac glycoside containing a 17α-lactone unit.25 These changes would result from the stereoelectronic effects of the C-17 lactone unit, the same as those reported for bicyclic alcohols.26

An external file that holds a picture, illustration, etc.
Object name is nihms836224f1a.jpg
An external file that holds a picture, illustration, etc.
Object name is nihms836224f1b.jpg

A. COSY (An external file that holds a picture, illustration, etc.
Object name is nihms836224ig1.jpg, 1HAn external file that holds a picture, illustration, etc.
Object name is nihms836224ig2.jpg1H) and key HMBC (An external file that holds a picture, illustration, etc.
Object name is nihms836224ig3.jpg, 1HAn external file that holds a picture, illustration, etc.
Object name is nihms836224ig2.jpg13C) correlations of 1. B. Selected NOESY (An external file that holds a picture, illustration, etc.
Object name is nihms836224ig4.jpg, 1HAn external file that holds a picture, illustration, etc.
Object name is nihms836224ig2.jpg1H) correlations of 1. C. Partial structure showing the conformation of the central ring system as generated by Mercury CSD Version 3.1.1,23 and based on the reported crystal structure of digoxin.10 D. Selected NOESY (An external file that holds a picture, illustration, etc.
Object name is nihms836224ig4.jpg, 1HAn external file that holds a picture, illustration, etc.
Object name is nihms836224ig2.jpg1H) correlations of digoxin.

An external file that holds a picture, illustration, etc.
Object name is nihms836224f2.jpg

A. ECD spectrum of digoxin induced by [Mo2(OAc)4] (IECD, 1.0 mM digoxin in a 1.2 mM DMSO solution of dimolybdenum tetraacetate [(MoAc2)2], 1.2 mM, in DMSO). B. The relationship between the absolute configuration of C-3'c and 4'c of digoxin and the sign of the O-C-C-O dihedral angle.

An external file that holds a picture, illustration, etc.
Object name is nihms836224f3.jpg

ECD (A and B) and UV (C and D) spectra of compounds 1–5 and digoxin.

Conformational effects have been also observed for other protons and carbons of cardenolides. For example, the chemical shift of C-4 at δC 41.3 of a 3α-hydroxycardenolide appeared at δC 36.9 in its 3-epimer.27 Also, the NMR resonances for H-7/C-7 at δH 2.0/δC 28.0 and H-9/C-9 at δH 0.9/δC 50.0 in a 5αH-cardenolide shifted to those at δH 1.5/δC 22.0 for H-7/C-7 and at δH 1.7/δC 35.0 for H-9/C-9 in its 5-epimer.28 Following these observations, the resonances for C-4 at δC 34.9, C-7/H-7 at δC 19.0/δH 1.39 and 2.59, and C-9/H-9 at δC 39.1/δH 1.57 (Table 2) of 1 supported the presence of 3-O-β-glycosyl and 5β-hydroxy groups.2729 The occurrence of the same conformation for the cardenolide core of 1 as that for digoxin was also confirmed by the NOESY correlations between H-3 and H-7α, H-7α and H-9, OH-5 and H-8, H-8 and H-18, and H-18 and H-19 of 1 (Figure 1).

Owing to the overlapped signals observed for H-1'–H-3' and H-5' of 1, determination of the relative configuration of the sugar unit proved to be difficult. Fortunately, this problem was resolved when acetone-d6 rather than CDCl3 was employed as the NMR solvent (Table 1). The NOESY correlations observed between H-1' and H-9, H-3', H-5', and OCH3-2', and between OH-4' and OH-5, OH-19, and H-6' (Figure 1), indicated that H-1', H-3', H-4', and H-5' are α-oriented, and H-2' and H-6' are β-oriented. Therefore, a (3S, 5S, 8R, 9S, 10R, 13R, 14S, 17R, 1'R, 2'R, 3'S, 4'S, 5'R) absolute configuration could be assigned for 1 [(+)-19-hydroxykamaloside], with the structure determined as (3β,5β)-3-[(2,3-di-O-methyl-β-d-fucopyranosyl)oxy]-5,14,19-trihydroxycard-20(22)-enolide.

Compound 2 was isolated as an amorphous colorless powder. The similar UV and IR spectra to those of 1 indicated that 2 is also a cardiac glycoside. The same molecular formula of 2 [positive-ion HRESIMS m/z 603.3152 (calcd for C31H48O10Na, 603.3140), as observed in the HRESIMS] to that of 1 showed these compounds to be regioisomers. Comparison of their 1H and 13C NMR spectroscopic data (Tables 1 and and2)2) suggested that both compounds contain a 5,14-dihydroxycardenolide core substituted with a 2',3'-di-O-methylglycose unit at C-3. The 19-hydroxymethyl and 6'-methyl groups of 1 were substituted by a 19-methyl group and a 6'-hydroxymethyl group in 2, as supported by the HMBC cross-peaks observed between H-19 and C-1 and C-9, H-6' and C-4', and H-3' and C-6' for 2 (Figure S14, Supporting Information).

A 17R absolute configuration could be defined for 2 from the positive CE at 236 nm observed in its ECD spectrum, consistent with that of 1, and this was supported by the resonances for H-17 at δH 2.81 and C-18 at δC 15.7, which were indicative of a 17β-lactone unit.25 A 3-O-β-glycosyl moiety and a 5β-hydroxy group were evident in 2 due to the resonances for C-4 at δC 34.2, C-7/H-7 at δC 23.8/δH 1.17 and 1.89, and C-9/H-9 at δC 39.2/δH 1.51 (Table 2).2729 These were confirmed by the NOESY correlations observed between H-3 and H-7α, H-7α and H-9, H-8 and H-19, and H-18 and H-19 (Figure S15, Supporting Information). Thus, the same conformation and relative configuration of the cardenolide moiety of 1 occurred in 2. The NOESY correlations observed between H-1' and H-3, H-3', and H-5', and between H-4' and H-2' and H-6' (Figure S15, Supporting Information), indicated an α-orientation for H-1', H-3', H-5' and a β-orientation for H-2', H-4', and H-6'. These assignments were supported by the resonances for C-1'–C-6' of 2, which were closely comparable with those of asperoside.6 Thus, a (3S, 5S, 8R, 9S, 10R, 13R, 14S, 17R, 1'R, 2'R, 3'S, 4'R, 5'R) absolute configuration could be defined for 2 [(+)-5-hydroxyasperoside], with its structure characterized therefore as (3β,5β)-3-[(2,3-di-O-methyl-β-d-glucopyranosyl)oxy]-5,14-dihydroxycard-20(22)-enolide.

Compound 3 was isolated as an amorphous colorless powder. The similar UV, IR, and NMR spectra with those of 1 and 2 indicated that 3 is a further cardiac glycoside. The molecular formula determined from the positive-ion HRESIMS m/z 573.3054 (calcd for C30H46O9Na, 573.3034) indicated an OCH2 unit to be missing from 3, when compared with either 1 or 2, as confirmed by the NMR resonances for a single methoxy group at δH 3.38 s and δC 57.1 observed for 3 (Tables 1 and and2).2). Comparison of the 1H and 13C NMR spectroscopic data of 3 with those of the known compound 4 (Tables 1 and and22 and Table S1, Supporting Information) indicated that both compounds possess the same cardenolide aglycone but a different sugar unit. A methoxy group was proposed at C-2' of 3, as indicated by the HMBC cross-peak observed between the methoxy group and C-2'. The 17R absolute configuration could be defined for 3 from a positive CE at 241 nm in the ECD spectrum, which was consistent with 1 and 2. The same conformation as that of 2 for its cardenolide unit was evident for 3, from the closely comparable NMR resonances for this moiety of 2 and 3. A (3S, 5S, 8R, 9S, 10R, 13R, 14S, 17R, 1'R, 2'R, 3'S, 4'R, 5'R) absolute configuration was characterized for 3 from the NOESY correlations observed between H-3 and H-7α and H-17, H-7α and H-9, H-8 and H-18 and H-19, H-18 and H-22, H-1' and H-3, H-3', H-4', and H-5', and between OH-4' and H-6' (Figure S15, Supporting Information). Therefore, compound 3 was assigned structurally as (3β,5β)-3-[(2-O-methyl-β-d-fucopyranosyl)oxy]-5,14-dihydroxycard-20(22)-enolide, and has been accorded the trivial name, (+)-3'-de-O-methylkamaloside.

The structures of the two known cardiac glycosides 4 and 5 isolated from the stem bark of S. asper in the present study were determined by comparison of their spectroscopic data (Tables 1 and and22 and Table S1, Supporting Information) with literature data for (+)-3-O-β-d-fucopyranosylperiplogenin (4)18 and (+)-strebloside (5),5 respectively. The (3S, 5S, 8R, 9S, 10R, 13R, 14S, 17R, 1'R, 2'R, 3'S, 4'R, 5'R) and (3S, 5S, 8R, 9S, 10S, 13R, 14S, 17R, 1'R, 2'R, 3'S, 4'S, 5'R) absolute configurations were evident for 4 and 5, respectively, from their ECD and 2D NOESY NMR spectra, which were consistent with those of 1 (Figures 1 and and33 and Figures S15 and S16, Supporting Information).

Compounds 1–5, along with a commercially available sample of digoxin, were evaluated for their cytotoxicity against the HT-29 human colon cancer cell line, using paclitaxel as the positive control (Table 3).30 The compounds obtained in sufficient quantity were also tested for their activity toward the human MV4–11 and Kasumi-1 leukemia cell lines31 and the H1299 human non-small cell lung cancer cell line,32 using silvestrol and paclitaxel as the positive controls, respectively (Table 3). All compounds tested showed potent cytotoxicity that was comparable with digoxin, with IC50 values in the range 93–690 nM (Table 3), indicating that they are the cytotoxic principles of the stem bark of S. asper. Interestingly, both compounds 1 and 4 showed more potent activity than paclitaxel toward H1299 cells (Table 3).

Table 3

Cytotoxicity against Human Colon and Lung Cancer and Leukemia Cell Lines of 15a

compoundHT-29bMV4–11cKasumi-1dH1299e
1160186181290
2690NTgNTgNTg
3360NTgNTgNTg
4909997NTg
51709086110
digoxin38011193460
paclitaxelf0.81500
silvestrolf1124
aIC50 values are the concentration (nM) required for 50% inhibition of cell viability for a given test compound and were calculated using nonlinear regression analysis with measurements performed in triplicate and representative of three independent experiments, where the values generally agreed within 10%.
bIC50 value toward the HT-29 human colon cancer cell line with 72 h treatment.
c,dIC50 value toward the human MV4–11 and Kasumi-1 leukemia cell lines, respectively, with 48 h treatment.
eIC50 value toward the H1299 human non-small cell lung cancer cell line with a 24 h treatment
fPositive control.
gNot tested.

The major active compound, (+)-strebloside (5), and digoxin were also tested for their selectivity, using normal human CCD-112CoN colon, NL20 lung, and peripheral blood mononuclear cells.3133 The potency of both agents toward normal human cells was found to be much lower than that against human cancer cells, indicating that both 5 and digoxin showed selective cytotoxicity toward human colon and lung cancer and leukemia cells. The selectivity of 5 observed toward HT-29 cells and CCD-112CoN cells was greater than digoxin (Tables 3 and and44).

Table 4

Growth Inhibitory Activity against Normal Human Cells of 5a

compoundCCD-112CoNbblood cellscNL20d
5>20>2010.0
digoxin2.4>207.0
aIC50 values are the concentration (µM) required for 50% inhibition of cell viability with a given test compound and were calculated using nonlinear regression analysis with measurements performed in triplicate and representative of three independent experiments, where the values generally agreed within 10%.
bIC50 value toward normal human colon CCD-112CoN cell line with a 72 h treatment.
cIC50 value toward normal human peripheral blood mononuclear cells with a 48 h treatment.
dIC50 value toward normal human lung NL20 cell line with a 24 h treatment.

To characterize the functional groups that were responsible for the cytotoxicity of (+)-strebloside (5), several derivatives (Figure 4) were synthesized from 5 (Scheme 1 and Schemes S1–S4, Supporting Information), which was isolated in a relatively large quantity from S. asper with a yield of 0.15‰ w/w. The synthesized compounds were defined structurally through comparison of their spectroscopic data with those of 5 (Tables 1 and and22 and Tables S1–S4, Supporting Information) and evaluated by their cytotoxicity against the HT-29 cell line (Table 5).30

An external file that holds a picture, illustration, etc.
Object name is nihms836224f4.jpg

Structure of synthesized derivatives of (+)-strebloside (5).

An external file that holds a picture, illustration, etc.
Object name is nihms836224f6.jpg

Plausible mechanism of formation of 5g from the reaction of 5 and TFA.

Table 5

Cytotoxicity against Human Colon Cancer Cell Line of 5 and 5a5ga

compoundHT-29b
50.17
5a0.47
5b1.2
5c>20
5d2.8
5e>20
5f>20
5g>20
paclitaxelc0.0008
aIC50 values are the concentration required for 50% inhibition of cell viability after 72 h incubation for a given test compound and were calculated using nonlinear regression analysis with measurements performed in triplicate and representative of three independent experiments, where the values generally agreed within 10%.
bIC50 value (µM) toward the HT-29 human colon cancer cell line.
cPositive control.

Previously, the cytotoxic potency of a lignan lactone was found to be increased by acetylation of its saccharide unit,33 and the presence of different glycosyl units of cardiac glycosides contributed diversely to the cytotoxicity of these compounds, as exemplified by 2 (IC50 690 nM) and 4 (IC50 90 nM) (Table 3). Thus, acetylation was included as the first step in the investigation of the structure-cytotoxicity relationship of 5.

When 5 was treated with Ac2O and pyridine, (+)-4'-O-acetylstrebloside (5a) was produced. This product showed a sodiated molecular ion peak at m/z 643.3109 for a molecular formula of C33H48O11Na and the NMR resonances for a C-4' acetyl group (Table S1, Supporting Information), as indicated by the HMBC cross-peak between H-4' and the acetyl carbonyl group (Figure S14, Supporting Information). A (3S, 5S, 8R, 9S, 10S, 13R, 14S, 17R, 1'R, 2'R, 3'S, 4'S, 5'R) absolute configuration was defined for 5a from its ECD and 2D NOESY NMR spectra, which were consistent with those of 5 (Figure 3 and Figures S15 and S16, Supporting Information).When tested against the HT-29 cell line, 5a was slightly less active than 5 (Table 5).

To further test the importance of the C-4' substituent, several new benzoylated derivatives were prepared from 5. When 5 was reacted with benzoyl chloride and pyridine, (+)-4'-O-benzoylstrebloside (5b) and (+)-4'-O-benzoyl-19-nor-kamaloside-10-carboxylic acid (5c) were obtained. The product 5b showed a sodiated molecular ion peak at m/z 705.3260 for a molecular formula of C38H50O11. It also exhibited the NMR resonances for an additional benzoyl group, when compared with those of 5 (Tables 1 and and22 and Tables S2 and S3, Supporting Information). The C-4' benzoyl group was implied by the HMBC between H-4' and the benzoyl carbonyl group (Figure S14, Supporting Information). A molecular formula of C38H50O12, deduced for 5c from its sodiated molecular ion peak at m/z 721.3214 (calcd 721.3194), indicated that this compound contains an additional oxygen atom, when compared with 5b. A hydroxycarbonyl rather than a formyl group was defined at C-10 of 5c, on account of the resonance for its C-19 at δC 176.1 rather than at δC 208.3 for the same carbon of 5b (Table S3, Supporting Information). The product 5c could be formed by the aerial oxidation of the C-10 formyl group of 5b.

When 5 was treated with 4-chlorobenzoyl chloride, pyridine, and dichloromethane, (+)-4'-O-(4-chlorobenzoyl)strebloside (5d), (+)-4'-O-(4-chlorobenzoyl)-19-nor-kamaloside-10-carboxylic acid (5e), and (+)-4'-O-(4-chlorobenzoyl)-14(15)-anhydrostrebloside (5f) were obtained. Compound 5d was defined structurally from its sodiated molecular ion peak at m/z 739.2862 for a molecular formula of C38H49O11Cl and the NMR resonances for an additional 4-chlorobenzoyl group, when compared with 5 (Tables 1 and and22 and Tables S2 and S3, Supporting Information). The 4-chlorobenzoyl group was shown to be at the C-4' position from the HMBC between H-4' and the benzoyl carbonyl group (Figure S14, Supporting Information). The product 5e gave a sodiated molecular ion peak at m/z 755.2805 for a molecular formula of C38H49O12Cl, 16 Da more than 5d, indicating that 5e contains an additional oxygen. A hydroxycarbonyl rather than a formyl group was linked at C-10 of 5e, as indicated by the resonance for C-19 at δC 176.1 (Table S3, Supporting Information). Similar to the formation of 5c, 5e might be generated from the aerial oxidation of 5d. The product 5f showed a sodiated molecular ion peak at m/z 721.2753 for a molecular formula of C38H47O10Cl, 18 Da (H2O) less than 5d, indicating that 5f is a dehydrated analogue of 5d. This was verified by comparison of 13C NMR spectroscopic data of 5f with those of 5d, in which the resonances for C-14 at δC 85.2 and C-15 at δC 32.0 of 5d appeared at δC 152.7 for C-14 and δC 117.6 for C-15 of 5f (Tables S3 and S4, Supporting Information).

The same (3S, 5S, 8R, 9S, 10S, 13R, 14S, 17R, 1'R, 2'R, 3'S, 4'S, 5'R) absolute configuration as determined for 5 was assigned for 5b–5e from their consistent ECD and 2D NOESY NMR spectra. In turn, a (3S, 5S, 8R, 9S, 10S, 13R, 17S, 1'R, 2'R, 3'S, 4'S, 5'R) absolute configuration was defined for 5f, from comparison of its ECD and 2D NOESY NMR spectra with those of 5 (Figure 3 and Figure S16, Supporting Information). Different from the ECD bands observed for 5 (Figure 3), a negative CE at 239 nm and a positive CE at 216 nm, indicative of a 17S configuration, occurred in the ECD spectrum of 5f (Figure S16, Supporting Information). This probably resulted from the impact of the c/d-ring conformational change upon formation of the C-14/C-15 double bond, as evidenced previously.34 When evaluated toward the HT-29 cell line,30 5b and 5d were cytotoxic, but 5c, 5e, and 5f were all inactive in this regard (Table 5).

To test the role of other substituents on mediating its cytotoxicity, (+)-strebloside (5) was subjected to acid hydrolysis. Treatment with trifluoroacetic acid (TFA) at 70 °C for 1 h afforded a new analogue, 5g, with the C-19 formyl group transferred to the C-3 hydroxy group, which was purified from the reaction mixture. Compound 5g showed a sodiated molecular ion peak at m/z 391.1899 for a molecular formula of C23H28O4, with no resonances for a saccharide residue observed from its 1H and 13C NMR spectra, when compared with those of 5 (Tables 1, ,2,2, and Table S4, Supporting Information). Three double bonds could be deduced from its 13C NMR resonances, for C-5 at δC 125.0 and C-10 at δC 129.6, C-14 at δC 153.3 and C-15 at δC 116.0, and C-20 at δC 170.8 and C-22 at δC 116.5 (Table S4, Supporting Information). These assignments were supported by the HMBC observed between H-1 and C-5, H-2 and C-10, H-7 and C-14, H-15 and C-14 and C-17, and H-17 and C-22 (Figure S14, Supporting Information). A formate group indicated by a resonance at δC 161.2 (Table S4, Supporting Information) was assigned at the C-3 position, as implied by the HMBC between H-3 and the formate carbonyl group (Figure S14, Supporting Information). Thus, 5g was characterized as (3β)-3-O-formyl-19-nor-card-5(10),14(15),20(22)-trienolide or (+)-19-nor-5(10),14-dianhydrostrophanthidin-3-yl formate.

A 17S configuration was indicated for 5g from its sequential negative and positive CEs at 242 and 213 nm in the ECD spectrum, which was consistent with that of 5f (Figure S16, Supporting Information), and a (3S, 8R, 9S, 13R, 17S) absolute configuration was assigned from its 1D and 2D NOESY NMR spectra (Figure S15, Supporting Information). When evaluated against the HT-29 cell line,30 5g did not show any activity (Table 5).

The mechanism of the formation of 5g from 5 could be proposed based on the retro-Prins reaction.35 As presumed in Scheme 1, isomerization of the cis a/b-ring system of 5 to a trans a/b-ring system was followed by hemiacetalization involving the C-3 hydroxy and C-10 formyl groups. Formate 5g then could be formed via a retro-Prins reaction35 of the intermediate hemiacetal. (Other related mechanisms for this reaction are also possible). Such a retro-Prins-type reaction based mechanism could also be extended to the biosynthesis of other 19-nor cardiac glycosides, as reported from Antiaris toxicaria recently.36

Investigation of the cytotoxicity of 1–5 and 5a–5g showed that the C-3 saccharide residue, along with the C-5 and C-14 hydroxy and C-10 formyl groups, was important in the mediation of the cytotoxicity of 5 toward HT-29 cells, as indicated by the IC50 values (µM) of 5 (0.17) and 5g (>20) (Table 5). Comparison of the data for 4 (IC50 0.09 µM) with 2 (IC50 0.69 µM) (Table 3) showed that different C-3 sugar residues contributed to the resultant activity differentially. Methylation of the 2′-hydroxy group decreased compound potency moderately, as shown by 3 (IC50 0.36 µM) and 4 (IC50 0.09 µM) (Table 3). The activity was reduced in a sequence following the bulk of the ester group when an acetyl, a benzoyl, or a 4-chlorobenzoyl group was introduced to 4'-OH of 5, as implied by the IC50 values (µM) of 5 (0.17), 5a (0.47), 5b (1.2), and 5d (2.8) (Table 5). Furthermore, the C-14 hydroxy group alone plays an important role in potentiating the cytotoxic effect against HT-29 cells, as indicated by comparison of 5d (IC50 2.8 µM) and 5f (IC50 >20 µM) (Table 5). The activity was retained when the C-10 formyl group of 5 was reduced to a C-10 hydroxymethyl group, as indicated by the closely similar cytotoxicity of 1 (IC50 0.16 µM) and 5 (IC50 0.17 µM) (Table 3). However, oxidation of this formyl group to a hydroxycarbonyl group led to abolishment of the activity, as shown by data for 5b (IC50 1.2 µM) and 5c (IC50 >20 µM), as well as 5d (IC50 2.8 µM) and 5e (IC50 >20 µM) (Table 5).

Consistent with these observations, several cardiac glycosides were found previously to be more active than the corresponding aglycones,36 and the activity against a small panel of human cancer cell lines was increased when the carbohydrate moiety was attached at both C-2 and C-3 through a 1,4-dioxane moiety.20 No cytotoxicity was evident for several cardiac glycosides that contain a 17-hydroxy (IC50 > 40 µM)19 or a 19-hydroxycarbonyl (IC50 > 10 µM) group.28

(+)-Strebloside (5), the main cytotoxic compound identified from the stem bark of S. asper, was tested further for its cytotoxicity toward the MDA-MB-435 human melanoma, MDA-MB-231 human breast cancer, and OVCAR3 human ovarian cancer cell lines.37 It was found to be potently cytotoxic in all cases in the present study (Tables 3 and and6).6). Hence, (+)-strebloside (5) exhibits a broad spectrum of action toward both human solid tumor and leukemic cells.

Table 6

Cytotoxicity against Human Breast and Ovarian Cancer and Melanoma Cell Lines of 5a

compoundMDA-MB-231bMDA-MB-435cOVCAR3d
51349044
paclitaxele2.70.23.3
aIC50 values are the concentration (nM) required for 50% inhibition of cell viability for a given test compound with a 72 h treatment and were calculated using nonlinear regression analysis with measurements performed in triplicate and representative of three independent experiments, where the values generally agreed within 10%.
bIC50 value toward the MDA-MB-231 human breast cancer cell line.
cIC50 value toward the MDA-MB-435 human melanoma cell line.
dIC50 value toward the OVCAR3 human ovarian cancer cell line.
ePositive control.

(+)-Strebloside (5) was then deemed worthy of evaluation for its antitumor efficacy in an in vivo hollow fiber assay.33 Immunodeficient NCr nu/nu mice were implanted with hollow fibers containing MDA-MB-231, MDA-MB-435, or OVCAR3 cells and treated with 5 at doses of 1.0, 5.0, 10.0, 15.0, or 30.0 mg/kg, or with the vehicle control or paclitaxel (positive control, 5 mg/kg). In all cases, intraperitoneal (ip) administration for four days was used. The relative cell survival values from the treatment of 5 at 5.0, 10.0, 15.0, or 30.0 mg/kg (ip) toward MDA-MB-231 cells and at 10.0, 15.0, or 30.0 mg/kg (ip) toward OVCAR3 cells were all statistically significantly different from those of the vehicle control (Figure 5). However, no efficacy was observed for 5 toward MDA-MB-435 cells using the same dose range (Figure S18, Supporting Information). The values from the treatment of 5 at all cases were significantly different from those of paclitaxel (Figure 5), and no gross toxicities were observed for any of the animals in this in vivo study. These results indicated that compound 5 showed activity toward MDA-MB-231 and OVCAR3 cells at doses down to 5 mg/kg and 10 mg/kg, respectively, although this efficacy was less potent than paclitaxel.

An external file that holds a picture, illustration, etc.
Object name is nihms836224f5.jpg

Effect of (+)-strebloside (5) on the growth of human breast MDA-MB-231 and ovarian OVCAR3 cancer cells implanted in NCr nu/nu mice tested in an in vivo hollow fiber assay. The results are shown as the average percentage cell growth relative to control [columns, mean in each group (n = 12 for the control group and n = 6 for the treatment group); bars, SE; **p ≤ 0.05, ***p ≤ 0.01, and ****p ≤ 0.001 for significant differences from the control treatment.

The cardiac glycoside drug digoxin is known to show a narrow therapeutic index11 and has been administered (i.p.) to mice at a non-toxic dose of 0.7 mg/kg/day,13 while its close analogue digitoxin exhibited an oral LD50 value of 10–80 mg/kg in mice.38 (+)-Strebloside (5), however, did not exhibit obvious toxicity to the mice at doses (i.p.) up to 30 mg/kg, which is much higher than its effective dose of 5 mg/kg. Previously, several cardiac glycosides have been evaluated in clinical trials for the treatment of cancer,7,15 and a mixture of two cardiac glycosides that contain the same cardenolide aglycone as 5 showed an LD50 value of more than 500 mg/kg in an acute oral toxicity test, using six-week old ddY mice.38 Mechanistically, most cardiac glycosides mediate their cytotoxicity through inhibition of Na+,K+-ATPase followed by a Ca+2 flux pathway.7,8,39 Therefore, (+)-strebloside (5) seems to be a promising lead compound worthy of further investigation for development as a new anticancer agent.

EXPERIMENTAL SECTION

General Experimental Procedures

Optical rotations were measured at room temperature on a Perkin-Elmer model 343 polarimeter with a path length of 10 mm. UV spectra were recorded on a Hitachi U2910 ultraviolet spectrophotometer. ECD measurements were performed using a JASCO J-810 spectropolarimeter. IR spectra were recorded on a Nicolet 6700 FT-IR spectrometer. 1H and 13C, DEPT 90, DEPT 135, HSQC, HMBC, NOESY, and COSY NMR spectra were recorded at room temperature on a Bruker Avance DRX 400, Bruker Avance II 400, Bruker Avance III HD 700, or a Bruker Avance III HD 800 MHz NMR spectrometer. ESIMS and HRESIMS data were collected on a Bruker Maxis 4G Q-TOF mass spectrometer in the positive-ion mode. Column chromatography was conducted using silica gel (65 × 250 or 230 × 400 mesh, Sorbent Technologies, Atlanta, GA, USA). Analytical thin-layer chromatography (TLC) was performed on precoated silica gel 60 F254 plates (Sorbent Technologies, Atlanta, GA, USA). Sephadex LH-20 was purchased from Amersham Biosciences, Uppsala, Sweden. For visualization of TLC plates, H2SO4 was used as spray reagent. All procedures were carried out using solvents purchased from commercial sources and employed without further purification. Digoxin, paclitaxel, and other reagents for chemical synthesis were purchased from Sigma-Aldrich (St. Louis, MO, USA) (purity ≥ 98%).

Plant Material

A sample of the stem bark (acquisition number AA06920) of S. asper was collected in January 2010 by D.D.S. and T.N.N. (voucher specimen: DDS 14729) from a tree (eight-meters tall) at Dahang Village (11° 40.53' N; 109° 10.3' E), Nui Chua National Park, Ninh Hai District, Ninh Thuan Province, Vietnam. The voucher herbarium specimen has been deposited at the John G. Searle Herbarium of the Field Museum of Natural History, Chicago, IL, USA, under the accession numbers F-2294556.

Extraction and Isolation

The milled air-dried stem bark of S. asper (sample AA06920, 3100 g) was extracted with MeOH (4 L × 7) at room temperature. The solvent was evaporated in vacuo, and the dried MeOH extract (254.5 g, 8.2%) was resuspended in 10% H2O in MeOH (1500 mL) and partitioned with n-hexane (1000, 600, and 500 mL) to yield an n-hexane-soluble residue (57.0 g, 1.8%). Then, 200 mL of H2O were added to the aqueous MeOH layer, and this was partitioned with CHCl3 (1000, 500, and 500 mL). The combined CHCl3 partition was washed with a 1% aqueous solution of NaCl, to partially remove plant polyphenols, and the solvent was evaporated to afford a CHCl3-soluble extract (13.0 g, 0.42%). The CHCl3-soluble extract exhibited cytotoxicity toward the HT-29 cell line (IC50 < 1.0 µg/mL). Both the n-hexane- and water-soluble extracts were inactive (IC50 > 20.0 µg/mL) in the bioassay system used. The CHCl3-soluble extract (12.5 g) was subjected to silica gel column chromatography (6.0 × 45 cm) and eluted with a gradient of n-hexane-acetone. Eluates were pooled by TLC analysis to give 19 combined fractions (D2F1–D2F19), and fractions D2F11–D2F19 were deemed active against HT-29 cells (IC50 < 1 µg/mL).

Fractions D2F11–D2F13 were combined and chromatographed over a silica gel column (4.5 × 40 cm), eluted with a gradient of CH2Cl2 (DCM)-MeOH to yield three pooled subfractions, D2F11F1–D2F11F3. Of these, fractions D2F11F2 and D2F11F3 were combined and further chromatographed over a silica gel column (4.5 × 40 cm), eluted with a gradient of DCM-MeOH, and purified by separation over a Sephadex LH-20 column, eluted with DCM-MeOH (1:1), affording (+)-strebloside (5, 420 mg).

Combined fractions D2F14–D2F16 were separated by silica gel chromatography (4.5 × 40 cm) eluted with a gradient of DCM-MeOH to yield two pooled subfractions, D2F14F1 and D2F14F2. Of these, fraction D2F14F1 was further chromatographed over a silica gel column (2.5 × 30 cm), eluted with a gradient of DCM-MeOH and purified by separation over a Sephadex LH-20 column, eluted with DCM-MeOH (1:1), to yield a further quantity of (+)-strebloside (5, 30 mg). Fraction D2F14F2 was separated over a series of silica gel columns (2.5 × 30 cm), eluted with a gradient mixture of DCM-MeOH, and purified by separation over Sephadex LH-20 columns, eluted with DCM -MeOH (1:1), to afford (+)-19-hydroxykamaloside (1, 20 mg), (+)-5-hydroxyasperoside (2, 2.0 mg), and (+)-3'-de-O-methylkamaloside (3, 1.0 mg).

Combined fractions D2F17–D2F19 were chromatographed over a silica gel column (2.5 × 30 cm), eluted with a gradient of DCM-MeOH, to yield two pooled subfractions, D2F17F1 and D2F17F2. Of these, fraction D2F17F1 was further chromatographed over a silica gel column (2.5 × 20 cm), eluted with a gradient of DCM-MeOH and purified by passage over a column containing Sephadex LH-20, eluted with DCM-MeOH (1:1), to yield an additional amount of (+)-19-hydroxykamaloside (1, 5.0 mg). Fraction D2F17F2 was separated over a series of silica gel columns (2.5 × 20 cm), eluted with a gradient of DCM-MeOH, and purified by separation over Sephadex LH-20 columns, eluted with DCM-MeOH (1:1), to afford (+)-3-O-β-d-fucopyranosylperiplogenin (4, 3.0 mg).

(+)-19-Hydroxykamaloside (1)

Amorphous colorless powder; [α]20D +13 (c 0.1, MeOH); UV (MeOH) λmax (log ε) 216 (3.95) nm; ECD (MeOH, nm) λmax (Δε) 238 (+2.76); IR (dried film) νmax 3459, 2939, 1743, 1621, 1447, 1371, 1169, 1067 cm−1; 1H and 13C NMR data, see Tables 1 and and2;2; positive-ion HRESIMS m/z 603.3153 (calcd for C31H48O10Na, 603.3140).

(+)-5-Hydroxyasperoside (2)

Amorphous colorless powder; [α]20D +83 (c 0.1, MeOH); UV (MeOH) λmax (log ε) 216 (4.13) nm; ECD (MeOH, nm) λmax (Δε) 236 (+4.05); IR (dried film) νmax 3445, 2928, 1739, 1627, 1450, 1378, 1274, 1077 cm−1; 1H and 13C NMR data, see Tables 1 and and2;2; positive-ion HRESIMS m/z 603.3152 (calcd for C31H48O10Na, 603.3140).

(+)-3'-De-O-methylkamaloside (3)

Amorphous colorless powder; [α]20D +50 (c 0.1, MeOH); UV (MeOH) λmax (log ε) 216 (4.08) nm; ECD (MeOH, nm) λmax (Δε) 241 (+3.99); IR (dried film) νmax 3479, 2931, 1743, 1621, 1449, 1365, 1222, 1166, 1079 cm−1; 1H and 13C NMR data, see Tables 1 and and2;2; positive-ion HRESIMS m/z 573.3054 (calcd for C30H46O9Na, 573.3034).

Acetylation of (+)-Strebloside (5)

To a dried 25 mL glass vial equipped with a magnetic stirrer, containing 11.6 mg (0.02 mmol) of (+)-strebloside (5), 50 µL of Ac2O and 1 mL pyridine were added, and the vial was sealed. After the mixture was stirred at 70 °C for 1 h, it was cooled to room temperature. Then, DCM (5 mL) was added, and the solution was extracted with distilled H2O. The organic layer was washed with distilled H2O, and evaporated at reduced pressure. The residue was purified by silica gel column chromatography, using n-hexane-acetone (5:1→1:1), to afford 8.0 mg (0.013 mmol) of 5a (64.5%).

(+)-4'-O-Acetylstrebloside or (3β,5β)-3-[(2,3-Di-O-methyl-4-O-acetyl-β-d-fucopyranosyl)oxy]-5,14-dihydroxy-19-oxocard-20(22)-enolide (5a)

Amorphous colorless powder; [α]20D +20 (c 0.1, MeOH); UV (MeOH) λmax (log ε) 216 (3.96) nm; ECD (MeOH, nm) λmax (Δε) 235 (+2.46); IR (dried film) νmax 3501, 2940, 1747, 1622, 1447, 1374, 1239, 1067 cm−1; 1H and 13C NMR data, see Table S1 (Supporting Information); positive-ion HRESIMS m/z 643.3109 (calcd for C33H48O11Na, 643.3094).

Benzoylation of (+)-Strebloside (5)

To a dried 25 mL glass vial equipped with a magnetic stirrer, containing 17.3 mg (0.03 mmol) of (+)-strebloside (5), 1 mL (8.6 mmol) of benzoyl chloride and 100 µL of pyridine were added, and the vial was sealed. After the mixture was stirred at 70 °C for 1 h, it was cooled to room temperature. Then, DCM (5 mL) was added, and the solution was extracted with distilled H2O. The organic layer was washed with distilled H2O, and evaporated at reduced pressure. The residue was purified by silica gel column chromatography, using n-hexane-acetone (5:1→1:1), to afford 2.0 mg (0.003 mmol) of 5b (10%) and 3.0 mg (0.004 mmol) of 5c (14.3%).

(+)-4'-O-Benzoylstrebloside or (3β,5β)-3-[(2,3-Di-O-methyl-4-O-benzoyl-β-d-fucopyranosyl)oxy]-5,14-dihydroxy-19-oxocard-20(22)-enolide (5b)

Amorphous colorless powder; [α]20D +10 (c 0.1, MeOH); UV (MeOH) λmax (log ε) 222 (3.97) nm; ECD (MeOH, nm) λmax (Δε) 235 (+2.02); IR (dried film) νmax 3501, 2926, 1722, 1621, 1452, 1383, 1275, 1161, 1068 cm−1; 1H and 13C NMR data, see Tables S2 and S3 (Supporting Information); positive-ion HRESIMS m/z 705.3260 (calcd for C38H50O11Na, 705.3245).

(+)-4'-O-Benzoyl-19-nor-kamaloside-10-carboxylic acid or (3β,5β,14β)-3-[(2,3-Di-O-methyl-4-O-benzoyl-β-d-fucopyranosyl)oxy]-5,14,21-trihydroxy-24-norchol-20(22)-ene-19,23-dioic acid-γ-lactone (5c)

Amorphous colorless powder; [α]20D +30 (c 0.1, MeOH); UV (MeOH) λmax (log ε) 224 (4.02) nm; ECD (MeOH, nm) λmax (Δε) 235 (+4.83); IR (dried film) νmax 3501, 2936, 1723, 1621, 1451, 1383, 1275, 1161. 1068 cm−1; 1H and 13C NMR data, see Tables S2 and S3 (Supporting Information); positive-ion HRESIMS m/z 721.3214 (calcd for C38H50O12Na, 721.3194).

4-Chlorobenzoylation of (+)-Strebloside (5)

To a dried 25 mL glass vial equipped with a magnetic stirrer, containing 17.3 mg (0.03 mmol) of (+)-strebloside (5), 216 µL (1.5 mmol) of 4-chlorobenzoyl chloride, 100 µL of pyridine, and 5 mL of DCM were added, and the vial was sealed. After the mixture was stirred at room temperature overnight, DCM (5 mL) was added, and the solution was extracted with distilled H2O. The organic layer was washed with distilled H2O, and evaporated at reduced pressure. The residue was purified by silica gel column chromatography, using n-hexane-acetone (5:1→1:1), to afford 2.5 mg (0.0035 mmol) of 5d (11.9%), 5.0 mg (0.007 mmol) 5e (22.7%), and 3.0 mg (0.004 mmol) of 5f (13.9%).

(+)-4'-O-(4-Chlorobenzoyl)strebloside or (3β,5β)-3-{[2,3–Di-O-methyl-4-O-(4-chloro)benzoyl-β-d-fucopyranosyl]oxy}-5,14-dihydroxy-19-oxocard-20(22)-enolide (5d)

Amorphous colorless powder; [α]20D +20 (c 0.1, MeOH); UV (MeOH) λmax (log ε) 238 (3.87), 201 (4.11) nm; ECD (MeOH, nm) λmax (Δε) 237 (+2.31); IR (dried film) νmax 3501, 2922, 1744, 1729, 1622, 1591, 1455, 1380, 1273, 1169, 1092 cm−1; 1H and 13C NMR data, see Tables S2 and S3 (Supporting Information); positive-ion HRESIMS m/z 739.2862 (calcd for C38H49O11ClNa, 739.2856).

(+)-4'-O-(4-Chlorobenzoyl)-19-nor-kamaloside-10-carboxylic acid or (3β,5β,14β)-3-{[2,3–Di-O-methyl-4-O-(4-chloro)benzoyl-β-d-fucopyranosyl]oxy}-5,14,21-trihydroxy-24-norchol-20(22)-ene-19,23-dioic acid-γ-lactone (5e)

Amorphous colorless powder; [α]20D +20 (c 0.1, MeOH); UV (MeOH) λmax (log ε) 233 (4.10), 201 (4.33) nm; ECD (MeOH, nm) λmax (Δε) 237 (+2.31); IR (dried film) νmax 3508, 2926, 1731, 1622, 1594, 1447, 1338, 1273, 1067 cm−1; 1H and 13C NMR data, see Tables S2 and S3 (Supporting Information); positive-ion HRESIMS m/z 755.2805 (calcd for C38H49O12ClNa, 755.2805).

(+)-4'-O-(4-Chlorobenzoyl)-14(15)-anhydrostrebloside or (3β,5β)-3-{[2,3–Di-O-methyl-4-O-(4-chloro)benzoyl-β-d-fucopyranosyl]oxy}-5-hydroxy-19-oxocard-14(15),20(22)-dienolide (5f)

Amorphous colorless powder; [α]20D +10 (c 0.1, MeOH); UV (MeOH) λmax (log ε) 238 (3.86), 201 (4.17) nm; ECD (MeOH, nm) λmax (Δε) 239 (–2.34), 216 (+3.28); IR (dried film) νmax 3508, 2931, 1749, 1716, 1629, 1594, 1488, 1337, 1273, 1170, 1092 cm−1; 1H and 13C NMR data, see Table S4 (Supporting Information); positive-ion HRESIMS m/z 721.2753 (calcd for C38H47O10ClNa, 721.2750).

Acid Hydrolysis of (+)-Strebloside (5)

To a 25 mL glass vial equipped with a magnetic stirrer, containing 17.3 mg (0.03 mmol) of (+)-strebloside (5), 775 µL (10 mmol) of trifluoroacetic acid and 5 mL of acetone were added, and the vial was sealed. After the mixture was stirred at 70 °C for 1 h, it was cooled to room temperature. Then, DCM (5 mL) was added, and the solution was extracted with distilled H2O. The organic layer was washed with distilled H2O, and evaporated at reduced pressure. The residue was purified by silica gel column chromatography, using n-hexane-acetone (5:1→1:1), to afford 2.0 mg (0.005 mmol) of 5g (18.0%).

(+)-19-Nor-5(10),14-dianhydrostrophanthidin-3-yl formate or (3β)-3-O-Formyl-19-nor-card-5(10),14(15),20(22)-trienolide (5g)

Amorphous colorless powder; [α]20D +10 (c 0.1, MeOH); UV (MeOH) λmax (log ε) 209 (3.64) nm; ECD (MeOH, nm) λmax (Δε) 242 (–2.51), 213 (+4.67); IR (dried film) νmax 3445, 2924, 1747, 1629, 1453, 1378, 1181, 1044 cm−1; 1H and 13C NMR data, see Table S4 (Supporting Information); positive-ion HRESIMS m/z 391.1899 (calcd for C23H28O4Na, 391.1880).

Determination of Absolute Configuration of the 1,2-Diol Unit of Digoxin

The ECD spectra of digoxin (1.0 mM in a DMSO solution) and [Mo2(OAc)4] (1.2 mM in a DMSO solution), and the induced ECD spectrum of digoxin [1.0 mM in a [Mo2(OAc)4] (1.2 mM) DMSO solution for 20 min) were measured on a JASCO J-810 spectropolarimeter in a 0.5 cm cell from 500 nm to 270 nm under identical conditions. The relation between the absolute configuration of the 1,2-diol moiety of digoxin and the sign of the O-C-C-O dihedral angles was performed following a reported procedure.24 [Mo2(OAc)4] (Sigma-Aldrich) and DMSO (Sigma Aldrich, anhydrous ≥99.9%) were commercially available and used without further purification.

Cell Lines and Peripheral Blood Cells

All cell lines were purchased from the American Type Culture Collection (ATCC, Manassas, VA, USA). Peripheral blood was obtained from healthy human donors under an Institutional Review Board-approved protocol from the Ohio State University (2016C0032), and mononuclear cells were isolated by Ficoll density gradient centrifugation. The HT-29 human colon cancer cell line was cultured in MEME medium (Hyclone, Logan, UT, USA) supplemented with heat-inactivated fetal bovine serum (FBS, 10%), penicillin (100 units/mL), streptomycin (100 µg/mL), and amphotericin B (fungizone, 0.25 µg/mL). The human MV4–11 and Kasumi-1 myeloid leukemia cell lines and normal human peripheral blood mononuclear cells were cultured in RPMI 1640 medium (Invitrogen, Carlsbad, CA, USA), supplemented with FBS (20%), penicillin (100 units/mL), and streptomycin (100 µg/mL). The MDA-MB-231 human breast cancer, MDA-MB-435 human melanoma, and OVCAR3 human ovarian cancer cell lines were cultured in RPMI 1640 medium, supplemented with FBS (10%), penicillin (100 units/mL), and streptomycin (100 µg/mL). The H1299 human non-small cell lung cancer cell line was maintained in Dulbecco’s Modified Eagle Medium (DMEM containing 25 mM glucose) supplemented with FBS (10%), penicillin (100 units/mL), and streptomycin (100 µg/mL). The CCD-112CoN normal human colon cell line was cultured in MEME medium supplemented with FBS (10%), penicillin G-streptomycin-fungizone solution (PSF, 1%), 1.0 mM sodium pyruvate, 0.1 mM non-essential amino acid, and 1.5 g/L sodium bicarbonate. The NL20 normal human lung cell line was maintained in Ham’s F12 medium, supplemented with FBS (4%), penicillin (100 units/mL), and streptomycin (100 µg/mL), 0.1 mM non-essential amino acids, 0.005 mg/mL insulin, 10 ng/mL epidermal growth factor, 0.001 mg/mL transferrin, and 500 ng/mL hydrocortisone. All cultures were maintained at 37 °C in 5% CO2.

Cytotoxicity Assay against the HT-29 Cell Line

The cytotoxicity of the test compounds and digoxin was screened against HT-29 cells by a previously reported procedure,30 with the vehicle and paclitaxel used as the negative and positive control, respectively.

Cytotoxicity Assay toward Human Leukemia Cells and Effect of Testing against Normal Human Peripheral Blood Mononuclear Cells

Cell growth inhibition/cytotoxicity was evaluated after 48 h incubation by an MTS assay (CellTiter 96, Promega, Madison WI, USA), using silvestrol.31 Briefly, MV4–11, Kasumi-1, or normal human peripheral blood mononuclear cells were seeded in 96-well plates and incubated with test samples or silvestrol (both dissolved in DMSO and diluted to different concentrations) or the vehicle (DMSO) for 48 h. Then, 20 µL of the CellTiter 96 reagent were added to each well. After an additional 4 h incubation, the optical density at 490 nm was measured, with IC50 values calculated with respect to the control samples.

Cytotoxicity Assay against the H1299 and NL20 Cell Lines

This assay was performed against the H1299 human non-small cell lung cancer and human non-tumorigenic NL20 lung cell lines using a reported procedure.32 Cell growth or proliferation was assessed using the MTT proliferation assay kit (Cayman, Ann Arbor, MI, USA). Briefly, 5000 cells were seeded in each well of a 96-well plate and cells were treated with or without test compounds. After cells were cultured for 24 h, they were treated with MTT for 4 h. Then, the medium was removed, and 100 µL Crystal Dissolving Solution was added to each well. The absorbance of the solution was measured at 570 nm, with IC50 values calculated from the vehicle control.

Cytotoxicity Assay against the CCD-112CoN Cell Line

Following a procedure reported previously,33 the cytotoxicity of compound 5, along with commercial digoxin, was screened against the CCD-112CoN cell line.

Cytotoxicity Assay against MDA-MB-231, MDA-MB-435, or OVCAR3 Cell Lines

Following a protocol used previously,37 MDA-MB-231, MDA-MB-435, or OVCAR3 cells in log phase growth were harvested by trypsinization and seeded in 96-well clear flat-bottomed plates (Microtest 96, Falcon). Cells were incubated at 37 °C in 5% CO2 overnight and then treated with the samples or paclitaxel (the positive control) (both dissolved in DMSO and diluted to different concentrations required) or the vehicle (DMSO) for 72 h. Viability of cells was evaluated by a commercial absorbance assay (CellTiter 96 AQueous One Solution Cell Proliferation Assay, Promega Corp, Madison, WI, USA), with the IC50 values calculated from the vehicle control.

Animals

Seven-week-old immunodeficient NCr nu/nu mice were purchased from The Jackson Laboratory (Bar Harbor, ME, USA) and housed in cages at room temperature with a relative humidity of 50–60% under 12:12 h light–dark cycle. This procedure was approved by the University of Illinois at Chicago Animal Care and Use Committee (protocol number 13-057), and the mice were treated in accordance with the institutional guidelines for animal care.

In Vivo Hollow Fiber Assay

An in vivo hollow fiber assay was performed for the evaluation of antitumor efficacy of 5 using procedures previously described in detail.33 In brief, seven-week-old immunodeficient NCr nu/nu mice were divided into seven groups, including a vehicle negative control group (12 mice), a paclitaxel positive control group (12 mice), and (+)-strebloside (5) treatment groups with dose of 1, 5, 10, 15, and 30 mg/kg, respectively (six mice for each group treated with compound 5). (+)-Strebloside (5) was dissolved initially in EtOH and subsequently diluted with Tween 20. The mixture was diluted with distilled water to 5% EtOH and 5% Tween 20. Hollow fibers containing MDA-MB-231, MDA-MB-435, or OVCAR3 cells were implanted into the abdominal cavity of mice on day 0. The mice were injected i.p. once daily for four days (from day 3 to day 6) with 5 (different dose as indicated) and the vehicle negative and paclitaxel (5 mg/kg) positive controls, respectively. On day 7, all the remaining mice were sacrificed. The fibers were retrieved, and viable cell mass was evaluated by a modified MTT [3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide] assay. The percentage of the net growth for the cells in each treatment group was calculated by subtracting the day 0 absorbance from the day 7 absorbance and dividing this difference by the net growth in the vehicle control (minus value between the day 7 and the day 0) (Figure 5). Each mouse was weighed daily during the study, and no signs of toxicity was found among the test animals even at the highest treatment dose (30 mg/kg) of 5.

Statistical Analysis

The in vitro measurements were performed in triplicate and are representative of three independent experiments, where the values generally agreed within 10%. The dose response curve was calculated for IC50 determinations using non-linear regression analysis (Table Curve2DV4; AISN Software Inc. Mapleton, OR, USA). Differences among samples were assessed by one-way ANOVA followed by Tukey-Kramer’s test, and the significance level was set at p < 0.05. The data from an in vivo hollow fiber assay were analyzed with linear mixed effect models to take account of the correlation of the observations from the same trial. Holm’s procedure was used to adjust for multiple comparisons of the multiple doses used for the same cell line. An adjusted p value less than 0.05 was considered statistically significant.

Supplementary Material

Supporting Information

Acknowledgments

This investigation was supported by grant P01 CA125066 and by grant P30 CA016058 funded by the National Cancer Institute, NIH, Bethesda, MD. The plant sample of Streblus asper was collected under a collaborative arrangement between the University of Illinois at Chicago (USA) and the Institute of Ecology and Biological Resources of the Vietnam Academy of Science and Technology, Hanoi (Vietnam). We thank Drs. David J. Hart and Judith C. Gallucci, Department of Chemistry and Biochemistry, The Ohio State University, for the helpful comments and suggestions about the mechanism of the formation of compound 5g and the crystal structure of digoxin, respectively. We thank Dr. Chia-Hsien Wu, Division of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, The Ohio State University, for his help for the chemical synthesis. Drs. Arpad Somogy, Nanette M. Kleinholz, and Yu Cao of the Mass Spectrometry and Proteomics of the Campus Chemical Instrument Center, The Ohio State University, are thanked for acquisition of the MS data. Drs. Craig A. McElroy (College of Pharmacy) and Chunhua Yuan (Nuclear Magnetic Resonance Laboratory, Campus Chemical Instrument Center), The Ohio State University, are thanked for access to some of the NMR instrumentations used in this investigation and for the 700 or 800 MHz NMR measurements, respectively.

Footnotes

ASSOCIATED CONTENT

Supporting Information.

The Supporting Information is available free of charge on the ACS Publications website at DOI: 10.1021/acschembio.5b01018.

  • Mass and NMR spectra of compounds 1–5 and 5a–5g; ECD and UV spectra of compounds 5a–5g; schemes of synthesis of 5a–5g from 5; diagrams of COSY, the key HMBC, and selective NOESY correlations of compounds 2–5 and 5a–5g; in vivo data of 5 against MDA-MB-435 ells; assignments of the 1H and 13C NMR data of the known compounds 4 and 5a and the synthetic derivatives 5a–5g; and analytical data of the known compounds 4 and 5.

ORCID

A. Douglas Kinghorn: 0000-0002-6647-8707

The authors declare no competing financial interest.

REFERENCES

1. Datwyler SL, Weiblen GD. Am. J. Bot. 2004;91:767–777. [Abstract] [Google Scholar]
2. Suresh Kumar RB, Kar B, Dolai N, Karmakar I, Bhattacharya S, Haldar PK. Interdiscip. Toxicol. 2015;8:125–130. [Europe PMC free article] [Abstract] [Google Scholar]
3. Khare MP, Bhatnagar SS, Schindler O, Reichstein T. Helv. Chim. Acta. 1962;45:1515–1534. [Google Scholar]
4. Saxena VK, Chaturvedi SK. Planta Med. 1985:343–344. [Abstract] [Google Scholar]
5. Fiebig M, Duh C-Y, Pezzuto JM, Kinghorn AD, Farnsworth NR. J. Nat. Prod. 1985;48:981–985. [Abstract] [Google Scholar]
6. Hano Y, Juma P, Abliz Z, Sun H-D, Nomura T. Heterocycles. 2003;59:805–809. [Google Scholar]
7. Newman RA, Yang P, Pawlus AD, Block KI. Mol. Intervent. 2008;8:36–49. [Abstract] [Google Scholar]
8. Slingerland M, Cerella C, Guchelaar HJ, Diederich M, Gelderblom H. Invest. New Drugs. 2013;31:1087–1094. [Abstract] [Google Scholar]
9. Smith S. J. Chem. Soc. 1930:508–510. [Google Scholar]
10. Go K, Kartha G. Acta Cryst. 1980;B36:1811–1819. [Google Scholar]
11. Eichhorn EJ, Gheorghiade M. Prog. Cardiovasc. Dis. 2002;44:251–266. [Abstract] [Google Scholar]
12. Stenkvist B, Bengtsson E, Eriksson O, Holmquist J, Nordin B, Westman-Naeser S. Lancet. 1979;313:563. [Abstract] [Google Scholar]
13. Svensson A, Azarbayjani F, Bäckman U, Matsumoto T, Christofferson R. Anticancer Res. 2005;25:207–212. [Abstract] [Google Scholar]
14. Platz EA, Yegnasubramanian S, Liu JO, Chong CR, Shim JS, Kenfield SA, Stampfer MJ, Willett WC, Giovannucci E, Nelson WG. Cancer Discov. 2011;1:68–77. [Europe PMC free article] [Abstract] [Google Scholar]
15. Cragg GM, Grothaus PG, Newman DJ. J. Nat. Prod. 2014;77:703–723. [Abstract] [Google Scholar]
16. Felth J, Rickardson L, Rosén J, Wickström M, Fryknäs M, Lindskog M, Bohlin L, Gullbo J. J. Nat. Prod. 2009;72:1969–1974. [Abstract] [Google Scholar]
17. Kinghorn AD, Carcache de Blanco EJ, Chai H-B, Orjala J, Farnsworth NR, Soejarto DD, Oberlies NH, Wani MC, Kroll DJ, Pearce CJ, Swanson SM, Kramer RA, Rose WC, Fairchild CR, Vite GD, Emanuel S, Jarjoura D, Cope FO. Pure Appl. Chem. 2009;81:1051–1063. [Europe PMC free article] [Abstract] [Google Scholar]
18. Wang T-M, Hojo T, Ran F-X, Wang R-F, Wang R-Q, Chen H-B, Cui J-R, Shang M-Y, Cai S-Q. J. Nat. Prod. 2007;70:1429–1433. [Abstract] [Google Scholar]
19. Araya JJ, Kindscher K, Timmermann BN. J. Nat. Prod. 2012;75:400–407. [Abstract] [Google Scholar]
20. Zhang R-R, Tian H-Y, Tan Y-F, Chung T-Y, Sun X-H, Xia X, Ye W-C, Middleton DA, Fedosova N, Esmann M, Tzen JTC, Jiang R-W. Org. Biomol. Chem. 2014;12:8919–8929. [Abstract] [Google Scholar]
21. Gottlieb HE, Kotlyar V, Nudelman A. J. Org. Chem. 1997;62:7512–7515. [Abstract] [Google Scholar]
22. Aulabaugh AE, Crouch RC, Martin GE, Ragouzeos A, Shockcor JP, Spitzer TD, Farrant RD, Hudson BD, Lindon JC. Carbohydr. Res. 1992;230:201–212. [Abstract] [Google Scholar]
23. Macrae CF, Bruno IJ, Chisholm JA, Edgington PR, McCabe P, Pidcock E, Rodriguez-Monge L, Taylor R, van de Streek J, Wood PA. J. Appl. Cryst. 2008;41:466–470. [Google Scholar]
24. Frelek J, Ikekawa N, Takatsuto S, Snatzke G. Chirality. 1997;9:578–582. [Google Scholar]
25. Gil RR, Lin L-Z, Chai H-B, Pezzuto JM, Cordell GA. J. Nat. Prod. 1995;58:848–856. [Abstract] [Google Scholar]
26. Seidl PR, Tostes JGR, Carneiro JWdeM, Taft CA, Dias JF. J. Mol. Struct. (Theochem) 2001;539:163–169. [Google Scholar]
27. Junior P, Wichtl M. Phytochemistry. 1980;19:2193–2197. [Google Scholar]
28. Tian D-M, Cheng H-Y, Jiang M-M, Shen W-Z, Tang J-S, Yao X-S. J. Nat. Prod. 2016;79:38–50. [Abstract] [Google Scholar]
29. Rovinski JM, Tewalt GL, Sneden AT. J. Nat. Prod. 1987;50:211–216. [Abstract] [Google Scholar]
30. Ren Y, Matthew S, Lantvit DD, Ninh TN, Chai H-B, Fuchs JR, Soejarto DD, Carcache de Blanco EJ, Swanson SM, Kinghorn AD. J. Nat. Prod. 2011;74:1117–1125. [Europe PMC free article] [Abstract] [Google Scholar]
31. Alachkar H, Santhanam R, Harb JG, Lucas DM, Oaks JJ, Hickey CJ, Pan L, Kinghorn AD, Caligiuri MA, Perrotti D, Byrd JC, Garzon R, Grever MR, Marcucci G. J. Hematol. Oncol. 2013;6:21. [Europe PMC free article] [Abstract] [Google Scholar]
32. Liu Y, Cao Y, Zhang W, Bergmeier S, Qian Y, Akbar H, Colvin R, Ding J, Tong L, Wu S, Hines J, Chen X. Mol. Cancer Ther. 2012;11:1672–1682. [Abstract] [Google Scholar]
33. Ren Y, Lantvit DD, Deng Y, Kanagasabai R, Gallucci JC, Ninh TN, Chai H-B, Soejarto DD, Fuchs JR, Yalowich JC, Yu J, Swanson SM, Kinghorn AD. J. Nat. Prod. 2014;77:1494–1504. [Europe PMC free article] [Abstract] [Google Scholar]
34. Emanuelsson R, Löfås H, Wallner A, Nauroozi D, Baumgartner J, Marschner C, Ahuja R, Ott S, Grigoriev A, Ottosson H. Chem Eur. J. 2014;20:9304–9311. [Abstract] [Google Scholar]
35. Al-Qallaf FAH, Johnstone RAW, Liu J-Y, Lu L, Whittaker D. J. Chem. Soc. Perkin Trans. 1999;2:1421–1423. [Google Scholar]
36. Shi L-S, Kuo S-C, Sun H-D, Morris-Natschke SL, Lee K-H, Wu T-S. Bioorg. Med. Chem. 2014;22:1889–1898. [Europe PMC free article] [Abstract] [Google Scholar]
37. Zhao M, Onakpa MM, Santarsiero BD, Chen W-L, Szymulanska-Ramamurthy KM, Swanson SM, Burdette JE, Che C-T. J. Nat. Prod. 2015;78:2731–2737. [Europe PMC free article] [Abstract] [Google Scholar]
38. Goda Y, Sakai S, Nakamura T, Akiyama H, Toyoda M. Shokuhin Eiseigaku Zasshi. 1998;39:256–265. [Google Scholar]
39. Durlacher CT, Chow K, Chen X-W, He Z-X, Zhang X, Yang T, Zhou S-F. Clin. Exp. Pharmacol. Physiol. 2015;42:427–443. [Abstract] [Google Scholar]

Citations & impact 


Impact metrics

Jump to Citations
Jump to Data

Citations of article over time

Alternative metrics

Altmetric item for https://www.altmetric.com/details/46345871
Altmetric
Discover the attention surrounding your research
https://www.altmetric.com/details/46345871

Smart citations by scite.ai
Smart citations by scite.ai include citation statements extracted from the full text of the citing article. The number of the statements may be higher than the number of citations provided by EuropePMC if one paper cites another multiple times or lower if scite has not yet processed some of the citing articles.
Explore citation contexts and check if this article has been supported or disputed.
https://scite.ai/reports/10.1021/acs.jnatprod.6b00924

Supporting
Mentioning
Contrasting
0
98
0

Article citations


Go to all (24) article citations

Data 


Data behind the article

This data has been text mined from the article, or deposited into data resources.

Similar Articles 


To arrive at the top five similar articles we use a word-weighted algorithm to compare words from the Title and Abstract of each citation.

Funding 


Funders who supported this work.

NCI NIH HHS (2)

National Cancer Institute (2)