You are on page 1of 8

Med Chem Res (2017) 26:1585–1592 MEDICINAL

DOI 10.1007/s00044-017-1871-4
CHEMISTRY
RESEARCH
REVIEW ARTICLE

Synthesis of Citrus polymethoxyflavonoids and their


antiproliferative activities on Hela cells
Van-Son Nguyen1,2 Wei Li1 Yue Li1 Qiuan Wang1
● ● ●

Received: 11 May 2015 / Accepted: 8 March 2017 / Published online: 17 March 2017
© Springer Science+Business Media New York 2017

Abstract A series of polymethoxyflavonoids (3–16) were Introduction


synthesized through dehydrogenation, O-methylation, gly-
coside hydrolysis, bromination, microwave-assisted aro- Polymethoxyflavonoids (PMFs) are a class of natural pro-
matic nucleophilic substitution, dimethyldioxirane ducts, which almost exclusively exist in Citrus species,
oxidation and regioselective demethylation, starting from particularly in the peel of sweet orange [Citrus sinensis (L.)
abundant and inexpensive natural sources naringin and Osbeck] and mandarin (Citrus reticulata Blanco) (Lewin
hesperidin. All the synthetic compounds were test for et al. 2010). Recently, quite a few studies focused on the
antiproliferative activities on human cervical carcinoma PMFs in Citrus plants because they were found to possess
Hela cell line by the standard CCK-8 assay, the result distinguished anticarcinogenic, anti-inflammatory, and
showed that most of the target compounds exhibited mod- antiviral activities (Miyata et al. 2013; Li et al. 2012; Chen
erate to potent antiproliferative activities on Hela cells et al. 2007).
comparable with the positive control cis-Platin. Among Over the past decade, many biological studies have
them, 5-hydroxypolymethoxy flavonoid 13 showed the focused on two of the most abundant PMFs in Citrus peels:
strongest activity (IC50 0.791 μM). tangeretin and nobiletin. For example, tangeretin (5,6,7,8,4′-
pentamethoxyflavone, 5) was demonstrated to be an anti-
Graphical Abstract

Keywords Polymethoxyflavonoids Synthesis ● ●

Antiproliferative activity Hela cells



proliferative agent against a variety of tumor types (Buisson
et al. 2007), to induce G1 cell-cycle arrest in human col-
orectal carcinoma and induce colon cancer cells apoptosis
(Pan et al. 2002), it was identified as an effective Multiple
* Qiuan Wang Drug Resistance modulator (Olga et al. 2012). Nobiletin
wangqa@hnu.edu.cn
(5,6,7,8,3′,4′-hexamethoxyflavone, 6), another abundant
1
College of Chemistry and Chemical Engineering, Hunan PMFs in Citrus peel, has been reported to exhibit anti-
University, Changsha 410082, China proliferative activity on HL-60 cell line (Li et al. 2007), to
2
Technology Faculty of Thanh Hoa Campus, Industrial University inhibit tyrosinase activity and to exhibit antimutagenic
of Ho Chi Minh City, Ho Chi Minh City, Vietnam activity (Okuno and Miyazawa 2004). In particular,
1586 Med Chem Res (2017) 26:1585–1592

Scheme 1 Reagents and conditions: a I2/pyridine, reflux; b (700 W); f AlCl3, CH3CN, reflux; g Oxone, acetone, CH2Cl2,
(CH3)2SO4, K2CO3, acetone, reflux; c concentrated H2SO4, ethanol, NaHCO3, Na2CO3, 5 °C; h prenyl bromide, K2CO3, acetone, r.t
reflux; d NBS, TFA, r.t; e CH3ONa, CuBr, DMF, microwave heating

increasing attention has been paid to its antitumor metastatic and commercially low-cost naringin or hesperidin by
activity due to the inhibition of gene expression and pro- dehydrogenation with I2/Py followed by base-induced
duction of some matrix metalloproteinases (MMP-1, -3, and elimination to rhoifolin or diosmin, respectively, then
-9) (Oshitari et al. 2011). O-methylation of phenol groups and acid hydrolysis of the
PMFs generally present as minor components in the neohesperidose residue.
Citrus plants. The synthesis of these compounds have been Next, our efforts were concentrated on the methoxylation
much less studied, the full potential of this group of com- of the 6,8-positions of 1 or 2 using the Ulmann-type reac-
pounds to be used as drugs or bioactive molecules has not tion. Upon treatment with 2 equiv N-bromosuccinimide
been realized. Recently we has reported the total synthesis (NBS) in trifluoroacetic acid, 1 or 2 was brominated to give
of some PMFs such as nobiletin, tangeretin and so on, as a 6,8-dibromoflavones 17 or 18 in good yield. Bromination at
continuation of our investigation of bioactive flavonoids both C-6 and C-8 were confirmed by 1H nuclear magnetic
and development of new antitumor activity compounds (Cai resonance (NMR) spectroscopy (loss of H-6 and H-8 sig-
et al. 2012; Wang et al. 2010). We reported the new nals of A-ring, unchanged signals for the AA’BB’ or ABX
synthesis of a series of Citrus PMFs. Furthermore, all spin system of the B-ring). The next step was substitution of
synthesized compounds were evaluated for their anti- the bromines at C-6 and C-8 by CuBr-mediated Ulmann-
proliferative activities on human cervical carcinoma Hela type reaction with a large excess amount of sodium meth-
cells by the standard CCK-8 assay. The synthetic pathways oxide, according to Guillaumet reported (Besson et al.
adopted for the synthesis of a series of Citrus PMFs are 1995), methoxylation of 6,8-dibromoflavones 17 or 18 by
illustrated in Scheme 1. heating it at 120 °C in Dimethyl Formamide (DMF)-MeOH
with MeONa (10 equiv) and CuBr (0.03 equiv) for 22 h led
to nucleophilic aromatic substitution at both C-6 and C-8
Results and discussion giving the expected flavone 3 or 4 only 25% yield in this
procedure, we suspected that decomposition of DMF caused
First, 7-hydroxy-5,4′-dimethoxyflavone (1) and 7-hydroxy- by prolonged heating at 120 °C had affected the yield,
5,3′,4′-trimethoxy flavone (2) were prepared from abundant therefore, the microwave-assisted reaction was tried in an
Med Chem Res (2017) 26:1585–1592 1587

attempt to shorten the reaction time. Gratifyingly, a very Table 1 Half-inhibitory concentration [IC50 (μM)] of compounds
1–18 on the Hela cells
good yield (85–90%) of 6,8-dimethoxy group-substituted
flavones 3 or 4 were obtained under microwave heating Compounds IC50 (μM) Compounds IC50 (μM)
(700 W) within 1 h. For the catalyst CuCl, CuCl2, and CuI
1 81.242 11 6.788
were found to work as well as CuBr to give the same 6,8-
2 59.623 12 27.014
dimethoxylated products. On the other hand, no reaction
3 >100 13 0.791
occurred in the absence of the Cu catalyst. O-methylation of
4 >100 14 18.243
7-hydroxyl group of 3 or 4 with (CH3)2SO4/ K2CO3 in
5 >100 15 97.391
acetone provided the PMFs tangeretin 5 and nobiletin 6,
6 46.185 16 37.946
respectively. The spectroscopic data for compounds 5 and 6
the obtained were identical to those reported previously by 7 >100 17 20.533
us (Cai et al. 2012). Oxidation of flavones to the 3- 8 69.763 18 21.084
hydroxyflavones (flavonols) proved to be challenging. The 9 11.763 cis-Platina 41.253
method was oxidation of polymethoxyflavone 5 or 6 with 10 16.847 Paclitaxela 0.003
dimethyldioxirane generated in situ from Oxone and acet- a
cis-Platin and paclitaxel were employed as positive control
one (Wu et al. 2012; Adam et al. 1987), followed by acid-
induced rearrangement that gave the 3-hydroxy poly-
methoxyflavone 7 or 8 in comparable yield. O-methylation
of 7 or 8 gave PMFs 9 or 10. Then promoted by a lewis acid 5-hydroxypolymethoxy flavonoid 13 showed the strongest
aluminum chloride and with anhydrous acetonitrile as the activity (IC500.791 μM), which suggested that 5-OH was an
solvent, the reaction of compounds 5, 6, 9, and 10 under- important functional group. Recently 5-OH-PMFs have
went regioselective demethylation of the C-5 methoxy gained more attention, as considerable evidence suggests
group to give 5-hydroxypolymethoxyflavone 11–14 in good that 5-OH-PMFs have much stronger health-promoting
yields due to the neighboring group participation of C-4 biological activities than permethoxylated PMFs because of
carbonyl oxygen. Prenyl moiety can potentially substitute the presence of 5-OH (Pan et al. 2007; Lai et al. 2007). The
for conventional lipids as lipophilic carriers of bioactive other four monohydroxylated PMFs, 3 and 4, having a
molecules. The introduction of a prenyl moiety in bioactive hydroxyl group at position 7 did not demonstrate any
molecules is an important post-translational modification antiproliferative activity below 100 μM concentration.
centre to many cellular processes (Michelle et al. 2007). Compounds 7 and 8, having a hydroxyl group at position 3,
Next, 3-hydroxylated polymethoxyflavone 7 and 8 were also showed the lowest activities. This suggested that the
reacted, respectively, with commercially available prenyl position of the hydroxyl group affects the antiproliferative
bromide in the presence of anhydrous potassium carbonate activity on Hela cells of PMFs. Compounds 9, 10 exhibited
and anhydrous acetone at ambient temperature to give O- stronger antiprotiferative activity than 5,6,7,8,4′-penta-
prenyl-substituted PMF derivatives 15 and 16. The structure methoxyflavone (5) and nobiletin (6), with the only struc-
of the synthesized compounds were confirmed by 1H NMR, tural difference between these four compounds being that 9,
13
C NMR, mass spectra (MS), and infrared (IR) spectra. 10 have a methoxyl group at 3-position in the C-ring, while
Antiproliferative activities on human cervical carcinoma 5, 6 have none. Compounds 7, 8, with a hydroxyl group at
Hela cells of these PMFs 3–16, as well as 7-hydroxy-5,4′- position 3, the antiproliferative were poor in comparison to
dimethoxyflavone (1), 7-hydroxy-5,3′,4′-dimethoxyflavone compounds 9, 10, with a methoxyl group at position 3.
(2) and intermediates 17, 18, were evaluated by the CCK-8 These indicates that the methoxyl group at 3-position in the
[2H-tetrazolium-5-(2,4-disulfophenyl)-3-(2-methoxy-4- C-ring contributes to the antiproliferative of PMFs. Our data
nitrophenyl) -2-(4-nitrophenyl)-, inner salt, sodium salt also showed that compound 9, 10, characterized by more
(1:1)] assay. The results are listed in Table 1. The methoxy group were more potent. Compounds 17 and 18,
dose–response curve for CCK-8 assay of compounds 9, 10, which possess two bromo substituents on the 6 and 8-
11, 13, 17and 18 on Hela cells proliferation are shown in position of flavones 1 and 2, were found to confer an
Fig. 1. cis-Platin and paclitaxel, two anticancer drug, were increase in antiproliferative activity. Compounds 15 and 16,
used as the positive control. The results showed that most of having an O-prenyl group at position 3, showed slightly
the target compounds exhibited moderate to potent anti- higher antiproliferative activity than 7 and 8. This showed
proliferative activities on Hela cells. the presence of the lipophilic substituents appears to
Compounds 11, 12, and 13, with a 5-OH in A-ring dis- improve the antiproliferative activity of PMFs by interac-
played higher Hela cells inhibition activity than the com- tion between PMFs and cell membrane.
pounds 5, 6, and 9, where the C-5 position on the A-ring The structure–activity relationship (SAR) analysis
were occupied by methoxy group, among them, revealed that the position of the hydroxyl group of PMFs
1588 Med Chem Res (2017) 26:1585–1592

Fig. 1 The dose–response curve for CCK-8 assay of compound 9, 10, 11, 13, 17, and 18 on Hela cells proliferation

and number of methoxyl group may be closely linked with observations on the antiproliferative activity of Hela cells
their antiproliferative activity against Hela cells, for good have not been reported previously. Although we have lim-
biological effect, the 5-OH or 3-OCH3 functionalities were ited information on the SAR, the findings of this study
essential, while the presence of 3-OH or 7-OH greatly provide important information for the exploitation and uti-
reduced the activity. To the best of our knowledge, such lization of Citrus PMFs as natural anticancer agents.
Med Chem Res (2017) 26:1585–1592 1589

Experimental yellow solid, m.p. 283–284 °C (lit (Shizuo and Hiroaki


1954), 284 °C); 1H NMR (400 MHz, DMSO-d6): δ 10.72 (s,
General methods 1H, 7-OH), 7.58 (d, J = 8.4 Hz, 1H, H-6′), 7.48 (s, 1H, H-
5′), 7.08 (d, J = 8.7 Hz, 1H, H-2′), 6.69 (s, 1H, H-3), 6.56
Melting points were measured on a XRC-I apparatus and (s, 1H, H-8), 6.37 (d, J = 2.5 Hz, 1H, H-6), 3.86, 3.83, 3.79
were uncorrected. Microwave reaction was performed with (each, s, each 3H, –OCH3);13C NMR (100 MHz, DMSO-
XH-MC-1 microwave reactor (Beijing Xianghu Science and d6): δ 178.8, 161.6, 160.6, 160.2, 151.7, 149.2, 124.1,
Technology Development Co., China). 1H NMR and 13C 119.2, 111.2, 108.5, 107.6, 104.2, 101.8, 96.8, 58.8, 56.3,
NMR spectra were recorded on a Bruker AM-400 instru- 56.1; EIMS: m/z 328 [M]+.
ment, using tetramethylsilane as an internal standard che-
mical shifts (δ) in ppm, and coupling constants (J) in Hz. Synthesis of 6,8-dibromo-7-hydroxy-5,4′-
MS or high-resolution mass spectrometry (HRMS) was dimethoxyflavone (17)
determined with VG Autospec-3000 or Mat 95 XP spec-
trometer by the EI method. Column chromatography was To a solution of compound 1 (3 g, 10 mmol) in 35 mL of
carried out on silica gel using 200–300 mesh (Qingdao trifluoroacetic acid (TFA), N-Bromosuccinimide (3.57 g,
Ocean Chemical Products of china). Commercially avail- 20.1 mmol) was added dropwise, and the reaction mixture
able AR or chemical pure reagents, and anhydrous solvent was stirred at room temperature for 5 h, then the mixture
removed water and redistilled were employed. was poured into cold water, the solids was filtered, washed
Rhoifolin and Diosmin were synthesized and purified with water, dried. The crude product was purified by silica
according to previous method by us (Liu et al. 2012; Shan gel column chromatography (petroleum ether/EtOAc, v/v,
et al. 2008). 3-Hydroxytangeretin (3-hydroxy-5,6,7,8,4′- 3:1) to afford 17 (4.3 g, yield: 95%) as white solid, m.p.
pentamethoxyflavone) (7), 3-hydroxynobiletein (3- 197–198 °C; 1H NMR (400 MHz, CDCl3): δ 10.25 (s, 1H,
hydroxy-5,6,7,8,3′,4′-hexamethoxyflavone) (8), 7-OH), 7.93 (d, J = 8.9 Hz, 2H, H-2′ and H-6′), 7.09 (d, J
3,5,6,7,8,4′-hexamethoxyflavone (9), 3,5,6,7,8,3′,4′-hepta- = 8.8 Hz, 1H, H-3′, and H-5′), 6.65 (s, 1H, H-3), 3.87 (d, J
methoxyflavone (10), 5-hydroxy-6,7,8,4′-tetramethoxy- = 3.5 Hz, 3H, 5-OCH3), 3.81 (s, 3H, 4′-OCH3); 13C NMR
flavone (11), 5-hydroxy-6,7,8,3′,4′-pentamethoxy-flavone (100 MHz, CDCl3): δ 176.8, 161.2, 158.4, 156.8, 156.2,
(12), 5-hydroxy-3,6,7,8,4′-pentamethoxyflavone (13) and 5- 131.7, 124.4, 114.4, 112.9, 110.8, 109.2, 106.7, 61.7, 57.3;
hydroxy- 3,6,7,8,3′,4′-hexamethoxyflavone (14) were syn- EIMS: m/z 454 [M]+.
thesized and purified according to previous method by us
(Cai et al. 2012; Li et al. 2014). Synthesis of 6,8-dibromo-7-hydroxy-5, 3′, 4′-
trimethoxyflavone (18)
Synthesis of 7-hydroxy-5,4′-dimethoxyflavone (1)
According to a similar procedure as described for the pre-
The solution of rhoifolin (10 g, 17.3 mmol) in 7% NaOH (150 paration of 17. 18 was obtained (yield: 95%) as yellow
mL) and (CH3)2SO4 (17 mL, 63.05 mmol) was stirred at room soild, m.p. 229–231 °C;1H NMR (400 MHz, CDCl3): δ 7.67
temperature for 6 h. Then 10 mL of concentrated sulfuric acid (d, J = 8.6 Hz, 1H, H-6′), 7.54 (d, J = 2.3 Hz, 1H, H-2′),
was added dropwise, The reaction mixture was stirred under 6.96 (d, J = 8.6 Hz, 1H, H-5′), 6.62 (s, 1H, H-3), 4.01, 3.98,
reflux for 4 h, then reaction mixture cooled and poured into 3.96 (each, s, each 3H, –OCH3); 13C NMR (100 MHz,
cold water, the solids was filtered, washed with water, dried CDCl3): δ 178.2, 162.3, 160.2, 158.6, 154.7, 149.8, 147.6,
and then crystallized in ethyl acetate to give 1 (4.6 g, yield: 123.8, 118.8, 116.6, 112.3, 109.3, 105.6, 101.1, 60.2, 56.2;
89%) as yellow solid, m.p. 296–297 °C (lit (Shizuo et al. EIMS: m/z 484 [M]+.
1952), 298 °C); 1H NMR (400 MHz, DMSO-d6): δ 7.93 (d, J
= 8.9 Hz, 2H, H-2′ and H-6′), 7.07 (d, J = 8.9 Hz, 2H, H-3′, Synthesis of 7-hydroxy-5,6,8,4′-tetramethoxyflavone (3)
and H-5′), 6.58 (s, 1H, H-3), 6.54 (d, J = 2.0 Hz, 1H, H-8),
6.37 (d, J = 2.0 Hz, 1H, H-6), 3.82, 3.78 (each, s, each 3H, To a solution of compound 17 (500 mg) in 10 mL DMF,
–OCH3); 13C NMR (100 MHz, DMSO-d6): δ 176.1, 162.9, CuBr (28 mg, 0.28 mmol) and NaOCH3 (0.22 g) in 5 mL
162.0, 160.9, 159.9, 159.4, 127.9, 123.3, 114.7, 107.3, 106.7, CH3OH were added under nitrogen protection. The reaction
96.8, 95.5, 56.1, 55.7; EIMS: m/z 298 [M]+. mixture was stirred under microwave heating (700 W) for
1 h. Then the reaction mixture was poured into ice-water
Synthesis of 7-hydroxy-5,3′, 4′-trimethoxyflavone (2) and adjusted to pH 6 using 2% HCl (aq), and extracted
by ethyl acetate (20 mL × 3), dried over anhydrous
According to a similar procedure as described for the pre- Na2SO4. The solvent was removed, and the residue was
paration of 1. 2 was obtained (87% yield from diosmin) as purified by silica gel column chromatography (petroleum
1590 Med Chem Res (2017) 26:1585–1592

ether / EtOAc, v/v, 5:1) to afford 3 (yield: 62%) as yellow (s, 6H, –OCH3); 13C NMR (CDCl3): δ 177.3, 161.1, 151.9,
solid, m.p. 182–184 °C; 1H NMR (400 MHz, CDCl3): δ 151.5, 149.2, 148.4, 147.7, 144.1, 123.9, 119.9, 114.7,
7.60 (d, J = 8.9 Hz, 2H, H-2′, and H-6′), 6.93 (d, J = 8.9 111.2, 108.5, 106.7, 62.3, 62.0, 61.8, 61.7, 56.1, 55.9; MS
Hz, 2H, H-3′, and H-5′), 6.53 (s, 1H, H-3), 4.01, 3.92, 3.85, (m/z, EI): 402 (M+), 387 (100), 371, 344, 326, 298, 283,
3.82 (each, s, each 3H, –OCH3); 13C NMR (100 MHz, 239, 225, 198, 197, 182, 162, 153, 139, 91, 83, 77; HRMS
CDCl3): δ 178.8, 162.5, 159.6, 147.3, 146.7, 143.0, 142.3, (EI): m/z [M]+ calcd for C21H22O8: 402.1315, found:
137.1, 136.5, 128.7, 122.8, 114.9, 105.7, 103.6, 61.3, 61.0, 402.1314.
60.2, 55.2; EIMS: m/z 358 [M]+.
Synthesis of 3-O-prenyl-5,6,7,8,4′-pentamethoxyflavonol
Synthesis of 7-hydroxy-5,6,8,3′, 4′-pentamethoxyflavone (15)
(4)
To a mixture of compound 7 (100 mg, 0.26 mmol) and
According to a similar procedure as described for the pre- anhydrous K2CO3 (110 mg, 0.8 mmol) in dry acetone
paration of 3. 4 was obtained (yield: 76%) as yellow soild, (20 mL), a solution of prenyl bromide (0.06 mL,
m.p. 115–117 °C; 1H NMR (400 MHz, CDCl3): δ 7.58 (d, J 0.26 mmol) in acetone (3 mL) was added dropwise upon
= 8.3 Hz, 1H, H-6′), 7.46 (d, J = 7.6 Hz, 1H, H-2′), 7.06 (d. stirring for 3 h, then the reaction mixture was filtered and
J = 7.3 Hz, 1H, H-5′), 6.68 (s, 1H, H-3), 4.12, 4.03, 3.83 evaporated. The residue was subjected to chromatography
(each, s, each 3H, –OCH3), 3.96 (s, 6H, –OCH3); 13C NMR on silica gel with petroleum ether-ethyl acetate (v/v,5:1) as
(100 MHz, CDCl3): δ 177.8, 162.6, 158.6, 149.3, 147.6, eluent to give 114 mg of compound 15 as a yellow soild
146.5, 143.0, 137.3, 136.7, 123.6, 119.6, 116.5, 112.6, (yield: 95%), m.p. 123–125 °C; 1H NMR (400 MHz,
105.9, 103.3, 61.0, 60.3, 60.0, 56.2; EIMS: m/z 388 [M]+. CDCl3): δ 8.19 (d, J = 8.8 Hz, 2H, H-2′ and 6′), 7.03 (d,
J = 8.8 Hz, 2H, H-3′ and 5′), 5.47 (s, 1H, CH), 4.57 (s, 2H,
Synthesis of 5,6,7,8,4′-pentamethoxyflavone (tangeretin) CH2), 4.11, 4.01, 3.98, 3.96, 3.91 (each, s, each 3H,
(5) -OCH3), 1.71 (s, 3H, CH3), 1.66 (s, 3H, CH3); 13C NMR
(100 MHz, CDCl3): δ 173.0, 160.2, 152.8, 150.1, 147.1,
Me2SO4 (0.1 mL, 3 equiv.) was added dropwise to stirred 145.8, 142.7, 138.3, 137.92, 136.8, 129.2. 122.6, 119.1,
solution of 3 (135 mg) in acetone and K2CO3 (0.91 g, 6.48 114.0, 112.8, 98.9, 67.6, 61.3, 61.0, 60.8, 60.6, 54.4, 24.8,
mmol) at 65 °C. The reaction was terminated after 5 h, and 17.1; IR (KBr) v 3417.03, 3234.66, 2029.75, 1635.74,
the mixture was filtered. The solution was evaporated to 1509.74, 1453.52, 1360.70, 1257.00, 1171.23, 1050.70,
afford a solid residue. The crude solid was chromato- 977.21, 801.96, 753.41, 621.55, 481.87 cm−1.
graphed on silica gel using petroleum ether/ethyl acetate (v/
v, 3:1) as eluent to afford light yellow crystals of 5 (115 mg, Synthesis of 3-O-prenyl-5,6,7,8,3′,4′-hexamethoxyflavonol
yield: 86%), m.p. 151–153 °C (lit[12]. 152–153 °C); 1H (16)
NMR (CDCl3): δ 7.80 (d, J = 8.8 Hz, 2H, H-2′ and 6′), 6.94
(d, J = 8.8 Hz, 2H, H-3′ and 5′), 6.52 (s, 1H, H-3), 4.03, Compound 16 was prepared from compound 8 and prenyl
3.94, 3.87, 3.81, (each, s, each 3H, –OCH3); 13C NMR bromide as described for the preparation of compound 15
(CDCl3): δ 176.3, 161.3, 160.1, 150.3, 147.3, 146.7, 143.0, from compound 7. Yellow solid, (yield: 94%), m.p.
137.1, 126.7, 122.8, 113.8, 113.5, 105.6, 61.2, 61.0, 60.8, 140–143 °C; 1H NMR (400 MHz, CDCl3): δ 7.83 (d, J =
60.6, 54.5; IR (KBr): ν 2946, 2843, 1650, 1607, 1587, 2.0 Hz, 1H, H-6′), 7.77 (dd, J = 8.6, 2.1 Hz, 1H, H-5′), 6.92
1512, 1462, 1363, 1265, 1181, 1074, 968, 830 cm–1; MS (d, J = 8.6 Hz, 1H, H-2′), 5.42 (t, J = 7.3 Hz, 1H, CH), 4.52
(m/z, EI): 372 (M+), 327, 315, 287, 259, 194, 135, 77, 57; (d, J = 7.3 Hz, 2H, CH2), 4.02, 3.93, 3.90, 3.88, 3.87 (s,
HRMS (EI): m/z [M]+ calcd for C20H20O7: 372.1209, each, s, each 3H, –OCH3), 1.63 (s, 3H, CH3), 1.56 (s, 3H,
found: 372.1204. CH3); 13C NMR (100 MHz, CDCl3): δ 173.0, 152.4, 150.2,
149.8, 147.5, 147.2, 145.7, 142.7, 138.6, 137.8, 136.8,
Synthesis of nobiletin (5,6,7,8,3′, 4′-hexamethoxyflavone, 122.7, 120.8, 119.3, 114,0, 110.4, 109.7, 67.9, 61.3, 60.9,
6) 60.8, 60.6, 54.9, 54.8, 24.7, 17.1; EIMS: m/z 486 [M]+

According to a similar procedure as described for the pre- Assay for antiproliferative activity
paration of 5. 6 was obtained (yield: 80%) as yellow soild;
m.p. 112–113 °C (lit (Wang et al. 2010), 112–113 °C); 1H The antiproliferative activity was tested using a CCK-8
NMR (CDCl3) δ 7.50 (dd, J = 8.5, 1.9 Hz, 1H, H-6′), 7.34 assay on Human cervix carcinoma cell line (Hela) (Xuan
(s, 1H, H-2′), 6.92 (d, J = 8.5 Hz, 1H, H-5′), 6.55 (s, 1H, H- et al. 2015). Briefly, cells (5 × 103 per well in a 96-well
3), 4.03, 3.96, 3.91, 3.89 (each, s, each 3H, –OCH3), 3.88 plate) were treated with different concentrations of
Med Chem Res (2017) 26:1585–1592 1591

compounds 1–18 (100, 25, 6.25, 1.56, 0.39, 0.0976, 0.0244, Chen KH, Weng MS, Lin JK (2007) Tangeretin suppresses IL-1β-
induced cyclooxygenase (COX)-2 expression through inhibition
0.0061 µM) for 48 h. Then 5% CCK-8-solution was added of p38 MAPK, JNK, and AKT activation in human lung carci-
into each well and incubated with 90% humidity and 5% noma cells. Biochem Pharmacol 73:215–227
CO2 for another 1–3 h. Color development was quantified Lai SC, Li S, Chai CY, Lo CY, Ho CT, Wang YJ, Pan MH (2007)
photometrically at 450 nm, and used an EL × 808 (Bio-Rad Inhibitory effect of citrus 5-hydroxy-3,6,7,8,3’,4’-hexamethoxy-
flavone on 12-O-tetradecanoylphorbol 13-acetate-induced skin
680) absorbance microplate reader to determine the con-
inflammation and tumor promotion in mice. Carcinogenesis 28
centration that killed 50% of cells (IC50). To stop the color (12):2581–2588
reaction by add 10 µL of 1% sodium dodecyl sulfate (SDS) Lewin G, Maciuk A, Thoret S, Aubert G, Dubois J, Cresteil T (2010)
[(dissolve 0.1 g SDS with phosphate buffer saline (PBS) Semisynthesis of natural flavones inhibiting tubulin polymeriza-
tion from hesperidin. J Nat Prod 73(4):702–706
buffer to prepare 10 mL solution)] or add 10 µL of 0.1 mol/
Li L, Xu XY, Cheng D, Yao XL, Pan SY (2012) Structure-activity
L acid such as hydrochloric acid. relationship of Citrus polymethoxylated flavones and their inhi-
bitory effects on Aspergillus niger. J Agric Food Chem 60
(17):4336–4341
Li S, Pan MH, Lai CS, Lo CY, Dushenkov S, Ho CT (2007) Isolation
Conclusion and syntheses of polymethoxyflavones and hydroxylated poly-
methoxyflavones as inhibitors of HL-60 cell lines. Bioorg Med
In summary, we succeeded in developing a new synthetic Chem 15:3381–3389
route for PMFs from abundant and commercially low-cost Li Y, Cai SL, He KL, Wang QA (2014) Semisynthesis of poly-
methoxyflavonoids from naringin and hesperidin. J Chem Res
naringin or hesperidin with CuBr-catalysted and 38:287–290
microwave-assisted aromatic nucleophilic substitution of Liu JD, Chen L, Cai SL, Wang QA (2012) Semisynthesis of apigenin
bromide by methoxy group as the key step. Their anti- and acacetin-7-O-β-D-glycosides from naringin and their cyto-
proliferative activities on human cervical carcinoma Hela toxic activity. Carbohydrate Res 357:41–45
Michelle K, Clark SA, Scott J, Wojtkowiak R, Chirco P, Mathieu J,
cell line were evaluated by the standard CCK-8 assay, the Reiners J, Raymond R, Mattingly RF, Borch R, Gibbs A (2007)
result showed that most of the target compounds exhibited Synthesis, biochemical, and cellular evaluation of farnesyl
moderate to potent antiproliferative activities on Hela cells monophosphate prodrugs as farnesyltransferase inhibitors. J Med
comparable with the positive control cis-Platin. Among Chem 50:3274–3282
Miyata Y, Oshitari T, Okuyama Y, Shimada A, Takahashi H, Natsu-
them, 5-hydroxy PMF 13 showed the strongest activity gari H, Kosano H (2013) Polymethoxyflavones as agents that
(IC50 0.791 μM). The result indicated that this compound is prevent formation of cataract: nobiletin congeners show potent
a potential antitumor agent and is promising for further growth inhibitory effects in human lens epithelial cells. Bioorg
development. Med Chem Lett 23:183–187
Okuno Y, Miyazawa M (2004) Biotransformation of nobiletin by
Aspergillus niger and the antimutagenic activity of a metabolite,
Acknowledgements We thank the National “Twelfth Five-year” plan 4’-hydroxy-5,6,7,8,3’-pentamethoxyflavone. J Nat Prod
for Science & Technology Support (NO. 2012BAD31B02) and the 67:1876–1878
Hunan Provincial Natural Science Foundation of China (NO. Olga W, Jerzy W, Kamila SP, Aleksandra BP, Maria P, Damuta D,
14JJ2048) for financial support. Noelia D, Ferreira MJU, Krystyna M (2012) Multidrug resistance
reversal and apoptosis induction in Human colon cancer cells by
Compliance with ethical standards some flavonoids present in Citrus plants. J Nat Prod
75:1896–1902
Conflict of interest The authors declare that they have no competing Oshitari T, Okuyama Y, Miyata Y, Kosano H, Takahashi H, Natsugari
interest. H (2011) B-Ring-modified and/or 5-demethylated nobiletin
congeners: Inhibitory activity against proMMP-9 production.
Bioorg Med Chem 19:7085–7092
Pan MH, Chen WJ, Lin-Shiau SY, Ho CT, Lin JK (2002) Tangeretin
References induces cell-cycle G1 arrest through inhibiting cyclin-dependent
kinases 2 and 4 activities as well as elevating Cdk inhibitors p21
Adam W, Chan YY, Cremer D, Gauss J, Scheutzow D, Schindler M and p27 in human colorectal carcinoma cells. Carcinogenesis
(1987) Spectral and chemical properties of dimethyldioxirane as 23:1677–1684
determined by experiment and ab initio calculations. J Org Chem Pan MH, Lai YS, Lai CS, Wang Li YS, Lo CY, Dushenkov S, Ho CT
52(13):2800–2803 (2007) 5-Hydroxy-3,6,7,8,3’,4’-hexamethoxyflavone induces
Buisson D, Quintin J, Lewin G (2007) Biotransformation of poly- apoptosis through reactive oxygen species production, growth
methoxylated flavonoids: access to their 4‘-O-demethylated arrest and DNA damage-inducible gene 153 expression, and
metabolites. J Nat Prod 70:1035–1038 caspase activation in human leukemia cells. J Agric Food Chem
Besson T, Ruiz N, Coudert G, Guillaumet G (1995) Synthesis of a 55:5081–5091
benzodioxinic analog of 8-methoxypsoralen. Tetrahedron Shan Y, Li GY, Wang QA, Li ZH (2008) Semisynthesis of five
51:3197–3204 bioactive flavonoids from hesperidin. Chin J Org Chem 28
Cai SL, Liu S, Liu L, Wang QA (2012) Synthesis of bioactive natural (6):1024–1028
polymethoxyflavones and their vinyl ether derivatives. Chem Res Shizuo H, Hiroaki M (1954) Glucoluteolin isolated from the leaves of
Chinese Universities 28:631–636 Sophora angustifolia. J Am Chem Soc 76:5792–5792
1592 Med Chem Res (2017) 26:1585–1592

Shizuo H, Masami S, Mototake K (1952) Studies on flavanone gly- Wu Z, Cai SL, Fan WJ, Wang QA (2012) Semisynthesis of bioactive
cosides. IV. The glycosides of ripe fruit peel and flower petals of flavonols and aurones from naringin. Chin J Org Chem
Citrus Aurantium L. J Am Chem Soc 74:3614–3615 32:1296–1301
Wang QA, Wu Z, Liu L, Zou LH, Luo M (2010) Synthesis of Citrus Xuan LN, Wang P, Zhang K, Shi YP, Liu YM, Zhu T, Chen BQ
bioactive polymethoxyflavonoids and flavonoid glucosides. Chin (2015) Synthesis and in vitro antiproliferative activity of novel
J Org Chem 30(11):1682–1688 benzisoselenazolone derivatives. Med Chem Res 24:543–552

You might also like