Abstract
DNA topoisomerase IB (TOP1) regulates DNA topological structure in many cellular metabolic processes and is a validated target for development of antitumor agents. Our previous study revealed that the benzophenanthridone scaffold is a novel chemotype for the discovery of TOP1 inhibitors. In this work, a series of novel 5-aminoethyl substituted benzophenanthridone derivatives have been synthesized and evaluated for TOP1 inhibition and cytotoxicity. Compound 12 exhibits the most potent TOP1 inhibition (+++) and cytotoxicity in human cancer cell lines with GI50 values at nanomolar concentration range. 12 induces the cellular TOP1cc formation and DNA damage, resulting in HCT116 cell apoptosis. The pharmacokinetics, acute toxicity and antitumor efficiency in vivo of 12 were also studied.
Keywords: Topoisomerase, Inhibitor, DNA damage, Cytotoxicity, Antitumor, Benzophenanthridone
1. Introduction
DNA topoisomerase IB (TOP1) is an essential nuclear enzyme that regulates DNA topological structure in many essential cellular metabolic processes including transcription and replication [1–3]. To perform its functions, TOP1 cleaves one strand of the DNA through a transesterification reaction by nucleophilic attack of its catalytic tyrosine (Tyr723 for human TOP1) to the DNA phosphodiester backbone to form the enzyme-DNA covalent cleavage complexes (TOP1cc). TOP1cc are transient intermediates under normal physiological circumstances as they reverse into the intact DNA and release TOP1 [4,5]. TOP1 inhibitor, such as camptothecin (CPT, Fig. 1) can bind at the interface of TOP1cc [6–8], which stabilizes TOP1cc and prevents further religation of the nicked DNA, resulting in DNA damage and triggering cell death [4,5]. Therefore, TOP1 is a validated target for the discovery of anticancer agents [4,6,9,10].
Fig. 1.
Structures of the camptothecins in clinical uses, indenoisoquinolines in clinical trials and the oxynitidine TOP1 inhibitors found in our laboratory.
To date, four well-known camptothecin derivatives (Fig. 1) have been approved for clinical treatment of cancers, including Topotecan and Irinotecan approved by FDA [11–13], 10-hydroxycamptothecin (HCPT, in China) and Belotecan (in South Korea) [4,14–16]. In spite of their effectiveness in solid tumors, camptothecin TOP1 inhibitors suffer from many shortcomings, such as chemical instability under physiological condition, poor solubility, bone marrow dose-limiting toxicity, severe gastrointestinal toxicity for Irinotecan and drug efflux-mediated resistance [9,17]. Therefore, many investigations have focused on the discovery of non-camptothecin TOP1 inhibitors. And three chemotypes of non-camptothecin TOP1 inhibitors, including indolocarbazole, dibenzonaphthyridinone and indenoisoquinoline derivatives, have been developed [9,18,19]. Three indenoisoquinolines LMP400L, MP744 and LMP776 (Fig. 1) are in clinical trials [19].
Natural products are important sources for medicinal chemistry and drug development. To find novel non-camptothecin TOP1 inhibitors, we studied our in-house natural product library and found several chemotypes inhibiting TOP1, including oxynitidine and meroterpenoid derivatives. Further investigation indicated that several meroterpenoid derivatives showed high TOP1 relaxing inhibition, but were not able to trap and stabilize TOP1cc and could be classified as TOP1 catalytic inhibitors [20,21]. On the contrary, oxynitidine (1, Fig. 1) exhibited weak TOP1 cleavage inhibitory activity (+/0) [22]. Structural optimization of 1 gave several TOP1 inhibitors with higher inhibitory activity, including NTD-96 (2, Fig. 1). 2 shows increased TOP1 inhibition (+++) and traps TOP1cc at genome binding sites different from CPT [22]. Further investigation indicated that 2 targets to TOP1 in cells, inducing cellular TOP1cc formation and DNA damage, and exhibits good antitumor activity both in vitro and in vivo [22]. Structure-activity relationship (SAR) analysis indicated that [22]: 1) the carbonyl group at the 6-position (benzophenanthridinone derivatives) is important for TOP1 inhibition. Most of the 6-aminoalkyloxy benzophenanthridine derivatives have no TOP1 inhibition at 100 μM concentration and show weak cytotoxicity. Furthermore, the reduced derivatives (dihydrobenzophenanthridine) show weak both TOP1 inhibition and cytotoxicity; 2) substitution of aminoethyl group at the 5-position may increase both TOP1 inhibition and cytotoxicity. It is noteworthy that bigger substituents at the 5-position, for example aminopropyl group did not significantly increase the potency of the drugs as TOP1 inhibitors. To further study the spatial effect of the terminus of the aminoethyl group at 5-position on the TOP1 inhibition, in this work, a series of novel 5-aminoethyl substituted benzophenanthridinone derivatives was synthesized and biologically evaluated.
2. Results and discussion
2.1. Chemistry
The synthesis of the designed 5-aminoethyl substituted benzophenanthridinone derivatives is outlined in Scheme 1. Similar to our previous publication [22], the hydroxy group of the Schiff base 3 prepared from the reaction of 6-bromoveratraldehyde with 2-aminoethanol was protected using methoxymethyl (MOM) group to give the intermediate 4, which was directly used for the next reaction without further purification. The intermediate 6 was obtained in two steps. First, under nickel-based catalysis [23], the cyclization reaction of 4 with 5, prepared through Sonogashira coupling reaction according to the reported method [24], gave a ternary ammonium salt intermediate. In the second step, the resulting ternary ammonium salt intermediate was oxidized by K3Fe(CN)6 to give the intermediate 6. Following the Swern oxidation of the hydroxy group of 6, the cyclization reaction under concentrated hydrochloric acid condition gave the intermediate 8 with simultaneous deprotection of MOM group. The replacement of hydroxy group of 8 with bromine gave the bromide 9 in 91% yield. Following replacement reaction of 9 with NaN3, the Pd/C catalytic reduction reaction under hydrogen atmosphere gave the target amine 11 in 51% yield for the two steps (from 9). 11 reacted with formaldehyde to form a Schiff base, which could be reduced by zinc powder to give the target product 12 in 59% yield. The acylation reaction of 12 with various acyl chloride in dichloromethane gave the target products 13–19 with N-methyl amide group as the terminus of the side chain at the 5-position. Similarly, the acylation of 11 gave the target products 20–25. Finally, we synthesized fifteen benzophenanthridone derivatives with various termini groups of the aminoethyl side chain at the 5-position, including amino group, methylamino group, amide group, sulfonamide group and phosphoamide group. Their structures and purity were assessed through HRMS, 1D and 2D NMR spectra, and HPLC method.
Scheme 1.
The synthesis of the target compounds Reagents and conditions: (a) MeOH, NH2(CH2)2OH, rt. (b) MOMCl, NaH, THF, 0 °C. (c) I) N2, Ni(cod)2, P(o-Tol)3, MeCN, 80 °C; ii) CsOH, K3[Fe(CN)6], MeOH, H2O, 80 °C. (d) (COCl)2, DMSO, TEA, DCM, −60 °C. (e) concd. hydrochloric acid, MeOH, rt. (f) PBr3, TCM, rt. g) NaN3, DMSO, rt. (h) Pd/C, H2, THF, rt. (i) aqueous HCHO, Zn, CH3CO2H, H2O, rt. (j) Acyl chloride, DIPEA, DCM, reflux. (k) Acyl chloride, DIPEA, DCM, rt.
2.2. TOP1 inhibition
The synthesized compounds were tested for TOP1 inhibitory activity through TOP1-mediated cleavage assay using a 3’-[32P]-labeled double-stranded DNA fragment as substrate along with CPT and LMP744, an indenoisoquinoline TOP1 inhibitor [25], as positive controls [26]. All compounds were tested at four concentrations, 0.1, 1.0, 10 and 100 μM. The TOP1-mediated cleavage activity of the compounds was semiquantitatively graded consistent with prior publications based on the number and intensities of the DNA cleavage bands relative to the TOP1 inhibition of CPT at 1 μM concentration [22,25]: 0, no inhibitory activity; +, between 20% and 50% inhibitory activity; ++, between 50% and 75% inhibitory activity; +++, between 75% and 95% inhibitory activity; ++++, equal inhibitory activity to CPT. The TOP1-mediated cleavage activity of the synthesized compounds is summarized in Table 1. Compared to the parent 1, most of the synthesized compounds showed increased TOP1 inhibitory activity except for three compounds 13, 22 and 24, which showed equal activity to 1. Compound 12 showed the most potent TOP1 inhibition of +++, equal to compound 2 [22]. Five compounds 11, 18, 19, 20 and 23 showed moderate TOP1 inhibitory activity (++). Compared with 2, the bigger termini of the side chain seem to decrease of TOP1 inhibitory activity. Representative TOP1-mediated cleavage assay gel is shown in Fig. 2. Compounds 11, 12, 18–20 and 23 exhibited the ability to induce TOP1-mediated cleavage bands in a dose-dependent manner with cleavage sites similar to LMP744 but not to CPT. For example, the cleavage sites 17, 35 and 79 could be induced by 11, 12, 18–20 and 23 but not by CPT, implying that the synthesized benzophenanthridinone derivatives trap TOP1cc at different DNA sequence from CPT.
Table 1.
TOP1 cleavage inhibitory activity and cytotoxicity of the synthesized compounds.
![]() | ||||||
---|---|---|---|---|---|---|
Compd. | R | TOP1 inhibitiona | GI50 ± SD(μM)b |
|||
HCT116 | MCF-7 | DU145 | A549 | |||
CPT | /c | ++++ | 0.009 ± 0.001 | 0.012 ± 0.002 | 0.21 ± 0.069 | 0.099 ± 0.017 |
1 | / | 0/+ | 74 ± 4.4 | 31 ± 4.6 | 64 ± 11 | 39 ± 26 |
2 | NMe2 | +++ | 0.076 ± 0.007 | 0.34 ± 0.098 | 0.018 ± 0.002 | 0.79 ± 0.11 |
11 | NH2 | ++ | 0.29 ± 0.019 | 0.10 ± 0.001 | 0.014 ± 0.001 | 0.98 ± 0.014 |
12 | NHMe | +++ | 0.036 ± 0.003 | 0.090 ± 0.001 | 0.002 ± 0.001 | 0.97 ± 0.001 |
13 | ![]() |
0/+ | 86 ± 2.5 | 35 ± 1.7 | 65 ± 1.2 | 42 ± 0.98 |
14 | ![]() |
+ | 7.9 ± 0.15 | 5.8 ± 0.89 | 10 ± 1.2 | 15 ± 1.5 |
15 | ![]() |
+ | 0.96 ± 0.04 | 1.5 ± 0.15 | 15 ± 0.59 | 7.9 ± 0.23 |
16 | ![]() |
+ | 2.5 ± 0.18 | 3.8 ± 0.25 | 3.1 ± 0.30 | 5.3 ± 0.41 |
17 | ![]() |
+ | 40 ± 1.2 | 64 ± 0.57 | 48 ± 1.2 | 35 ± 2.5 |
18 | ![]() |
++ | 9.2 ± 0.14 | 10 ± 0.54 | 5.3 ± 0.21 | 14 ± 1.4 |
19 | ![]() |
++ | 5.9 ± 1.2 | 1.8 ± 0.47 | 3.6 ± 0.25 | 1.9 ± 0.034 |
20 | ![]() |
++ | 5.2 ± 0.13 | 3.0 ± 0.63 | 1.3 ± 0.32 | 4.3 ± 0.21 |
21 | ![]() |
+ | 30 ± 2.1 | 21 ± 1.0 | 45 ± 1.2 | 35 ± 0.14 |
22 | ![]() |
0/+ | 34 ± 1.1 | 38 ± 0.78 | 79 ± 1.0 | 61 ± 0.15 |
23 | ![]() |
++ | 1.1 ± 0.047 | 0.89 ± 0.12 | 1.6 ± 0.021 | 2.9 ± 0.027 |
24 | ![]() |
0/+ | 51 ± 1.5 | 48 ± 1.9 | 39 ± 2.2 | 41 ± 2.0 |
25 | ![]() |
+ | 5.3 ± 0.11 | 2.2 ± 0.27 | 7.8 ± 0.52 | 1.3 ± 0.006 |
TOP1 cleavage inhibitory activity of the synthesized compounds was semiquantitatively graded relative to CPT at 1 μM concentration: 0, no inhibition; +, between 20% and 50% activity; ++, between 50% and 75% activity; +++, between 75% and 95% activity; ++++, equal activity.
GI50 values (means ± SD) were calculated from MTT assay, and defined as the concentrations of compounds that resulted in 50% cell growth inhibition. The experiments were repeated at least three times independently.
“/” means “inapplicable”.
Fig. 2.
Representative gel of TOP1-mediated DNA cleavage assay. Lane 1, DNA alone; lane 2, DNA and TOP1; lane 3, DNA and TOP1 with CPT (1.0 μM); lane 4, DNA and TOP1 with LMP744 (1.0 μM); lanes 5–28, DNA and TOP1 with the indicated compounds at 0.1, 1.0, 10 and 100 μM concentration, respectively. The arrows at left indicate the cleavage site positions.
To inspect the molecular binding mode of the synthesized TOP1 inhibitors within the TOP1-DNA complex, molecular modeling was performed. A hypothetical binding model was built using in-silico docking from the X-ray crystal of the TOP1-DNA-ligand ternary complex (PDB ID: 1K4T) [7]. Compounds were energy-minimized and docked into the binding model. As shown in Fig. 3A, the benzophenanthridinone scaffold of 12 intercalates in the DNA break made by TOP1 and readily stacks with the +1 and −1 base pairs flanking the DNA cleavage site, similar to that of 2 [22]. The A- and B-ring of 12 stack with the bases of non-cleaved strand (C and A), while the C- and D-ring stack with the scissile strand bases (G and T). In addition, the methylaminoethyl side chain of 12 extends into the minor groove of the DNA and binds to a limited space (Fig. 3B), which might be the reason why the bigger termini of the side chain decrease TOP1 inhibitory activity. Also, a hydrogen bond (2.9 Å) was observed between the lactam oxygen atom and R364 residue (Fig. 3A), implying the importance of hydrogen bond acceptor, which is consistent with the cytotoxicity of 12 against the prostate cancer cells DU145-RC0.1, resistant cells with a R364H mutation of TOP1 [27]. DU145-RC0.1 cells showed high resistance to 12 (Table 3). In addition, a hydrogen bond observed between the oxygen atom in the dioxole ring and Asn722 (3.5 Å) might also contribute to the TOP1cc inhibition.
Fig. 3.
Hypothetical binding mode of 12 in the ternary TOP1-DNA-drug complex (PDB ID: 1K4T). 12 is shown as green carbon atoms ball and stick representation. (A) TOP1 is shown in cartoon and the base pairs are displayed in blue lines. (B) TOP1 is shown in surface and DNA is shown in cartoon. All distances are measured from heavy atom to heavy atom. (For interpretation of the references to colour in this figure legend, the reader is referred to the Web version of this article.)
Table 3.
The cytotoxicity of the compound 12 in drug-resistant human cancer cell lines.
Compd. | GI50 ± SD(μM)a |
Resisitance Ratiob | |
---|---|---|---|
Parental cell line | Resistant subline | ||
HCT116 | HCT116-siTOP1 | ||
CPT | 0.009 ± 0.001 | 0.075 ± 0.014 | 8.3 |
12 | 0.036 ± 0.003 | 0.32 ± 0.035 | 8.9 |
DU145 | DU145-RC0.1 | ||
CPT | 0.021 ± 0.016 | 4.73 ± 0.68 | 225 |
12 | 0.002 ± 0.001 | 0.43 ± 0.006 | 215 |
MCF-7 | MCF-7/ADR | ||
DOX | 0.15 ± 0.003 | 11.67 ± 1.94 | 77.8 |
12 | 0.10 ± 0.001 | 1.37 ± 0.55 | 13.7 |
GI50 values (means ± SD) were calculated from MTT assay and defined as the concentrations of compounds that resulted in 50% cell growth inhibition after 72 h of drug exposure. The experiments were repeated at least three times independently.
Resistance ratio was calculated by dividing the GI50 of the mutant cell line by the GI50 of the corresponding parental cell line.
2.3. Cytotoxicity
The cytotoxicity of the synthesized benzophenanthridone derivatives was evaluated through MTT assay against four human tumor cell lines: colon cancer HCT116, breast cancer MCF-7, prostate cancer DU-145 and non-small cell lung cancer A549 cell lines. The compounds were incubated with cells for 72 h in a five-dose assay ranging from 0.01 to 100 μM concentration. At the end of the incubation, MTT solution was added to test the percentage growth of tumor cells. The GI50 values, defined as the concentrations of the compounds that resulted in 50% cell growth inhibition, are calculated and summarized in Table 1.
With the increased TOP1 inhibitory activity (+, ++ and +++), the novel synthesized benzophenanthridone analogues 11, 12, 14–21, 23 and 25 exhibited increased cytotoxicity against these four tumor cell lines compared with the parent 1 with TOP1 inhibition of +/0, except for 17, which exhibited decreased cytotoxicity against MCF-7 cells.12 with the most potent TOP1 inhibition of +++ showed the highest cytotoxicity against HCT116 (GI50 = 0.036 μM), MCF-7 (GI50 = 0.090 μM), DU145 (GI50 = 0.002 μM) and A549 (GI50 = 0.97 μM) cell lines. Although 12 has the similar structure and equal TOP1 inhibition to 2, it showed higher cytotoxicity against HCT116, MCF-7 and DU145 cells, which might possibly due to its good solubility and cellular permeability. Indeed, 12 has higher bioavailability (20.4%, Table 4) in vivo than 2 (15.5%) [22]. Furthermore, 12 showed highest cytotoxicity against DU145 cells at low nanomolar concentration (0.002 μM). With the bigger steric terminus of the side chain at the 5-position, the acylated analogues 13–25 showed decreased both TOP1 inhibitory activity and cytotoxicity compared 12, which is consistent with molecular modelling analysis.
Table 4.
Pharmacokinetic parameters of 12.
parameters | Mean ± SD (n = 2) |
|
---|---|---|
iv (1 mg/kg)a | ig (5 mg/kg)b | |
Tmax (h) | / | 2 ± 0 |
Cmax (ng/ml) | / | 19 ± 9 |
AUC0→t (h·ng/ml) | 47.2 ± 1.8 | 48.3 ± 28 |
AUC0→∞ (h·ng/ml) | 49.8 ± 1.5 | / |
MRTINF (h) | 1.46 ± 0.027 | / |
T1/2 (h) | 1.21 ± 0.14 | / |
F (%) | / | 20.4 ± 11.9 |
iv means intravenous injection.
ig means intragastrical administration.
Compounds 11 and 12 were submitted to the National Cancer Institute (NCI, USA) for further study on cytotoxicity against the 60 cancer cell lines representation nine tissue types (NCI-60) [28–30]. According to the NCI established procedures, the cells were incubated with 11 or 12 for 48 h and stained with sulforhodamine B dye. The GI50 values were plotted and summarized in Table 2. The results indicate that 12 has a higher mean graph midpoint (MGM) for growth inhibition of all cancer cell lines of 0.0977 μM than that of 11 (0.525 μM) and 2 (0.145 μM) [22]. 12 shows high cytotoxicity against 28 cancer cell lines at nanomolar range (<100 nM) and the most cytotoxic against leukemia SR with GI50 of 0.0173 μM.
Table 2.
Cytotoxicity of 11 and 12 against individual NCI-60 cell lines.
Panel | Cell line | GI50 (μM)a |
Panel | Cell line | GI50 (μM) |
||
---|---|---|---|---|---|---|---|
11 | 12 | 11 | 12 | ||||
MGMb | 0.525 | 0.0977 | Colon Cancer | COLO 205 | 0.29 | 0.0605 | |
Leukemia | CCRF-CEM | 0.15 | 0.0201 | HCC-2998 | 1.02 | 0.156 | |
K-562 | 0.161 | 0.0394 | HCT116 | 0.285 | 0.0515 | ||
MOLT-4 | 0.0633 | 0.0234 | HCT-15 | 0.42 | 0.116 | ||
RPMI-8226 | 0.257 | 0.0763 | HT29 | 0.168 | 0.0508 | ||
SR | 0.0907 | 0.0173 | KM12 | 1.1 | 0.238 | ||
Non-Small Cell Lung Cancer | A549/ATCC | 0.584 | 0.0948 | SW-620 | 0.243 | 0.0658 | |
EKVX | 1.36 | 0.202 | Renal Cancer | 786–0 | 0.464 | 0.0703 | |
HOP-62 | 0.399 | 0.0439 | A498 | 1.23 | 0.127 | ||
HOP-92 | 3.56 | 0.184 | ACHN | 0.366 | 0.0856 | ||
NCI-H226 | 1.34 | 0.172 | CAKI-1 | 0.295 | 0.0644 | ||
NCI-H23 | 0.451 | 0.0906 | RXF 393 | 0.929 | 0.16 | ||
NCI-H322M | 0.99 | 0.147 | SN 12C | 0.739 | 0.171 | ||
NCI-H460 | 0.23 | 0.0212 | TK-10 | 1.35 | 0.316 | ||
NCI-H522 | 0.0875 | 0.0274 | UO-31 | 0.35 | 0.104 | ||
CNS Cancer | SF-268 | 0.442 | 0.0903 | Breast Cancer | MCF-7 | 0.164 | 0.0292 |
SF-295 | 0.279 | 0.0346 | MDA-MB-231/ATCC | 1.28 | 0.362 | ||
SF-539 | 1.06 | 0.124 | HS 578T | 1.96 | 0.904 | ||
SNB-19 | 0.398 | 0.116 | BT-549 | 0.918 | 0.202 | ||
SNB-75 | 0.596 | 0.0885 | T-47D | 0.368 | 0.0757 | ||
U251 | 0.26 | 0.0394 | MDA-MB-468 | 0.157 | 0.0599 | ||
Melanoma | LOX IMVI | 0.246 | 0.0389 | Ovarian Cancer | IGROV1 | 0.5 | 0.145 |
MALME-3M | 0.613 | 0.174 | OVCAR-3 | 0.826 | 0.166 | ||
M14 | 0.324 | 0.0581 | OVCAR-4 | 0.782 | 0.154 | ||
MDA-MB-435 | 0.759 | 0.118 | OVCAR-5 | 1.2 | 0.183 | ||
SK-MEL-2 | 3.01 | 0.393 | OVCAR-8 | 0.93 | 0.132 | ||
SK-MEL-28 | 3.05 | 0.573 | NCI/ADR-RES | 1.01 | 0.165 | ||
SK-MEL-5 | 0.614 | 0.0909 | SK-OV-3 | 1.24 | 0.157 | ||
UACC-257 | 1.9 | 0.122 | Prostate Cancer | PC-3 | 0.532 | 0.112 | |
UACC-62 | 0.278 | 0.0451 | DU145 | 0.527 | 0.13 |
GI50 values were defined as the compound concentrations that resulted in 50% cell growth inhibition. The cells were incubated for two days with the tested compounds.
MGM: mean graph midpoint for growth inhibition of all human cancer cell lines.
The cytotoxicity of 12 was further assessed against a panel of isogenic CPT- and doxorubicin-resistant cell lines through MTT assay. The HCT116-siTOP1 subline was established by transfection of colon cancer parental cells HCT116 with short hairpin RNA vectors expressing siRNA for TOP1 [31]. TOP1 is the only known cellular target of CPT [9,32]. HCT116-siTOP1 subline exhibits 8.3-fold resistant to CPT compared to the parental HCT116 cells (Table 3). Similarly, HCT116-siTOP1 subline exhibits 8.9-fold resistant to 12, implying that TOP1 is a major cellular target of 12. The CPT-resistant prostate cancer DU145-RC0.1 cells have a R364H mutation in TOP1 relative to the wild-type parental DU145 cells [27]. The TOP1 with R364H mutation is catalytically active but leads DU145-RC0.1 cells resistance to CPT because the R364 residue is close to the catalytic tyrosine and can stabilize the open form of TOP1cc [7,8]. Compared to the parental DU145 cells, the DU145-RC0.1 cells show 225-fold resistant to CPT and 215-fold resistant to 12 (Table 3), which is consistent with the molecular modeling result (Fig. 3) showing a hydrogen bond between 12 and R364 residue. P-Glycoprotein (P-gp) mediated drug efflux is generally responsible for classical multiple drug resistance [33]. The chemotherapeutic agent doxorubincin (DOX) is a substrate of P-gp. Compared to the parental MCF-7 cells, the breast cancer MCF-7/ ADR cells overexpressing P-gp are highly resistant to DOX (77.8-fold) and less resistant to 12 (13.7-fold, Table 3) [34], implying 12 might not be a substrate of P-gp. These results indicate that 12 acts as TOP1 inhibitor in cancer cells, similar to its analogue 2 [22].
2.4. Induction of cellular TOP1cc and DNA damage
To assess the induction of TOP1cc by 12, the immunocomplex of enzyme to DNA (ICE) assay in HCT116 cells was performed. As shown Fig. 4A, both the positive control CPT and 12 induce the formation of cellular TOP1cc in a dose-dependent manner. And 12 shows higher ability to induce cellular TOP1cc at 25 μM than CPT.
Fig. 4.
(A) Detection of TOP1-DNA covalent cleavage complexes by in vivo complex of enzyme (ICE) assay in human colon cancer HCT116 cells. Left: lane 1, control; lanes 2 and 3, cells treated with CPT at 25 and 50 μM concentration, respectively. Right: lanes 1–3, cells treated with 12 at 25, 50 and 100 μM concentration, respectively. (B) Histone γH2AX foci induced by 12 in HCT116 cells. Cells were treated with CPT or 12 at 1 μM concentration for 3 h. DNA was stained with DAPI (blue). (For interpretation of the references to colour in this figure legend, the reader is referred to the Web version of this article.)
To evaluate the DNA damaging effect of 12 in cancer cells, γH2AX foci were assessed by immunofluorescence microscopy in human prostate cancer HCT116 cells. After incubation with drugs for 3 h, HCT116 cells were stained with γH2AX antibodies. As shown in Fig. 4B, similar to CPT,12 effectively induces γH2AX foci at 1 μM concentration, implying that DNA damage is induced mainly due to the trapping of cellular TOP1cc by 12.
2.5. Cancer cell apoptosis
To evaluate the induction of apoptosis by 12, flow cytometry assays were performed in HCT116 cells. HCT116 cells were incubated with 12 for 24 h and detected for the apoptotic cells. As shown in Fig. 5, 12 could obviously induce HCT116 cells apoptosis in a dose-dependent manner. 27.95% early apoptotic cells were observed after incubation with 12 at 2 μM.
Fig. 5.
Flow cytometry histograms. HCT116 cells were incubated with 12 for 24 h at 0.5 μM, 1 μM and 2 μM concentration, respectively
2.6. Pharmacokinetic parameters
The pharmacokinetic (PK) study in vivo of 12 was performed in Sprague-Dawley (SD) rats. The SD rats were randomly divided into two groups (n = 2) and treated by intravenous injection (iv) at 1 mg/kg dose and intragastrical administration (ig) at 5 mg/kg dose, respectively. Plasma samples were collected postdosing and the concentration of 12 was measured. The PK parameters were calculated and summarized in Table 4. After iv treatment, the AUC0→t is 47.2 ± 1.8 h ng/ml and T1/2 is 1.21 ± 0.14 h. After ig treatment, Tmax is 2 ± 0 h and Cmax is 19 ± 9 ng/ml. The bioavailability (F, 20.4%) of 12 is higher than that of the analogue 2 (15.5%), which might be the reason why 12 shows higher cytotoxicity in vitro and antitumor efficiency in vivo (Fig. 7) [22].
Fig. 7.
Antitumor efficiency of 12 in HCT116 (A) and MCF-7 (B) xenograft models. The effects of 12 on tumor size (left) and tumor weight (right) at the dose of 5 mg/kg, 10 mg/kg and 20 mg/kg, respectively. Statistically significant difference in mean tumor weight compared with the control, **: P < 0.01, ***: P < 0.001.
2.7. Acute toxicity in vivo
The acute toxicity of 12 was assessed in Kunming male mice. The mice were randomly divided into six groups (n = 6) and treated with 12 by intraperitoneal injection (ip) at single doses of 300, 240, 192, 153.6 and 122.9 mg/kg. The control group was treated with sterile water. As shown in Fig. 6, after 7 days of administration with 12, all mice survived in the group of 122.9 mg/kg dose, and four mice survived in the 153.6 mg/kg dose group, three mice survived in the 192 mg/kg dose group, two mice survived in the 240 mg/kg dose group. All mice died within 5 days in the 300 mg/kg dose group. The median lethal dose (LD50), defined as the dose to kill half of mice after 7 days, is 192 mg/kg.
Fig. 6.
Effect of 12 on mice survival. Mice were treated with 12 at dose 300 mg/kg, 240 mg/kg, 192 mg/kg 153.6 mg/kg and 122.9 mg/kg, respectively.
2.8. Antitumor efficiency in vivo
The in vivo antitumor efficiency of 12 was assessed in both human colon cancer HCT116 and human breast cancer MCF-7 xenograft mude mice models. For both models, the mice were randomly divided into four groups (n = 6) and treated with 12 at 20 mg/kg, 10 mg/kg or 5 mg/kg dose by ip administration daily. The control group was treated with saline. As shown in Fig. 7, administration of 12 significantly reduced the tumor volume in a dose-dependent manner in both HCT116 and MCF-7 xenograft models. 12 is more antitumor efficient in HCT116 xenograft model than MCF-7 xenograft model. The tumor weight inhibitions (TWI) of 12 at 20 μM dose are 76.9% (in HCT116 model) and 71.8% (in MCF-7 model), respectively.
3. Conclusion
In summary, a series of novel 5-aminoethyl substituted benzophenanthridinone derivatives have been synthesized and evaluated for biochemical activity as TOP1 inhibitors and cellular responses against four human cancer cell lines (HCT116, MCF-7, DU145 and A549). The TOP1-mediated cleavage assay indicates that the derivatives with bigger termini of the side chain at the 5-position show decreased TOP1 inhibitory activity. Compound 12 with methylamino ethyl group at the 5-position exhibits the most potent TOP1 inhibition of +++, and cytotoxicity in human cancer cell lines at nanomolar concentration range GI50 values. 12 also shows consistent cytotoxicity in the NCI-60 cell lines with nanomolar MGM value of 0.0977 μM. Cell-based assays indicate that 12 induces the formation of cellular TOP1cc and DNA damage in HCT116 cells, promotes apoptosis, and is not a substrate of P-gp, a drug efflux protein responsible for multidrug resistance. 12 was also evaluated for PK, acute toxicity and antitumor efficiency in vivo. The results indicate that 12 exhibits antitumor efficiency in both HCT116 and MCF-7 xenograft nude mice models, and good bioavailability (20.4%) in rat model, higher than its analogue 2 (15.5%) with a dimethylaminoethyl group at 5-position, which might be the reason why 12 shows more potent antitumor activity both in vitro and in vivo. These results suggest that benzophenanthridinone scaffold is a chemotype for TOP1 inhibitors, and worth further development.
4. Experimental section
4.1. General experiments
All the required chemical reagents used for synthesis were purchased from Sigma-Aldrich, Alfa Aesar or Aladdin Reagent Database Inc (Shanghai) and used without any further purification unless otherwise indicated. Melting points were determined in open capillary tubes on a MPA100 Optimelt Automated Melting Point System without being corrected. Silica gel GF254 thin layer chromatography (TLC) was used to monitor the progress of the chemical reaction. Nuclear magnetic resonance spectra were recorded on a Bruker AVANCE III 400 MHz spectrometer using tetramethylsilane as an internal reference. Mass spectra were analyzed on an Agilent 6120 (Quadrupole LC-MS) mass spectrometer. The high-resolution mass spectra were analyzed on an SHIMADZU LCMS-IT-TOF mass spectrometer. All compounds tested for biological activities were analyzed by HPLC and their purities were more than 95%. The analysis condition is: detection at 220 nm, 1.0 ml/min flow rate, a linear gradient of 50%–15% PBS buffer (pH 3) and 50%–85% MeOH in 35 min.
All animals were obtained from Laboratory Animal Center of Sun Yat-sen University. All procedures were approved by the Animal Ethics Committee of Sun Yat-sen University, in accordance with National Institute of Health and Nutrition Guidelines for the Care and Use of Laboratory Animals.
4.2. 12-(2-bromoethyl)-2,3-dimethoxy-[1,3]dioxolo[4’,5’:4,5]benzo [1,2-c]phenanthridin-13(12H)-one (9)
To a solution of 8 (0.5 mmol) in freshly distilled chloroform (10 mL), PBr3 (1 mL) was added slowly with a syringe. The mixture was stirred at room temperature for 1 h. The resulting gray precipitate was filtered and washed with chloroform, saturated sodium bicarbonate and water, respectively. The crude solid was dried and purified by silica gel column chromatography to give the gray solid 9, yield 91%, 1H NMR (DMSO) δ 8.84 (d, J = 9.1 Hz, 1H), 8.31 (s, 1H), 8.26 (s, 1H), 8.16 (d, J = 9.0 Hz, 1H), 7.73 (s, 1H), 7.69 (s, 1H), 6.33 (s, 2H), 5.76 (t, J = 9.0 Hz, 2H), 5.35 (t, J = 9.0 Hz, 2H), 4.21 (s, 3H), 4.04 (s, 3H). ESI-MS m/z: 455.1 (100%), 457.1 (100%) [M + H]+.
4.3. 12-(2-aminoethyl)-2,3-dimethoxy-[1,3]dioxolo[4’,5’:4,5]benzo [1,2-c]phenanthridin-13(12H)-one (11)
To a solution of 9 (456 mg, 1 mmol) in DMSO (50 mL), NaN3 (130 mg, 2 mmol) was added. The mixture was stirred at room temperature for 16 h. And then, the reactive solution was poured into water (100 mL). The formed gray precipitate was filtered and washed with saturated sodium bicarbonate and water, respectively. The crude solid (10) was dried for the next synthesis without further purification.
The crude solid 10 was dissolved in THF (100 mL). Pd/C (42 mg) was added to the solution. The mixture was stirred at room temperature under hydrogen atmosphere for 2 h. The reaction mixture was filtered and washed with THF. The filtrate was dried over MgSO4 and concentrated under reduced pressure. The residue was purified by silica gel column chromatography to give the white solid 11, yield 51% for the two steps, mp = 264.3–267.1 °C. 1H NMR (CDCl3) δ 7.98 (d, J = 8.4 Hz,1H), 7.91 (s,1H), 7.59–7.54 (m, 2H), 7.52 (s, 1H), 7.18 (s, 1H), 6.10 (s, 2H), 4.60 (t, J = 6.0 Hz, 2H), 4.11 (s, 3H), 4.06 (s, 3H), 3.13 (t, J = 6.0 Hz, 2H). 13C NMR (CDCl3) δ 164.9, 153.8, 149.9, 147.7, 147.6, 135.6, 131.9, 129.1, 123.6, 121.3, 119.6, 118.5, 117.4, 109.0, 105.0, 103.0, 102.4, 101.7, 56.4, 56.3, 55.1, 41.5. HRMS (ESI) m/z: 393.1439 [M + H]+, calcd for C22H21N2O5 393.1445.
4.4. 2,3-Dimethoxy-12-(2-(methylamino)ethyl)-[1,3]dioxolo[4’,5’:4,5]benzo[1,2-c]phenanthridin-13(12H)-one (12)
To a solution of 11 (470 mg, 0.12 mmol) in acetic acid (140 mL) and water (100 mL), aqueous formaldehyde solution (40%, 90 mL) and Zn (156 mg, 2.4 mmol) was added. The mixture was stirred at room temperature overnight. Ammonia water was added to quench the reaction. The solution was exacted with chloroform (3 × 50 mL). The combined organic layer was dried over MgSO4 and concentrated under reduced pressure. The residue was purified by silica gel column chromatography to give the white solid 12, yield 59%, mp = 211.8–212.0 °C. 1H NMR (CDCl3) δ 7.97 (d, J = 8.8 Hz, 1H), 7.90 (s, 1H), 7.59–7.54 (m, 2H), 7.54 (s, 1H), 7.18 (s, 1H), 6.10 (s, 2H), 4.61 (t, J = 6.4 Hz, 2H), 4.10 (s, 3H), 4.05 (s, 3H), 3.11 (t, J = 6.4 Hz, 2H), 2.36 (s, 3H).13C NMR (CDCl3) δ 165.0,153.9,149.9,147.7,147.6,135.6, 131.9,129.2,123.6,121.2,119.6,118.5,117.4,108.9,105.0,103.1,102.4, 101.7, 56.4, 56.3, 52.0, 51.3, 36.2. HRMS (ESI) m/z: 407.1601 [M + H]+,calcd for C23H23N2O5 407.1601.
4.5. General procedure for the synthesis of compounds 13–25
To a solution of 12 (or 11, 0.24 mmol) and DIPEA (2.4 mmol) in freshly distilled dichloromethane (30 mL), the solution of acyl chlorides (0.32 mmol) in dichloromethane (5 mL) was added slowly at an ice bath. And then, the reaction solution was stirred and heated under reflux for 1 h (for 13–19, or at room temperature for 20–25). The reaction solution was cooled to room temperature. The solvent was evaporated under reduced pressure. The residue was purified by silica gel column chromatography to give target product.
4.5.1. N-(2-(2,3-dimethoxy-13-oxo-[1,3]dioxolo[4’,5’:4,5]benzo[1,2-c]phenan-thridin-12(13H)-yl)ethyl)-1,1,1-trifluoro-N-methylmethanesulfonamide (13)
White solid, yield 40%, mp = 217.4–217.8 °C. 1H NMR (CDCl3) δ 7.99 (d, J = 8.8 Hz, 1H), 7.87 (s, 1H), 7.62–7.56 (m, 2H), 7.41 (s, 1H), 7.20 (s, 1H), 6.13 (s, 2H), 4.81 (t, J = 6.6 Hz, 2H), 4.11 (s, 2H), 4.06 (s, 2H), 3.82 (s, 2H), 2.87 (s, 3H). 13C NMR (CDCl3) δ 164.6, 153.9, 149.8, 147.8, 147.7, 134.6, 131.8, 129.2, 123.9, 120.5, 119.2, 118.4, 117.7, 108.6, 105.2, 102.9, 101.7, 101.6, 56.2, 56.2, 49.0, 48.8, 35.9. HRMS (ESI) m/z: 539.1116 [M + H]+, calcd for C24H22N2O7F3S 539.1094.
4.5.2. N-(2-(2,3-dimethoxy-13-oxo-[1,3]dioxolo[4’,5’:4,5]benzo [1,2-c]phenanth-ridin-12(13H)-yl)ethyl)-N-methylcyclopropanesulfonamide (14)
White solid, yield 80%, mp > 280 °C. 1H NMR (CDCl3) δ 7.96 (d, J = 8.5 Hz, 1H), 7.88 (s, 1H), 7.56 (d, J = 6.0 Hz, 2H), 7.52 (s, 1H), 7.17 (s, 1H), 6.11 (s, 2H), 4.74 (t, J = 6.5 Hz, 2H), 4.10 (s, 2H), 4.05 (s, 2H), 3.74 (t, J = 6.6 Hz, 2H), 2.78 (s, 3H), 2.08 (d, J = 4.5 Hz, 1H), 1.07 (d, J = 3.6 Hz, 2H), 0.83 (d, J = 6.2 Hz, 2H). 13C NMR (CDCl3) δ 164.6, 153.8, 149.7, 147.7, 147.7, 135.1, 131.8, 129.2, 123.7, 120.7, 119.2, 118.3, 117.4,108.6,104.9,102.9,102.1,101.7, 56.2, 56.1, 50.2, 48.5, 35.7, 26.9, 4.5, 4.5. HRMS (ESI) m/z: 511.1536 [M + H]+, calcd for C26H27N2O7S 511.1533.
4.5.3. N-(2-(2,3-dimethoxy-13-oxo-[1,3]dioxolo[4’,5’:4,5]benzo [1,2-c]phenan-thridin-12(13H)-yl)ethyl)-N-methyldimethylamine-1-sulfonamide (15)
White solid, yield 83%, mp = 234.7–235.9 °C · 1H NMR (CDCl3) δ 7.97 (d, J = 8.6 Hz, 1H), 7.88 (s, 1H), 7.60–7.49 (m, 3H), 7.18 (s, 1H), 6.12 (s, 2H), 4.74 (t, J = 6.5 Hz, 2H), 4.11 (s, 3H), 4.05 (s, 3H), 3.71 (t, J = 6.5 Hz, 2H), 2.68 (s, 3H), 2.59 (s, 6H). 13C NMR (CDCl3) δ 164.6, 153.8,149.7,147.7,147.6,135.2,131.8,129.2,123.6,120.8,119.3,118.3, 117.4,108.6,104.9,102.8,102.1,101.7, 56.3, 56.2, 49.8, 48.9, 37.8, 37.8, 36.1. HRMS (ESI) m/z: 514.1669 [M+ H]+, calcd for C25H28N3O7S 514.1642.
4.5.4. N-(2-(2,3-dimethoxy-13-oxo-[1,3]dioxolo[4’,5’:4,5]benzo [1,2-c]phena-nthridin-12(13H)-yl)ethyl)-N-methylpyrrolidine-1-sulfonamide (16)
White solid, yield 81%, mp = 223.4–224.3 °C. 1H NMR (CDCl3) δ 7.97 (d, J = 8.7 Hz, 1H), 7.89 (s, 1H), 7.56 (d, J = 6.5 Hz, 2H), 7.54 (s, 1H), 7.18 (s,1H), 6.12 (s, 2H), 4.75 (t, J = 6.6 Hz, 2H), 4.11 (s, 3H), 4.05 (s, 3H), 3.69 (t, J = 6.6 Hz, 2H), 3.03 (t, J = 6.7 Hz, 4H), 2.64 (s, 3H), 1.82–1.75 (m, 4H).13C NMR (CDCl3) δ 164.6,153.8,149.7,147.7,147.6, 135.3,131.8,129.2,123.5,120.9,119.4,118.2,117.4,108.7,104.9,102.9, 102.2, 101.7, 56.3, 56.2, 50.1, 48.8, 47.9, 47.9, 35.9, 25.5, 25.5. HRMS (ESI) m/z: 540.1811 [M + H]+, calcd for C27H30N3O7 540.1799.
4.5.5. N-(2-(2,3-dimethoxy-13-oxo-[1,3]dioxolo[4’,5’:4,5]benzo [1,2-c]phenan-thridin-12(13H)-yl)ethyl)-N-methylmorpholine-4-sulfonamide (17)
White solid, yield 85%, mp = 205.7–206.2 °C. 1H NMR (CDCl3) δ 7.96 (d, J = 8.7 Hz, 1H), 7.88 (s, 1H), 7.56 (t, J = 4.3 Hz, 2H), 7.27 (s, 1H), 7.17 (s,1H), 6.11 (s, 2H), 4.75 (t, J = 6.5 Hz, 2H), 4.11 (s, 3H), 4.06 (s, 3H), 3.69 (t, J =6.5 Hz, 2H), 3.55 (t, J = 4.0 Hz, 4H), 2.91(t, J = 4.0 Hz, 3H), 2.64 (s, 3H). 13C NMR (CDCl3) δ 164.5, 153.8, 149.8, 147.7, 147.7, 135.1, 131.7, 129.1, 123.6, 120.8, 119.3, 118.3, 117.5, 108.6, 104.9, 102.8, 102.1, 101.7, 66.2, 66.2, 56.5, 56.2, 49.9, 48.9, 45.9, 45.9, 36.3. HRMS (ESI) m/z: 556.1778 [M + H]+, calcd for C27H30N3O8S 556.1748.
4.5.6. Dimethyl(2-(2,3-dimethoxy-13-oxo-[1,3]dioxolo[4’,5’:4,5] benzo[1,2-c]phe-nanthridin-12(13H)-yl)ethyl)(methyl) phosphoramidate (18)
White solid, yield 30%, mp = 220.0–221.7 °C. 1H NMR (CDCl3) δ 7.97 (d, J = 8.6 Hz, 1H), 7.90 (s, 1H), 7.57 (s, 1H), 7.54 (d, J = 5.9 Hz, 2H), 7.18 (s,1H), 6.11 (s, 2H), 4.73 (t, J = 6.5 Hz, 2H), 4.10 (s, 3H), 4.06 (s, 3H), 3.55–3.47 (m, 2H), 3.44 (s, 3H), 3.42 (s, 3H), 2.49 (d, J = 9.8 Hz, 3H). 13C NMR (CDCl3) δ 164.5, 153.6, 149.7, 147.6, 147.5, 135.3,131.7,129.1,123.4,120.9,119.4,118.3,117.4,108.8,104.8,102.8, 102.3, 101.6, 56.3, 56.1, 52.7, 52.7, 50.3, 47.4, 34.3. HRMS (ESI) m/z: 515.1584 [M + H]+, calcd for C25H28N2O8P 515.1578.
4.5.7. 2,3-Dimethoxy-12-(2-(methyl(2-oxido-1,3,2-dioxaphospholan-2-yl)amino)ethyl)-[1,3]dioxolo[4’,5’:4,5]benzo [1,2-c]phenanthridin-13(12H)-one (19)
White solid, yield 40%, mp = 221.3–222.4 °C. 1H NMR (CDCl3) δ 7.97 (d, J = 8.8 Hz, 1H), 7.86 (s, 1H), 7.56 (dd, J = 11.7, 6.9 Hz, 2H), 7.51 (s, 1H), 7.18 (s, 1H), 6.12 (s, 2H), 4.74–4.66 (m, 2H), 4.32–4.25 (m, 2H), 4.24–4.19 (m, 2H), 4.11 (s, 3H), 4.05 (s, 3H), 3.53–3.41 (m, 2H), 2.65 (d, J = 10.2 Hz, 2H). 13C NMR (CDCl3) δ 164.6, 153.6, 149.6, 147.6, 147.5, 135.0, 131.7, 129.2, 123.5, 120.7, 119.3, 118.3, 117.5, 108.5, 104.9,102.9,102.0,101.7, 65.7, 65.6, 56.3, 56.1, 49.1, 47.0, 34.5. HRMS (ESI) m/z: 513.1446 [M + H]+, calcd for C25H36N2O8P 513.1421.
4.5.8. N-(2-(2,3-dimethoxy-13-oxo-[1,3]dioxolo[4’,5’:4,5]benzo [1,2-c]phenanthridin-12(13H)-yl)ethyl)cyclopropanesulfonamide (20)
White solid, yield 60%, mp = 245.4–246.2 °C. 1H NMR CDCl3) δ 7.97 (d, J = 8.7 Hz, 1H), 7.86 (s, 1H), 7.57 (d, J = 9.7 Hz, 2H), 7.30 (s, 1H), 7.18 (s, 1H), 6.12 (s, 2H), 5.52 (t, J = 6.1 Hz, 1H), 4.62 (t, J = 5.5 Hz, 2H), 4.11 (s, 3H), 4.05 (s, 3H), 3.78 (dd, J = 11.4, 5.7 Hz, 2H), 2.26 (td, J = 8.0, 4.1 Hz, 1H), 1.14–1.02 (m, 2H), 0.92–0.81 (m, 2H). 13C NMR (CDCl3) δ 165.0, 153.9, 149.8, 147.7, 147.6, 135.1, 131.8, 129.2, 123.7, 120.7, 118.9, 118.4, 117.3, 108.6, 105.1, 102.9, 101.7, 101.7, 56.3, 56.2, 52.6, 43.5, 30.2, 5.4, 5.4. HRMS (ESI) m/z: 497.1394 [M + H]+, calcd for C25H25N2O7S 497.1377.
4.5.9. N-(2-(2,3-dimethoxy-13-oxo-[1,3]dioxolo[4’,5’:4,5]benzo [1,2-c]phenanthridin-12(13H)-yl)ethyl)dimethylamine-1-sulfonamide (21)
White solid, yield 65%, mp > 280 °C. 1H NMR (CDCl3) δ 7.98 (d, J = 8.7 Hz, 1H), 7.86 (s, 1H), 7.57 (d, J = 8.0 Hz, 2H), 7.29 (s, 1H), 7.19 (s, 1H), 6.11 (s, 2H), 5.58 (t, J = 5.9 Hz, 1H), 4.59 (t, J = 5.4 Hz, 2H), 4.11 (s, 3H), 4.06 (s, 3H), 3.70 (t, J = 5.4 Hz, 2H), 2.70 (s, 6H).13C NMR (CDCl3) δ 165.1, 153.9, 149.8, 147.7, 147.6, 135.1, 131.9, 129.2, 123.8, 120.7, 118.9, 118.4, 117.3, 108.6, 105.1, 102.9, 101.7, 101.7, 56.3, 56.2, 52.4, 44.2, 37.9. 37.9. HRMS (ESI) m/z: 500.1531 [M + H]+, calcd for C24H26N3O7S 500.1486.
4.5.10. N-(2-(2,3-dimethoxy-13-oxo-[1,3]dioxolo[4’,5’:4,5]benzo [1,2-c]phenanthridin-12(13H)-yl)ethyl)pyrrolidine-1-sulfonamide (22)
White solid, yield 85%, mp = 249.7–250.8 °C. 1H NMR (CDCl3) δ 7.98 (d, J = 8.7 Hz, 1H), 7.87 (s, 1H), 7.58 (d, J = 7.2 Hz, 2H), 7.31 (s, 1H), 7.19 (s, 1H), 6.12 (s, 2H), 5.41 (t, J = 5.9 Hz, 1H), 4.62 (t, J = 5.4 Hz, 2H), 4.11 (s, 3H), 4.06 (s, 3H), 3.70–3.67 (m, 2H), 3.19 (t, J = 6.6 Hz, 4H), 1.82 (t, J = 6.6 Hz, 4H). 13C NMR (CDCl3) δ 165.0, 153.9,149.8,147.6,147.6,135.2,131.9,129.2,123.7,120.8,119.1,118.4, 117.4,108.7,105.1,102.9,101.8,101.7, 56.3, 56.2, 52.3, 48.0, 48.0, 43.9, 25.5, 25.5.HRMS (ESI) m/z:526.1640 [M + H]+,calcd for C26H28N3O7S 526.1642.
4.5.11. N-(2-(2,3-dimethoxy-13-oxo-[1,3]dioxolo[4’5’:4,5]benzo [1,2-c]phenanthridin-12(13H)-yl)ethyl)morpholine-4-sulfonamide (23)
White solid, yield 70%, mp = 221.6–222.3 °C. 1H NMR (CDCl3) δ 7.98 (d, J = 8.5 Hz,1H), 7.85 (s,1H), 7.58 (d, J = 11.4 Hz, 2H), 7.27 (s, 1H), 7.19 (s, 1H), 6.12 (s, 2H), 5.66 (s, 1H), 4.61 (t, J = 4.8 Hz 2H), 4.11 (s, 3H), 4.06 (s, 3H), 3.68 (t, J = 4.8 Hz, 2H), 3.64 (s, 4H), 3.06 (s, 4H). 13C NMR (CDCl3) δ 165.2,154.0,149.9,147.7,147.7,135.0,131.9,129.2, 123.8, 120.7, 118.9, 118.4, 117.4, 108.6, 105.1, 102.9, 101.7, 101.6, 66.1, 66.1, 56.3, 56.2, 52.3, 46.1, 44.4, 44.4. HRMS (ESI) m/z: 542.1623 [M + H]+, calcd for C26H28N3O8S 542.1592.
4.5.12. Dimethyl(2-(2,3-dimethoxy-13-oxo-[1,3]dioxolo[4’,5’:4,5] benzo[1,2-c]phena-nthridin-12(13H)-yl)ethyl)phosphoramidate (24)
White solid, yield 62%, mp = 197.2–197.8 °C. 1H NMR (CDCl3) δ 7.97 (d, J = 8.7 Hz, 1H), 7.88 (s, 1H), 7.56 (d, J = 7.5 Hz, 2H), 7.37 (s, 1H), 7.19 (s,1H), 6.11 (s, 2H), 4.60 (t, J = 5.8 Hz, 2H), 4.11 (s, 3H), 4.06 (s, 3H), 3.55 (s, 3H), 3.52 (s, 3H), 3.50–3.46 (m, 2H). 13C NMR (CDCl3) δ 164.8, 153.8, 149.8, 147.6, 147.5, 135.4, 131.7, 129.1, 123.6, 120.9, 119.3, 118.4, 117.3, 108.7, 104.9, 102.9, 101.9, 101.6, 56.3, 56.2, 53.6, 52.9, 52.9, 41.1. HRMS (ESI) m/z: 501.1423 [M + H]+, calcd for C24H26N2O8P 501.1421.
4.5.13. N-(2-(2,3-dimethoxy-13-oxo-[1,3]dioxolo[4’,5’:4,5]benzo [1,2-c]phena-nthridin-12(13H)-yl)ethyl)cyclopropanecarboxamide (25)
White solid, yield 80%, mp = 237.6–238.1 °C. 1H NMR (CDCl3) δ 7.97 (d, J = 8.7 Hz, 1H), 7.89 (s, 1H), 7.57 (d, J = 9.7 Hz, 2H), 7.42 (s, 1H), 7.17 (s,1H), 6.73 (s,1H), 6.10 (s, 2H), 4.60 (t, J = 4.9 Hz, 2H), 4.11 (s, 3H), 4.07 (s, 3H), 3.88 (d, J = 5.0 Hz, 2H), 0.87 (d, J = 6.7 Hz, 1H), 0.81 (s, 2H), 0.63 (d, J = 4.7 Hz, 2H). 13C NMR (CDCl3) δ 173.5, 165.2, 153.8, 149.7, 147.7, 147.6, 135.4, 131.8, 129.3, 123.7, 120.9, 119.1, 118.2, 117.1, 108.5, 104.9, 102.9, 102.1, 101.7, 56.3, 56.2, 52.2, 40.3, 14.7, 7.0, 7.0. HRMS (ESI) m/z: 461.1732 [M + H]+, calcd for C26H25N2O6 461.1707.
4.6. TOP1-mediated cleavage assay
DNA cleavage assays were performed using a 3’-[32P]-labeled 117-bp DNA oligonucleotide as substrate according to the previously reported method [26]. Approximately 2 nM radiolabeled DNA substrate was incubated with recombinant TOP1 in 20 mL of reaction buffer (10 mM Tris-HCl pH 7.5, 50 mM KCl, 5 mM MgCl2, 0.1 mM EDTA, and 15 mg/mL BSA) at 25 °C for 20 min in the presence of various concentrations of test compounds. The reactions were terminated by adding SDS (0.5% final concentration) followed by the addition of two volumes of loading dye (80% formamide, 10 mM sodium hydroxide, 1 mM sodium EDTA, 0.1% xylene cyanol, and 0.1% bromophenol blue). Aliquots of each reaction mixture were subjected to 20% denaturing PAGE. Gels were dried and visualized by using a phosphoimager and ImageQuant software (Molecular Dynamics). Cleavage sites are numbered to reflect actual sites on the 117-bp oligonucleotide.
4.7. Molecular modelling
The molecular modelling was conducted as previously reported method [22]. Briefly, the X-ray crystal structures of the ternary TOP1-DNA-ligand complex (PDB ID: 1K4T) was obtained and cleaned, inspected for errors and missing residues, hydrogens were added, and the water molecules and the ligand were deleted. The ternary complex ligand centroid coordinates for docking were defined using the ligand in the complex structure as the center of the binding pocket. The compounds constructed using ChemDraw were saved in SDFfile formats and corrected using MOE software. The top 30 docking poses per ligand were inspected visually following the docking runs. The highest-ranked poses for these ligands were merged into the crystal structure. Energy minimizations were performed for the highest-ranked poses for these ligands. The AMBER forcefield was utilized within the MOE software for energy minimization.
4.8. Cell culture and MTT assay
The cells were cultured on RPMI-1640 medium at 37 °C in a humidified atmosphere with 5% CO2. All cells to be tested in the following assays had a passage number of 3–6. For the drug treatment experiments, the cancer cells were treated with the compounds (predissolved in DMSO) at a five-dose assay ranging from 0.01 to 100 μM concentration. After incubation for 72 h at 37 °C, MTT solution (50 μL, 1 mg/mL) in PBS (PBS without MTT as the blank) was fed to each well of the culture plate (containing 100 mL medium). After 4 h incubation, the formazan crystal formed in the well was dissolved with 100 mL of DMSO for optical density reading at 570 nm [35]. The GI50 value was calculated by nonlinear regression analysis (GraphPad Prism).
4.9. Immunodetection of cellular TOP1-DNA complex
The ICE assays for cellular TOP1-DNA adduct was performed according to the reported method [36]. Briefly, mid-log phase HCT116 cells were incubated with drugs at the indicated concentration for 1 h. And then, the cells were lysed with DNAzol Reagent (1 mL) at 25 °C for 30 min. Ethanol (0.5 ml, 100%) was subsequently added and mixed with the lysate and the solution was incubated overnight at −20 °C. The genomic DNA was collected by centrifugation (12,000 rpm) at 25 °C for 10 min and washed with 75% ethanol. The precipitated DNA was dissolved in NaOH (8 mM, 0.2 ml). The pH value was adjusted to 7.2 by adding HEPES (1 M). After centrifugation, supernatant was used to quantify the DNA concentration. DNA (2 μg) were dissolved in NaH2PO4 buffer (30 μL, 25 mM, pH 6.5) and then loaded onto nitrocellulose membranes. Membranes were incubated with rabbit monoclonal to human TOP1 (Abcam, 1:1000) overnight at 4 °C, and then incubated with the appropriate HRPconjugated secondary antibodies (Cell Signaling Technology, 1:3000) at room temperature for 1 h. Reactive dots were detected using Immobilon Western Chemiluminescent HRP Substrate (Millipore).
4.10. γH2AX detection
γH2AX staining was performed as described [37]. Briefly, HCT116 cells (2 ×104 cells/mL) were grown in culture medium and treated with compounds for 3 h at 37 °C. After incubation, cells were fixed in 4% paraformaldehyde/PBS for 15 min at 25 °C and washed three times with PBS buffer. Cells were permeabilized with 0.5% Triton X-100 in PBS at 0 °C for 30 min. Dish was blocked with 5% goat serum/PBS at 37 °C for 3 h. Immunofluorescence assay was performed using standard methods, and the slides were incubated alternately with phospho-γH2AX (Cell Signaling Technology) at 37 °C overnight. The cover slips were washed six times with blocking buffer and then incubated with anti-rabbit alexa 488-conjugated antibody (A21206, Life Technology) and 2.0 μg/mL of 4’,6-diamidino-2-phenylindole (DAPI, Invitrogen) at 37 °C for 2 h. The dishes were again washed six times with blocking buffer. Digital images were recorded using an LSM710 microscope (Zeiss, Germany) and analyzed with ZEN software.
4.11. Pharmacokinetic study in vivo
The PK study was conducted according to our previous method [22]. Briefly, male SD rats (weighing 220–250 g, n = 2) were treated with compound 12 pre-dissolved in10% DMSO and 10% Kolliphor® HS15 (a non-ionic solubilizer) by iv (1 mg/kg) and by ig administration (5 mg/kg), respectively. Blood samples (200 μL) were collected into heparinized tubes via the jugular vein at the following times: 0.083, 0.25, 0.5, 1, 2 and 4 h after dosing. Plasma samples (100 μL) were obtained after centrifugation for 10 min at 3000 rpm and stored at −20 °C until used for analysis. The plasma was detected through LC-MS-MS.
4.12. In vivo acute toxicity
Based on the preliminary experiments, the Kuning male mice were randomly divided into six groups (n = 6) and administered by intraperitoneal (ip) injection. The control group was treated with an equivalent volume of sterile water. The testing groups were treated with compound 12 in a single dose 300, 240, 192, 153.6 and 122.9 mg/kg, respectively. The mice were kept under observation for 7 days post-treatment in order to check for any behavioral (poisoning symptoms and body weight) and death. All animals were euthanized by cervical dislocation at the end of the experiments.
4.13. In vivo antitumor activity
Athymic nude mice bearing the nu/nu gene were obtained from Laboratory Animal Center of Sun Yat-sen University and maintained in pathogen-free conditions to establish the model of xenografts of HCT116 and MCF-7. Male nude mice 4–5 weeks old weighing 12–15 g were used. Tumor pre-induced in the mice by subcutaneously injecting of cancer cells (100 μL, 1× 107 cells) was implanted. When the implanted tumors had reached a volume of about 80 mm3, the mice were randomly divided into four groups (n = 6) and administered by intraperitoneal injection. The testing groups were treated with 12 in 20 mg/kg, 10 mg/kg and 5 mg/kg dose once every day, respectively. The negative control group was treated with an equivalent volume of saline. Tumor volumes (V) were monitored by caliper measurement of the length and width, and calculated using the formula: V = (larger diameter) × (smaller diameter)2/2, and growth curves were plotted using average tumor volume within each experimental group at the set time points. At the end of the experiment, the animals were euthanized by cervical dislocation. The tumors were removed and weighed. The tumor weight inhibition (TWI) was calculated according to the formula: TWI = (1-Mean tumor weight of the experimental group/Mean tumor weight of the control group) × 100%.
4.14. Statisticl analysis
All data are expressed as the mean ± standard deviation. Statistical comparisons were conducted using a one-way analysis of variance (ANOVA) using the Prism statistical software package (GraphPad Software, USA), followed by Tukey’s test.
Supplementary Material
Acknowledgement
This work was supported by the National Natural Science Foundation of China (No. 81373257), Natural Science Foundation of Guangdong Province (No. S2013010015609), Guangdong Provincial Key Laboratory of Construction Foundation (No. 2017B030314030) and supported by the Intramural Program of the National Cancer Institute (Center for Cancer Research), National Institutes of Health, Bethesda, Maryland, USA (Z01 BC 006150-19 and Z01 BC006161-171).
Footnotes
Appendix A. Supplementary data
Supplementary data to this article can be found online at https://doi.org/10.1016/j.ejmech.2019.05.074.
References
- [1].Wang JC, DNA topoisomerases, Annu. Rev. Biochem. 65 (1996) 635–692. [DOI] [PubMed] [Google Scholar]
- [2].Stewart L, Redinbo MR, Qui X, Hol WGJ, Champoux JJ, A model for the mechanism of human topoisomerase I, Science 279 (1998) 1534–1541. [DOI] [PubMed] [Google Scholar]
- [3].Champoux JJ, DNA topoisomerases: structure, function, and mechanism, Annu. Rev. Biochem. 70 (2001) 369–413. [DOI] [PubMed] [Google Scholar]
- [4].Pommier Y, Leo E, Zhang H, Marchand C, DNA topoisomerases and their poisoning by anticancer and antibacterial drugs, Chem. Biol. 17 (2010) 421–433. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [5].Pommier Y, Sun Y, Huang SN, Nitiss JL, Roles of eukaryotic topoisomerases in transcription, replication and genomic stability, Nat. Rev. Mol. Cell Biol. 17 (2016) 703–721. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [6].Pommier Y, DNA topoisomerase I inhibitors: chemistry, biology, and interfacial inhibition, Chem. Rev. 109 (2009) 2894–2902. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [7].Staker BL, Hjerrild K, Feese MD, Behnke CA, Burgin AB Jr., L. Stewart, The mechanism of topoisomerase I poisoning by a camptothecin analog, Proc. Natl. Acad. Sci. U. S. A. 99 (2002) 15387–15392. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [8].Staker BL, Feese MD, Cushman M, Pommier Y, Zembower D, Stewart L, Burgin AB, Structures of three classes of anticancer agents bound to the human topoisomerase I-DNA covalent complex, J. Med. Chem. 48 (2005) 2336–2345. [DOI] [PubMed] [Google Scholar]
- [9].Pommier Y, Topoisomerase I inhibitors: camptothecins and beyond, Nat. Rev. Canc. 6 (2006) 789–802. [DOI] [PubMed] [Google Scholar]
- [10].Beretta GL, Perego P, Zunino F, Targeting topoisomerase I: molecular mechanisms and cellular determinants of response to topoisomerase I inhibitors, Expert Opin. Ther. Targets 12 (2008) 1243–1256. [DOI] [PubMed] [Google Scholar]
- [11].Choi S-H, Tsuchida Y, Yang H-W, Oral versus intraperitoneal administration of irinotecan in the treatment of human neuroblastoma in nude mice, Cancer Lett. 124 (1998) 15–21. [DOI] [PubMed] [Google Scholar]
- [12].Brogden RN, Wiseman LR, Topotecan a review of its potential in advanced ovarian cancer, Drugs 56 (1998) 709–723. [DOI] [PubMed] [Google Scholar]
- [13].Bracher F, Tremmel T, From lead to drug utilizing a mannich reaction: the topotecan story, Arch. Pharm. Chem. Life Sci. 350 (2017), e1600236. [DOI] [PubMed] [Google Scholar]
- [14].Zhang R, Li Y, Cai Q, Liu T, Sun H, Chambless B, Preclinical pharmacology of the natural product anticancer agent 10-hydroxycamptothecin, an inhibitor of topoisomerase I, Cancer Chemother, Pharmacologist 41 (1998) 257–267. [DOI] [PubMed] [Google Scholar]
- [15].Jianghe S, Fengming H, Jianwei F, Clinical study on treatment of non-small cell lung cancer with 10-hydroxycamptothecin, Chin. J. Clin. Oncol. 30 (2003) 26–28. [Google Scholar]
- [16].Kim SJ, Kim JS, Kim SC, Kim YK, Kim YK, Kang JY, Yoon HK, Song JS, Lee SH, Moon HS, Kim JW, Kim KH, Kim CH, Shim BY, Kim HK, A multicenter phase II study of belotecan, new camptothecin analogue, in patients with previously untreated extensive stage disease small cell lung cancer, Lung Cancer 68 (2010) 446–449. [DOI] [PubMed] [Google Scholar]
- [17].Lian Q, Xu J, Yan S, Huang M, Ding H, Sun X, Bi A, Ding J, Sun B, Geng M, Chemotherapy-induced intestinal inflammatory responses are mediated by exosome secretion of double-strand DNA via AIM2 inflammasome activation, Cell Res. 27 (2017) 784–800. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [18].Li T-K, Houghton PJ, Desai SD, Daroui P, Liu AA, Hars ES, Ruchelman AL, LaVoie EJ, Liu LF, Characterization of ARC-111 as a novel topoisomerase Itargeting anticancer drug, Cancer Res. 63 (2003) 8400–8407. [PubMed] [Google Scholar]
- [19].Burton JH, Mazcko CN, LeBlanc AK, Covey JM, Ji JJ, Kinders RJ, Parchment RE, Khanna C, Paoloni M, Lana SE, Weishaar K, London CA, Kisseberth WC, Krick E, Vail DM, Childress MO, Bryan JN, Barber LG, Ehrhart EJ, Kent MS, Fan TM, Kow KY, Northup N, Wilson-Robles H, Tomaszewski JE, Holleran JL, Muzzio M, Eiseman J, Beumer JH, Doroshow JH, Pommier Y, NCI comparative oncology Program testing of non-camptothecin indenoisoquinoline topoisomerase I inhibitors in naturally occurring canine lymphoma, Clin. Cancer Res. 24 (2018) 5830–5840. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [20].Qin XJ, Yu Q, Yan H, Khan A, Feng MY, Li PP, Hao XJ, An LK, Liu HY, Meroterpenoids with antitumor activities from guava (Psidium guajava), J. Agric. Food Chem. 65 (2017) 4993–4999. [DOI] [PubMed] [Google Scholar]
- [21].Qin XJ, Jin LY, Yu Q, Liu H, Khan A, Yan H, Hao XJ, An LK, Liu HY, Eucalypglobulusals A-J, Formyl-phloroglucinol-terpene meroterpenoids from Eucalyptus globulus fruits, J. Nat. Prod. 81 (2018) 2638–2646. [DOI] [PubMed] [Google Scholar]
- [22].Zhang XR, Wang HW, Tang WL, Zhang Y, Yang H, Hu DX, Ravji A, Marchand C, Kiselev E, Ofori-Atta K, Agama K, Pommier Y, An LK, Discovery, synthesis, and evaluation of oxynitidine derivatives as dual inhibitors of DNA topoisomerase IB (TOP1) and tyrosyl-DNA phosphodiesterase 1 (TDP1), and potential antitumor agents, J. Med. Chem. 61 (2018) 9908–9930. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [23].Liu C-C, Korivi RP, Cheng C-H, Cobalt-catalyzed regioselective synthesis of indenamine from o-iodobenzaldimine and alkyne: intriguing difference to the nickel-catalyzed reaction, Chem. Eur J. 14 (2008) 9503–9506. [DOI] [PubMed] [Google Scholar]
- [24].Korivi RP, Cheng CH, Protecting-group-free total synthesis of isoquinoline alkaloids by nickel-catalyzed annulation of o-halobenzaldimine with an alkyne as the key step, Chem. Eur J. 16 (2010) 282–287. [DOI] [PubMed] [Google Scholar]
- [25].Cushman M, Jayaraman M, Vroman JA, Fukunaga AK, Fox BM, Kohlhagen G, Strumberg D, Pommier Y, Synthesis of new indeno[1,2-c]isoquinolines: cytotoxic non-camptothecin topoisomerase I inhibitors, J. Med. Chem. 43 (2000) 3688–3698. [DOI] [PubMed] [Google Scholar]
- [26].Dexheimer TS, Pommier Y, DNA cleavage assay for the identification of topoisomerase I inhibitors, Nat. Protoc. 3 (2008) 1736–1750. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [27].Urasaki Y, Laco GS, Pourquier P, Takebayashi Y, Kohlhagen G, Gioffre C, Zhang H, Chatterjee D, Pantazis P, Pommier Y, Characterization of a novel topoisomerase I mutation from a camptothecin resistant human prostate cancer cell line, Cancer Res. 61 (2001) 1964–1969. [PubMed] [Google Scholar]
- [28].Shoemaker RH, The NCI60 human tumour cell line anticancer drug screen, Nat. Rev. Canc. 6 (2006) 813e823. [DOI] [PubMed] [Google Scholar]
- [29].Holbeck SL, Collins JM, Doroshow JH, Analysis of Food and Drug Administration-approved anticancer agents in the NCI60 panel of human tumor cell lines, Mol. Cancer Ther. 9 (2010) 1451–1460. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [30].Reinhold WC, Sunshine M, Varma S, Doroshow JH, Pommier Y, Using cellMiner 1.6 for systems pharmacology and genomic analysis of the NCI-60, Clin. Cancer Res. 21 (2015) 3841–3852. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [31].Miao ZH, Player A, Shankavaram U, Wang YH, Zimonjic DB, Lorenzi PL, Liao ZY, Liu H, Shimura T, Zhang HL, Meng LH, Zhang YW, Kawasaki ES, Popescu NC, Aladjem MI, Goldstein DJ, Weinstein JN, Pommier Y, Nonclassic functions of human topoisomerase I: genome-wide and pharmacologic analyses, Cancer Res. 67 (2007) 8752–8761. [DOI] [PubMed] [Google Scholar]
- [32].Hsiang Y-H, Hertzberg R, Hecht S, Liu L, Camptothecin induces protein-linked DNA breaks via mammalian DNA topoisomerase I, J. Biol. Chem. 260 (1985) 14873–14878. [PubMed] [Google Scholar]
- [33].Wu CP, Hsieh CH, Wu YS, The emergence of drug transporter-mediated multidrug resistance to cancer chemotherapy, Mol. Pharm. 8 (2011) 1996–2011. [DOI] [PubMed] [Google Scholar]
- [34].Zhu J, Wang R, Lou L, Li W, Tang G, Bu X, Yin S, Jatrophane diterpenoids as modulators of P-Glycoprotein-Dependent multidrug resistance (MDR): advances of structure-activity relationships and discovery of promising MDR reversal agents, J. Med. Chem. 59 (2016) 6353–6369. [DOI] [PubMed] [Google Scholar]
- [35].Carmichael J, DeGraff WG, Gazdar AF, Minna JD, Mitchell JB, Evaluation of a tetrazolium-based semiautomated colorimetrie assay: assessment of chemosensitivity testing, Cancer Res. 47 (1987) 936–942. [PubMed] [Google Scholar]
- [36].Yalowich JC, Wu X, Zhang R, Kanagasabai R, Hornbaker M, Hasinoff BB, The anticancer thiosemicarbazones Dp44mT and triapine lack inhibitory effects as catalytic inhibitors or poisons of DNA topoisomerase IIalpha, Biochem. Pharmacol. 84 (2012) 52–58. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [37].Sooryakumar D, Dexheimer TS, Teicher BA, Pommier Y, Molecular and cellular pharmacology of the novel noncamptothecin topoisomerase I inhibitor Genz-644282, Mol. Cancer Ther. 10 (2011) 1490–1499. [DOI] [PMC free article] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.