Accepted Manuscript [1,2]Oxazolo[5,4-e]isoindoles as promising tubulin polymerization inhibitors Virginia Spanò, Marzia Pennati, Barbara Parrino, Anna Carbone, Alessandra Montalbano, Alessia Lopergolo, Valentina Zuco, Denis Cominetti, Patrizia Diana, Girolamo Cirrincione, Nadia Zaffaroni, Paola Barraja PII:
S0223-5234(16)30739-5
DOI:
10.1016/j.ejmech.2016.09.013
Reference:
EJMECH 8879
To appear in:
European Journal of Medicinal Chemistry
Received Date: 20 July 2016 Revised Date:
31 August 2016
Accepted Date: 3 September 2016
Please cite this article as: V. Spanò, M. Pennati, B. Parrino, A. Carbone, A. Montalbano, A. Lopergolo, V. Zuco, D. Cominetti, P. Diana, G. Cirrincione, N. Zaffaroni, P. Barraja, [1,2]Oxazolo[5,4-e]isoindoles as promising tubulin polymerization inhibitors, European Journal of Medicinal Chemistry (2016), doi: 10.1016/j.ejmech.2016.09.013. This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.
ACCEPTED MANUSCRIPT
N OMe MeO
Ctr 6j
400
200
0 0
10
20
30
40
RI PT
N O
Tumor Volume (mm3)
600
50
60
AC C
EP
TE D
M AN U
SC
Days after cells injection
ACCEPTED MANUSCRIPT [1,2]OXAZOLO[5,4-e]ISOINDOLES AS PROMISING TUBULIN POLYMERIZATION INHIBITORS Virginia Spanò1*, Marzia Pennati2*, Barbara Parrino1, Anna Carbone1, Alessandra Montalbano1,
Nadia Zaffaroni2, Paola Barraja1§°
1
Dipartimento di Scienze e Tecnologie Biologiche Chimiche e Farmaceutiche (STEBICEF),
SC
Università di Palermo, 90123 Palermo, Italy; 2
RI PT
Alessia Lopergolo2, Valentina Zuco2, Denis Cominetti2, Patrizia Diana1, Girolamo Cirrincione1,
Molecular Pharmacology Unit, Department of Experimental Oncology and Molecular Medicine,
M AN U
Fondazione IRCCS Istituto Nazionale dei Tumori, 20133 Milano, Italy.
*
°
TE D
These authors contributed equally to this work
Corresponding author: Paola Barraja, Dipartimento di Scienze e Tecnologie Biologiche
Chimiche e Farmaceutiche (STEBICEF), Università di Palermo, via Archirafi 32, 90123 Palermo,
AC C
EP
Italy; e-mail:
[email protected]
ACCEPTED MANUSCRIPT
Abstract A series of [1,2]Oxazolo[5,4-e]isoindoles has been synthesized through a versatile and high yielding sequence. All the new structures showed in the 1HNMR spectra, the typical signal in the
RI PT
8.34-8.47 ppm attributable to the H-3 of the [1,2]oxazole moiety. Among all derivatives, methoxy benzyl substituents at positions 3 and 4 or/and 5 were very effective in reducing the growth of different tumor cell lines, including diffuse malignant peritoneal mesothelioma (DMPM), an
SC
uncommon and rapidly malignancy poorly responsive to available therapeutic options. The most active compound 6j was found to impair tubulin polymerization, cause cell cycle arrest at G2/M
M AN U
phase and induce apoptosis in DMPM cells, making it as a new lead for the discovery of new potent
AC C
EP
TE D
antimitotic drugs.
Keywords: [1,2]oxazolo[5,4-e]isoindoles; α-hydroxyalkyl ketones; Antitubulin agents; Diffuse malignant peritoneal mesothelioma
ACCEPTED MANUSCRIPT
1. Introduction Despite the remarkable progress made in recent years, the treatment of cancer still remains the most challenging task for the mankind. In the last decades, both natural and synthetic products have
RI PT
appreciably contributed to the development of a large number of anticancer drugs [1-5]. Among these, tubulin-binding molecules represent an important class of antineoplastic agents, with broad activity in both solid and hematologic malignancies [6-8]. Such chemotherapeutic agents block cell
SC
division at the metaphase/anaphase junction of mitosis by affecting microtubule dynamics and interfering with the function of the mitotic spindle, thus leading to cell death [6-8]. The therapeutic
M AN U
potential of anti-microtubule agents (including taxanes, vinca alkaloids, macrolides and peptides) has been extensively exploited in clinical practice [7,8]. However, limitations in the use of these molecules are often related to their short half-life and the frequent incidence of tumor cell resistance (i.e., cellular efflux of the compound, ineffective interaction with the target, alterations in
TE D
microtubules dynamics, and deficient activation of programmed cell death), as well as to the development of severe toxicity [7,9]. Recently, Combretastatin A4 (CA4, Chart 1), a natural product isolated from the bark of the South African bush willow Combretum caffrum, has been
EP
considered a promising lead compound for the design and synthesis of novel microtubule targeting agents able to overcome the above mentioned limitations [8,10,11]. The IC50 of CA4 against tubulin
AC C
polymerization was found to range from 0.53 to 3.0 µM [12]. Besides its potent cytotoxicity against a broad spectrum of human cancer lines, CA4 shows high potency against MDR positive cancer cell lines. However, CA4 does not show in vivo efficacy, in part, due to its poor pharmacokinetics [13], and for this reason considerable effort has been addressed to the improvement of this issue. One example is the water soluble disodium phosphate of CA4 [14]. Moreover, cis double bond in CA4 has been replaced by several heterocycles, such as imidazole, oxazole, pyrazole, tetrazole or a thiazole [15-19]. In particular, an example of CA4 analogues demonstrating oral in vivo antitumor activity against solid tumor was achieved by incorporation of an N-methyl group into the central
ACCEPTED MANUSCRIPT imidazole ring, and much improved pharmacokinetic profiles. The in vivo studies confirmed the remarkable oral efficacy at the maximum tolerated doses (MTD) (30 mg/kg/day), with strong reduction in mean tumor mass in the animals studied [18]. Naphtylcombretastin and its analogues incorporating the isoxazole moiety displayed potent cytotoxic effect and inhibition of tubulin
RI PT
polymerization (Chart 1) [20]. In particular, 5-(naphthalen-2-yl)-4-(TMP)-1,2-oxazole (X = O, Y = CH) and 4-(naphthalen-2-yl)-5-(TMPl)-1,2-oxazole (X = CH, Y = O) (Chart 1) showed the same inhibitory potency of naphtylcombretastin and in the same order of magnitude of CA4. Another
SC
significative example is the indole-based oxazoline, A-289099, which was the S-isomer among two enantiomers, demonstrating as a potent and orally active antimitotic agent against various cancer
M AN U
cell lines including those that express the MDR phenotype [21].
A-289099 decreased the
proliferation of a variety of cells with EC50 values ranging from 5.1 to 12.8 nM, and studies on the mechanism of action indicated that A-289099 was able to depolymerize microtubules in human HCT-15 colon carcinoma cells at a concentration of 44 nM. In competition-binding assays, A-
TE D
298099 competed with [3H]colchicine for binding to tubulin (Ki = 0.65 µM). Molecular modeling studies indicated that the structure of A289099 was superimposable to that of combretastatin A4, having the trimethoxylphenyl moiety as a common pharmacophore and the 2,5-disubstituted
EP
oxazoline mimicking the cis-double bond, with a binding constant at the colchicine site comparable to colchicine at 0.63 µM but 3-fold higher than that of combretastatin A4. The pharmacokinetic
AC C
properties of A-289099 in four different species (mouse, rat, dog and monkey) indicated good oral bioavailabilities ranging from 6.5 to 18.6%. Moreover, in vivo studies against M5076 murine ovarian sarcoma in mice gave remarkable results, comparable to other antimitotic drugs such as E7010, a potent an orally active sulfonamide antitumor agent [22]. In fact, when administrated at the maximum tolerated dose (100 mg/kg/day), A-289099 achieved 206% increase in life span and an impressive 28 day delay to 1 g tumor volume in mice with M5076 murine ovarian sarcoma, copared to 27 of E7010. Chart 1. Structures of tubulin polymerization inhibitors.
ACCEPTED MANUSCRIPT
MeO
MeO
MeO
MeO OMe
OMe
OR OMe
RI PT
CA4 CA4P
Naphtylcombretastatin
OMe
Y N X
MeO
OMe
N
MeO
Me
O
OMe
N
M AN U
OMe
SC
R=H R = PO3Na2
TMP = Trimethoxyphenyl 5-(naphthalen-2-yl)-4-(TMP)-1,2-oxazole (X = O, Y = CH) 4-(naphthalen-2-yl)-5-(TMPl)-1,2-oxazole (X = CH, Y = O)
A-289099
TE D
Since many years, we have been involved in studies dealing with nitrogen heterocycles which are widely represented in natural products and potentially active drugs [23-29]. Among them, several indole derivatives have been described as potent tubulin polymerization inhibitors, thus
EP
highlighting the indole scaffold as valuable pharmacophore [30-33].. However, only few examples
AC C
have been reported in the literature about isoindole derivatives belonging to this class of anticancer agent [28,34, 35].
Isoxazoles or [1,2]oxazoles represent the core structure of many drug candidates, thus representing an attractive scaffold in medicinal chemistry continuously facing the challenge of designing new and leading drugs [36]. In particular, compounds containing the isoxazole moiety have been reported as pharmaceutically interesting agents because of their biological activities (i.e., antitumor, antiinflammatory, antidepressant, and antiviral) [37-43], and some of them were used for the treatment of pathologies, such as cystic fibrosis [44], neurological disorders [45] and tuberculosis [46].
ACCEPTED MANUSCRIPT Based on the evidence that small molecule therapeutic drugs are generally highly specific, exhibit best efficacy and mostly contain heterocyclic scaffolds, we have devoted our efforts to develop small molecules with antitumor properties [47-53]. Among these, we have studied the synthesis and biological properties of tricyclic systems of type 1-3 (Figure 1) based on the
RI PT
isoindole scaffold [35,54-58]. In addition, due to the promising antitumor properties of [1,2]oxazole derivatives, we have already investigated two classes of pyrazole- and pyrrole-fused systems incorporating the [1,2]oxazole unit of type 4 [58] and 5 [59]. In particular, two derivatives
SC
belonging to the latter class of compounds showed potent antiproliferative activity against the NCI human tumor cell lines panel at nanomolar concentrations. Starting from these promising results
M AN U
and in the search for novel promising antimitotic agents, here we report the synthesis and in vitro antitumor activity of [1,2]oxazolo[5,4-e]isoindoles 6 as positional isomers of compounds 5.
Results and Discussion
TE D
2.1 Chemistry
Thus far, only two reports on the title ring system, have been reported describing the same synthetic approach to the tricyclic system with limited yields and poor possibility of obtaining
obtain
EP
derivatives with a wide substitution pattern [60,61]. The synthetic strategy optimized by us to [1,2]oxazolo[5,4-e]isoindoles
10
is
outlined
in
Scheme
1.
We
started
from
AC C
tetrahydroisoindoles 7a-c, previously reported by us [35,54], which were ideal building blocks for our purpose considering that the α position to the carbonyl is suitable for the introduction of a second electrophilic centre, thus allowing cyclization with dinucleofiles.
Scheme 1. Synthesis of [1,2]oxazolo[5,4-e]isoindoles 6a-r
ACCEPTED MANUSCRIPT O
R2 NH
O
O
R2
a
NR
R1
b
HO
NR
c
8
3
R2 7
NR 4
9a-r
8a-r
1
N O
R1
R1
7a-c
2
R2
5
6
R1
6a-r (Table 1)
a R= Me, R1= R2= H; b R= Bn, R1= R2= H; c R= 4-MeBn, R1= R2= H; d R= 4-OMeBn, R1= R2= H; e R= 3-OMeBn, R1= R2= H; f R= 2-OMeBn, R1= R2= H; g R= 2,3(OMe)2Bn, R1= R2= H; h R= 2,5-(OMe)2Bn, R1= R2= H; i R= 3,4-(OMe)2Bn, R1= R2= H; j R= 3,5-(OMe)2Bn, R1= R2= H; k R= 3,4,5-(OMe)3Bn, R1= R2= H; l R= Ph, R1= R2= H; m R= 4-OMePh, R1= R2= H; n R= R2= Me, R1= COOEt; o R= Bn, R1= COOEt, R2= Me; p R= 4-OMeBn, R1= COOEt, R2= Me; q R= R2= Me, R1= H; r R= Bn, R1= H, R2= Me
RI PT
a R1= R2= H b R1= COOEt, R2= Me c R1 = H, R2= Me
SC
Reagents and conditions: (a) for 8a-d,n-r,[35,54] for 8e-k NaH, DMF, 0 °C to rt, 1 h, then benzylchlorides at 0 °C to rt, 1-24 h, 60-79%; for 8l,[24,43] 8m K2CO3, N-methylpyrrolidone, N2,
M AN U
rt, 1 h, then Cu(I)Br, rt, 1 h, then 4-OMePhI, reflux, 3 h, 60%; (b) t-BuOK, toluene, N2, 0 °C to rt, 3 h, then HCOOEt, 0 °C to rt, 24 h, for 9a-d,l,n-r,[35,54] 9e-k,m 63–97%; (c) NH2OH•HCl, ethanol, reflux, 50 min, 56–78%.
Functionalization of the nitrogen atom was achieved by nucleophilic reactions with the proper
TE D
alkyl or aralkyl halides in dimethylformamide to give N-substituted ketones 8a-d,n-r [35,54] and 8e-k (60-79%). Instead, the N-phenyl derivatives 8l [35,54] and 8m (60%) were obtained by a modified Ulmann cross-coupling reaction in the presence of potassium carbonate as the base,
EP
copper(I)bromide and iodobenzene [35] or iodoanisole. Subsequent formylation with ethyl formate in the presence of potassium t-buthoxyde gave the corresponding α-hydroxymethylidene ketones
AC C
9a-d,l,n-r [35,54] and 9e-k,m (63-97%), which were in turn subjected to annelation of the [1,2]oxazole moiety using hydroxylamine hydrochloride as dinucleofile in refluxing ethanol yielding the corresponding [1,2]oxazole[5,4-e]isoindoles 6a-r in 56–78% yields (Table 1).
2.2 Biology 2.2.1 [1,2]oxazole[5,4-e]isoindoles impair tumor cell growth The cytotoxic activity of new [1,2]oxazole[5,4-e]isoindole derivatives was examined in a panel of human tumor cell lines of different histological origin, including melanoma (JR8 and M14),
ACCEPTED MANUSCRIPT breast cancer (MCF-7 and MDA-MB-231), castration-resistant prostate carcinoma (PC3 and DU145) and diffuse malignant peritoneal mesothelioma (DMPM; STO and MP8). Cells were exposed to increasing concentrations (0.01 to 100 µM) of each derivative for 72 h, and the effect on cell proliferation was determined by MTS assay. Results indicated that only [1,2]oxazolo[5,4-
RI PT
e]isoindoles unsubstituted at positions 6 and 8 (R1= R2= H) of the pyrrole moiety were active at micromolar/submicromolar level in all tested human cell lines with the exception of 6c and 6g. Moreover, within this series two compounds (6d and 6h) did not reduce the growth of DMPM cells
SC
(Table 2).
Although a structure-activity relationship of general application is not easy, some observations
M AN U
can be done. The presence of a benzyl or substituted benzyl groups at the pyrrole nitrogen is crucial in conferring good activity to [1,2]oxazolo derivatives (Table 2). By contrast, the presence of a methyl (6a), phenyl (6l) or 4-methoxyphenyl group (6m) significantly reduced their activity. Moreover, among the N-benzyl substituted derivatives, the presence of a methoxy substitution
TE D
generally seems to be relevant. In fact, the most potent compounds are 6e, 6i, 6j and 6k, bearing a 3-methoxy, 3,4-dimethoxy, 3,5-dimethoxy and 3,4,5-trimethoxy benzyl substitution, respectively. The presence of a methoxy group in the position 2 (6f, 6g and 6h) or its removal (6b) significantly
EP
decreased the activity, whereas the replacement of the 4-methoxy substituent with a 4-methyl in the benzyl moiety (6c) led to the inactivation of the compound.
AC C
Interestingly, the most active compound 6j showed a remarkable antiproliferative activity against DMPM, a highly chemoresistant tumor type [62,63], without affecting the growth of both normal human lung fibroblast (WI38) and adult human breast (MCF10A) cell lines (IC50 >100 µM). The antitumor activity of 6j was also evaluated on STO cells following subcutaneous xenotransplantation into athymic nude mice. As shown in Figure 2B, the intraperitoneal administration of the compound resulted in a tumor growth delay, with a maximum tumor volume inhibition of 51%. Moreover, the compound was well tolerated without any appreciable sign of toxicity.
ACCEPTED MANUSCRIPT 2.2.2 6j derivative inhibits tubulin polymerization, promotes cell cycle arrest and induces apoptosis Since previous evidence indicated that derivatives structurally related to our [1,2]oxazolo[5,4e]isoindoles interfere with tubulin polymerization [64], we investigated whether 6j affected tubulin dynamics in DMPM models. Western blot results revealed that 6j markedly reduced the
RI PT
polymerized compared to soluble fraction of tubulin in both STO and MP8 cells, with a mechanism superimposable to that observed following exposure to vinorelbine but opposite to that induced by taxol (Figure 3).
SC
The exposure of DMPM cells to 6j also resulted in a marked alteration of the distribution of the cells in the different phases of the cell cycle (Figure 4A), as assessed by flow cytometric analysis.
M AN U
Specifically, the treatment of STO and MP8 cells with 6j (at specific IC50 and IC80 of each cell line) induced a time- and dose-dependent accumulation of cells in the G2/M phase, which was paralleled by a marked reduction in the percentage of cells in the G1 phase (Figure 4A). Such an effect was comparable to that observed in the same cell lines following exposure to taxol and vinorelbine
TE D
(Figure 4A). Consistently, fluorescence microscopy analysis of DMPM cells revealed the presence of 35-70% mitotic cells within the overall cell population following 72-h exposure of STO and MP8 cells to equitoxic (IC50) concentrations of 6j, vinorelbine and taxol (Figure 4B). In addition, at the
EP
molecular level, treatment of STO and MP8 cells with 6j resulted in a significant dose- and timedependent increase in caspase-3 catalytic activity, determined in vitro by the hydrolysis of the
AC C
specific fluorogenic substrate (Figure 5). Based on the evidence that a cell cycle arrest at G2/M phase could be also due to a reduced activity of the cyclin-dependent kinase 1 (CDK1) [65], we investigated the ability of compound 6j to interfere with the enzyme’s in vitro catalytic activity. To this purpose, increasing amounts of 6j were incubated with purified recombinant CDK1/cyclin B complex and its potential kinase inhibitory effect was measured using a fluorimetric assay. Results revealed that the compound failed to modify the activity of the kinase at doses up to 100 µM, whereas Purvalanol A -a well-
ACCEPTED MANUSCRIPT known CDK1 inhibitor [66] used as a positive control- showed activity toward CDK1 at a very low concentration (IC50=0.61±0.03 µM).
3. Conclusions
RI PT
In this study, we have reported the synthesis of a new series of [1,2]oxazolo[5,4-e]isoindoles through a convenient, versatile and high yielding sequence by annelation of the [1,2]oxazole ring into the isoindole moiety. The analysis of the structure-activity relationship indicated that the
SC
existence of a benzyl (or substituted benzyl) groups at the pyrrole nitrogen is crucial in conferring good activity to the compounds. In particular, the presence of N-methoxy groups at positions 3, 4
M AN U
and/or 5 allowed to obtain the greatest response. Indeed, in vitro studies indicated that these derivatives significantly impaired the growth of human cancer cells lines of different histological origin, including experimental models of DMPM, a lethal disease lacking of effective therapeutic approaches. Notably, these derivatives did not appreciably interfere with the growth of human cells,
TE D
thus suggesting a preferential activity against malignant cells.
As far as the mechanisms underlying the antiproliferative effect of 6j were concerned, we demonstrated that the compound impaired microtubule assembly during mitosis, in a vinca alkaloid-
EP
like manner, and consequently induced a dose- and time-dependent cell cycle arrest at the G2/M compartment, which was paralleled by an induction of apoptosis.
AC C
Overall, our results indicated this derivative as a new lead for the discovery of new potent antimitotic drugs, and suggest [1,2]oxazolo[5,4-e]isoindoles as a possible new therapeutic approach for the treatment of DMPM.
4. Experimental Section 4.1. Chemistry 4.1.1. General Methods
ACCEPTED MANUSCRIPT All melting points were taken on a Büchi melting point M-560 apparatus. IR spectra were determined in bromoform with Shimadzu FT/IR 8400S spectrophotometer. 1H and
13
C NMR
spectra were measured at 200 and 50.0 MHz, respectively, in DMSO-d6 or CDCl3 solution using a Bruker Avance II series 200 MHz spectrometer. Column chromatography was performed with
RI PT
Merck silica gel (230−400 mesh ASTM) or a Büchi Sepacor chromatography module (prepacked cartridge system). Elemental analyses (C, H, N) were within ±0.4% of theoretical values and were performed with a VARIO EL III elemental analyzer. The purity of all the tested compounds was
SC
>95%, determined by HPLC (Agilent 1100 series).
Compounds 7a-c, 8a-d,l,n-r and 9a-d,l,n-r were prepared according to procedures earlier reported
M AN U
by us.[24,43] Representative spectra of compounds 6e, 6i, 6j, 6k, 6m were included as supplementary material.
4.1.2 General procedure for the synthesis of 2-substituted-2,5,6,7-tetrahydro-4H-isoindol-4-ones (8).
TE D
To a solution of 7a (1.35 g, 10 mmol) in anhydrous DMF (15 mL) NaH (0.24 g, 10 mmol) was added at 0 °C and the reaction was stirred for 1 h at rt. The suitable benzyl chloride (15 mmol) was added at 0 °C and the reaction mixture was stirred at rt up to completeness (TLC). Then it was
EP
poured into crushed ice. The aqueous solution was extracted with dichloromethane (3 x 50 mL), and the organic phase was dried over Na2SO4. After evaporation of the solvent at reduced pressure, the
eluent.
AC C
crude residue was purified by chromatography column using dichloromethane/ethyl acetate as
4.1.2.1. 2-(3-Methoxybenzyl)-2,5,6,7-tetrahydro-4H-isoindol-4-one (8e). This compound was obtained from reaction of 7a with 3-methoxybenzylchloride after 1 h and 30 min. White solid; yield 69%; Rf=0.41 (CH2Cl2/EtOAc, 9:1); m.p.:120–121 °C; IR cm-1: 1649 (CO); 1H NMR (DMSO-d6, 200 MHz, ppm): δ 1.83–1.96 (m, 2H, CH2), 2.29 (t, 2H, J = 6.0 Hz, CH2), 2.57 (t, 2H, J = 6.0 Hz, CH2), 3.73 (s, 3H, CH3), 5.06 (s, 2H, CH2), 6.67 (s, 1H, H-1), 6.82–6.88 (m, 3H, Ar), 7.26 (t, 1H, J = 8.0 Hz, Ar), 7.43 (s, 1H, H-3); 13C NMR (DMSO-d6, 50 MHz, ppm): δ 21.1 (t), 24.8 (t), 38.8 (t),
ACCEPTED MANUSCRIPT 52.6 (t), 55.0 (q), 112.9 (d), 113.6 (d), 117.3 (d), 119.8 (d), 121.3 (s), 122.1 (d), 126.4 (s), 129.7 (d), 139.3 (s), 159.4 (s), 193.6 (s). Anal calcd for C16H17NO2 (255.31): C 75.27, H 6.71, N 5.49. Found: C 75.43, H 6.90, N 5.15. 4.1.2.2 2-(2-Methoxybenzyl)-2,5,6,7-tetrahydro-4H-isoindol-4-one (8f). This compound was
RI PT
obtained from reaction of 7a with 2-methoxybenzylchloride after 1 h and 30 min. Yellow oil; yield 69%; Rf=0.44 (CH2Cl2/EtOAc, 9:1); IR cm-1: 1658 (CO); 1H NMR (DMSO-d6, 200 MHz, ppm): δ 1.83–1.96 (m, 2H, CH2), 2.29 (t, 2H, J = 6.1 Hz, CH2), 2.57 (t, 2H, J = 6.1 Hz, CH2), 3.83 (s, 3H,
SC
CH3), 5.06 (s, 2H, CH2), 6.63 (s, 1H, H-1), 6.91 (t, 1H, J = 7.3 Hz, Ar), 7.01–7.08 (m, 2H, Ar), 7.24–7.43 (m, 2H, H-3 and Ar); 13C NMR (DMSO-d6, 50 MHz, ppm): δ 21.6 (t), 25.3 (t), 39.3 (t),
M AN U
48.2 (t), 55.9 (q), 111.4 (d), 118.0 (d), 121.0 (d), 121.6 (s), 122.6 (d), 125.9 (s), 126.6 (s), 129.5 (d), 130.0 (d), 157.1 (s), 194.0 (s). Anal calcd for C16H17NO2 (255.31): C 75.27, H 6.71, N 5.49. Found: C 75.40, H 6.82, N 5.36.
4.1.2.3 2-(2,3-Dimethoxybenzyl)-2,5,6,7-tetrahydro-4H-isoindol-4-one (8g). This compound was
TE D
obtained from reaction of 7a with 2,3-dimethoxybenzylchloride after 24 h. Yellow oil; yield 79%; Rf=0.34 (CH2Cl2/EtOAc, 9:1); IR cm-1: 1653 (CO); 1H NMR (DMSO-d6, 200 MHz, ppm): δ 1.83– 1.95 (m, 2H, CH2), 2.29 (t, 2H, J = 6.0 Hz, CH2), 2.57 (t, 2H, J = 6.0 Hz, CH2), 3.72 (s, 3H, CH3),
EP
3.80 (s, 3H, CH3), 5.08 (s, 2H, CH2), 6.62 (s, 1H, H-1), 6.69–6.77 (m, 1H, Ar), 7.02–7.05 (m, 2H,
AC C
Ar), 7.34 (s, 1H, H-3); 13C NMR (DMSO-d6, 50 MHz, ppm): δ 21.6 (t), 25.3 (t), 39.3 (t), 48.2 (t), 56.2 (q), 60.6 (q), 113.3 (d), 117.9 (d), 121.3 (d), 121.5 (s), 122.6 (d), 124.6 (d), 126.7 (s), 131.3 (s), 146.7 (s), 152.8 (s), 194.0 (s). Anal calcd for C17H19NO3 (285.34): C 71.56, H 6.71, N 4.91. Found: C 71.75, H 6.60, N 5.21.
4.1.2.4 2-(2,5-Dimethoxybenzyl)-2,5,6,7-tetrahydro-4H-isoindol-4-one (8h). This compound was obtained from reaction of 7a with 2,5-dimethoxybenzylchloride after 24 h. Yellow oil; yield 64%; Rf=0.38 (CH2Cl2/EtOAc, 9:1); IR cm-1: 1651 (CO); 1H NMR (DMSO-d6, 200 MHz, ppm): δ 1.83– 1.96 (m, 2H, CH2), 2.29 (t, 2H, J = 6.1 Hz, CH2), 2.57 (t, 2H, J = 6.1 Hz, CH2), 3.67 (s, 3H, CH3), 3.77 (s, 3H, CH3), 5.01 (s, 2H, CH2), 6.64 (s, 1H, H-1), 6.70 (d, 1H, J = 2.9 Hz, Ar), 6.83–6.98 (m,
ACCEPTED MANUSCRIPT 2H, Ar), 7.33 (s, 1H, H-3); 13C NMR (DMSO-d6, 50 MHz, ppm): δ 21.6 (t), 25.2 (t), 39.3 (t), 48.3 (t), 55.9 (q), 56.4 (q), 112.7 (d), 114.0 (d), 116.4 (d), 118.2 (d), 121.4 (s), 122.7 (d), 126.7 (s), 127.0 (s), 151.4 (s), 153.8 (s), 194.5 (s). Anal calcd for C17H19NO3 (285.34): C 71.56, H 6.71, N 4.91. Found: C 71.30, H 6.60, N 5.13.
RI PT
4.1.2.5 2-(3,4-Dimethoxybenzyl)-2,5,6,7-tetrahydro-4H-isoindol-4-one (8i). This compound was obtained from reaction of 7a with 3,4-dimethoxybenzylchloride after 1 h. Light brown solid; yield 60%; Rf=0.31 (CH2Cl2/EtOAc, 9:1); m.p.: 102–103 °C; IR cm-1: 1651 (CO); 1H NMR (DMSO-d6,
SC
200 MHz, ppm): δ 1.82–1.95 (m, 2H, CH2), 2.28 (t, 2H, J = 6.1 Hz, CH2), 2.56 (t, 2H, J = 6.1 Hz, CH2), 3.72 (s, 3H, CH3), 3.74 (s, 3H, CH3), 4.99 (s, 2H, CH2), 6.67 (s, 1H, H-1), 6.81–7.01 (m, 3H,
M AN U
Ar), 7.41 (s, 1H, H-3); 13C NMR (DMSO-d6, 50 MHz, ppm): δ 21.1 (t), 24.8 (t), 38.8 (t), 52.5 (t), 55.4 (q), 55.5 (q), 111.8 (d), 112.0 (d), 117.1 (d), 120.3 (d), 121.1 (s), 121.8 (d), 126.3 (s), 130.0 (s), 148.4 (s), 148.7 (s), 193.5 (s). Anal calcd for C17H19NO3 (285.34): C 71.56, H 6.71, N 4.91. Found: C 71.69, H 6.58, N 5.15.
TE D
4.1.2.6 2-(3,5-Dimethoxybenzyl)-2,5,6,7-tetrahydro-4H-isoindol-4-one (8j). This compound was obtained from reaction of 7a with 3,5-dimethoxybenzylchloride after 1 h. White solid; yield 63%; Rf=0.34 (CH2Cl2/EtOAc, 9:1); m.p.: 90–91 °C; IR cm-1: 1649 (CO); 1H NMR (DMSO-d6, 200
EP
MHz, ppm): δ 1.84–1.96 (m, 2H, CH2), 2.29 (t, 2H, J = 6.1 Hz, CH2), 2.58 (t, 2H, J = 6.1 Hz, CH2),
AC C
3.72 (s, 6H, 2 x CH3), 5.01 (s, 2H, CH2), 6.44 (s, 1H, H-1), 6.45–6.47 (m, 2H, H-2’ and H-6’), 6.68 (s, 1H, H-4’), 7.43 (s, 1H, H-3); 13C NMR (DMSO-d6, 50 MHz, ppm): δ 21.1 (t), 24.8 (t), 38.9 (t), 52.6 (t), 55.2 (2 x q), 99.1 (d), 105.9 (2 x d), 117.4 (d), 121.2 (s), 122.1 (d), 126.3 (s), 139.9 (s), 160.6 (2 x s), 193.6 (s). Anal calcd for C17H19NO3 (285.34): C 71.56, H 6.71, N 4.91. Found: C 71.67, H 6.55, N 5.09. 4.1.2.7 2-(3,4,5-Trimethoxybenzyl)-2,5,6,7-tetrahydro-4H-isoindol-4-one (8k). This compound was obtained from reaction of 7a with 3,4,5-trimethoxybenzylchloride after 24 h. White solid; yield 60%; Rf=0.28 (CH2Cl2/EtOAc, 9:1); m.p.: 101–102 °C; IR cm-1: 1657 (CO); 1H NMR (DMSO-d6, 200 MHz, ppm): δ 1.83–1.95 (m, 2H, CH2), 2.29 (t, 2H, J = 6.4 Hz, CH2), 2.57 (t, 2H, J = 6.4 Hz,
ACCEPTED MANUSCRIPT CH2), 3.62 (s, 3H, CH3), 3.75 (s, 6H, 2 xC H3), 4.98 (s, 2H, CH2), 6.73 (s, 3H, H-2’, H-6’ and H-1), 7.45 (s, 1H, H-3); 13C NMR (DMSO-d6, 50 MHz, ppm): δ 21.1 (t), 24.8 (t), 38.8 (t), 52.9 (t), 55.9 (2 x q), 59.9 (q), 105.7 (2 x d), 117.2 (d), 121.1 (d), 121.9 (s), 126.3 (s), 133.1 (s), 137.0 (s), 152.9 (2 x s), 193.6 (s). Anal calcd for C18H21NO4 (315.36): C 68.55, H 6.71, N 4.44. Found C 68.36, H 6.87,
RI PT
N 4.32.
4.1.3 Synthesis of 2-(4-methoxyphenyl)-2,5,6,7-tetrahydro-4H-isoindol-4-one (8m).
To a solution of 7a (20 mmol) in N-methyl-2-pyrrolidone (40 mL) anhydrous K2CO3 (2.76 g, 20
SC
mmol) was added under nitrogen atmosphere and the reaction mixture was stirred at rt for 1 h. Then Cu(I)Br (5.74 g, 40 mmol) was added and the reaction mixture was stirred at rt for 1 h and finally 4-
M AN U
iodoanisole (16.38 g, 70 mmol) was added. The reaction mixture was heated under reflux for 3 h. After cooling, HCl (5%, 20 mL) was added and the mixture was stirred for 1 h, then ethyl acetate (20 mL) was added and the mixture was stirred for further 30 min. Then the resulting solution was filtered through celite and washed with ethyl acetate (20 mL). The organic layer was stirred for 1 h
TE D
with ice and water, separated, dried over Na2SO4 and the solvent evaporated at reduced pressure. The crude product was purified by chromatography column using dichloromethane/ethyl acetate (9:1) as eluent. Brown solid; yield 60%; Rf=0.50 (CH2Cl2/EtOAc, 9:1); m.p.: 88–89 °C; IR cm-1:
EP
1655 (CO); 1H NMR (DMSO-d6, 200 MHz, ppm): δ 1.91–2.04 (m, 2H, CH2), 2.38 (t, 2H, J = 6.4
AC C
Hz, CH2), 2.68 (t, 2H, J = 6.4 Hz, CH2), 3.79 (s, 3H, CH3), 7.03 (d, 2H, J = 9.0 Hz, H-3’ and H-5’), 7.14 (d, 1H, J = 2.2 Hz, H-1), 7.57 (d, 2H, J = 9.0 Hz, H-2’ and H-6’), 7.77 (d, 1H, J = 2.2 Hz, H3); 13C NMR (DMSO-d6, 50 MHz, ppm): δ 21.1 (t), 24.6 (t), 38.9 (t), 55.4 (q), 114.7 (2 x d), 116.1 (d), 119.6 (d), 121.7 (2 x d), 122.7 (s), 127.4 (s), 132.6 (s), 157.8 (s), 193.9 (s). Anal calcd for C15H15NO2 (241.29): C 74.67, H 6.27, N 5.81. Found: C 74.49, H 6.16, N 5.99. 4.1.4 General procedure for the synthesis of 5-(hydroxymethylidene)-2-substituted-2,5,6,7tetrahydro-4H-isoindol-4-ones (9). To a suspension of t-BuOK (3.7 g, 36 mmol) in anhydrous toluene (30 mL), a solution of 8 (12 mmol) in the same solvent (40 mL) was added dropwise under N2 at 0 °C. After 3 h stirring at rt the
ACCEPTED MANUSCRIPT reaction was cooled at 0 °C and a solution of ethyl formate (2.90 mL, 36 mmol) in anhydrous toluene (20 mL) was added and the mixture was kept stirring at rt for 24 h, then the solvent was removed at reduced pressure. The residue was dissolved in water and the solution was washed with diethyl ether. The acqueous solution was then acidified with HCl 6 M and in case of a precipitate
RI PT
formed it was filtered and dried, otherwise the solution was extracted with dichloromethane (3 x 30 mL). The organic phase was dried over Na2SO4 and the solvent evaporated at reduced pressure. The crude product was purified by chromatography column using dichloromethane as eluent.
SC
4.1.4.1 5-(Hydroxymethylidene)-2-(3-methoxybenzyl)-2,5,6,7-tetrahydro-4H-isoindol-4-one (9e). This compound was obtained from reaction of 8e. Yellow oil; yield 87%; Rf=0.36 (CH2Cl2); IR cm: 2935 (OH), 1633 (CO); 1H NMR (CDCl3, 200 MHz, ppm): δ 2.47 (t, 2H, J = 6.9 Hz, CH2), 2.63
M AN U
1
(t, 2H, J = 6.9 Hz, CH2), 3.77 (s, 3H, CH3), 4.98 (s, 2H, CH2), 6.42 (s, 1H, H-1), 6.69–6.87 (m, 3H, Ar), 7.22 (s, 1H, H-3), 7.26–7.30 (m, 1H, Ar), 7.48 (s, 1H, CH), 14.35 (bs, 1H, OH);
13
C NMR
(CDCl3, 50 MHz, ppm): δ 21.1 (t), 25.8 (t), 54.0 (t), 55.3 (q), 109.4 (s), 113.3 (d), 113.4 (d), 117.4
TE D
(d), 119.7 (d), 121.0 (s), 122.7 (d), 125.9 (s), 130.0 (d), 137.9 (s), 160.0 (s), 165.5 (d), 186.8 (s). Anal calcd for C17H17NO3 (283.32): C 72.07, H 6.05, N 4.94. Found: C 72.38, H 6.34, N 4.82. 4.1.4.2 5-(Hydroxymethylidene)-2-(2-methoxybenzyl)-2,5,6,7-tetrahydro-4H-isoindol-4-one (9f).
: 2937 (OH), 1633 (CO); 1H NMR (CDCl3, 200 MHz, ppm): δ 2.47 (t, 2H, J = 6.7 Hz, CH2), 2.63
AC C
1
EP
This compound was obtained from reaction of 8f. Yellow oil; yield 70%; Rf=0.40 (CH2Cl2); IR cm-
(t, 2H, J = 6.7 Hz, CH2), 3.84 (s, 3H, CH3), 5.02 (s, 2H, CH2), 6.45 (s, 1H, H-1), 6.87–7.03 (m, 3H, Ar), 7.25–7.34 (m, 2H, H-3 and Ar), 7.46 (s, 1H, CH), 14.39 (bs, 1H, OH);
13
C NMR (CDCl3, 50
MHz, ppm): δ 21.1 (t), 25.9 (t), 49.0 (t), 55.4 (q), 109.4 (s), 110.5 (d), 117.5 (d), 120.6 (s), 120.7 (d), 122.9 (d), 124.8 (s), 125.4 (s), 129.2 (d), 129.7 (d), 157.0 (s), 165.0 (d), 187.0 (s). Anal calcd for C17H17NO3 (283.32): C 72.07, H 6.05, N 4.94. Found: C 72.32, H 6.26, N 4.87. 4.1.4.3
2-(2,3-Dimethoxybenzyl)-5-(hydroxymethylidene)-2,5,6,7-tetrahydro-4H-isoindol-4-one
(9g). This compound was obtained from reaction of 8g. Yellow oil; yield 65%; Rf=0.28 (CH2Cl2); IR cm-1: 2935 (OH), 1633 (CO); 1H NMR (CDCl3, 200 MHz, ppm): δ 2.47 (t, 2H, J = 6.6 Hz, CH2),
ACCEPTED MANUSCRIPT 2.62 (t, 2H, J = 6.6 Hz, CH2), 3.75 (s, 3H, CH3), 3.87 (s, 3H, CH3), 5.04 (s, 2H, CH2), 6.46 (s, 1H, H-1), 6.67–6.72 (m, 1H, Ar), 6.88–7.07 (m, 2H, Ar), 7.30 (s, 1H, H-3), 7.47 (s, 1H, CH), 14.39 (bs, 1H, OH);
13
C NMR (CDCl3, 50 MHz, ppm): δ 21.1 (t), 25.9 (t), 49.0 (t), 55.8 (q), 60.6 (q), 109.4
(s), 112.7 (d), 117.4 (d), 120.7 (s), 121.1 (d), 122.7 (d), 124.3 (d), 125.6 (s), 130.1 (s), 146.9 (s),
RI PT
152.8 (s), 165.2 (d), 186.9 (s). Anal calcd for C18H19NO4 (313.35): C 68.99, H 6.11, N 4.47. Found: C 68.67, H 6.33, N 4.35. 4.1.4.4
2-(2,5-Dimethoxybenzyl)-5-(hydroxymethylidene)-2,5,6,7-tetrahydro-4H-isoindol-4-one
SC
(9h). This compound was obtained from reaction of 8h. Yellow oil; yield 72%; Rf=0.38 (CH2Cl2); IR cm-1: 2933 (OH), 1633 (CO); 1H NMR (CDCl3, 200 MHz, ppm): δ 2.47 (t, 2H, J = 6.7 Hz, CH2),
M AN U
2.63 (t, 2H, J = 6.7 Hz, CH2), 3.73 (s, 3H, CH3), 3.80 (s, 3H, CH3), 5.00 (s, 2H, CH2), 6.45 (s, 1H, H-1), 6.59 (s, 1H, Ar), 6.81 (d, 2H, J = 1.6 Hz, Ar), 7.30 (s, 1H, H-3), 7.48 (s, 1H, CH), 14.37 (bs, 1H, OH);
13
C NMR (CDCl3, 50 MHz, ppm): δ 21.1 (t), 25.9 (t), 49.0 (t), 55.7 (q), 55.9 (q), 109.4
(s), 111.5 (d), 113.5 (d), 115.7 (d), 117.5 (d), 120.7 (s), 122.9 (d), 125.5 (s), 125.9 (s), 151.2 (s),
C 68.72, H 6.34, N 4.19.
TE D
153.6 (s), 165.1 (d), 187.0 (s). Anal calcd for C18H19NO4 (313.35): C 68.99, H 6.11, N 4.47. Found:
4.1.4.5 2-(3,4-Dimethoxybenzyl)-5-(hydroxymethylidene)-2,5,6,7-tetrahydro-4H-isoindol-4-one (9i).
: 2933 (OH), 1633 (CO); 1H NMR (CDCl3, 200 MHz, ppm): δ 2.44 (t, 2H, J = 7.4 Hz, CH2), 2.62
AC C
1
EP
This compound was obtained from reaction of 8i. Yellow oil; yield 71%; Rf=0.24 (CH2Cl2); IR cm-
(t, 2H, J = 7.4 Hz, CH2), 3.84 (s, 3H, CH3), 3.87 (s, 3H, CH3), 4.96 (s, 2H, CH2), 6.42 (s, 1H, H-1), 6.69–6.86 (m, 3H, Ar), 7.28 (s, 1H, H-3), 7.48 (s, 1H, CH), 14.36 (bs, 1H, OH); 13C NMR (CDCl3, 50 MHz, ppm): δ 21.1 (t), 25.9 (t), 53.9 (t), 55.8 (q), 55.9 (q), 109.4 (s), 110.8 (d), 111.2 (d), 117.2 (d), 120.3 (d), 120.9 (s), 122.4 (d), 125.9 (s), 128.6 (s), 149.1 (s), 149.3 (s), 165.3 (d), 186.9 (s). Anal calcd for C18H19NO4 (313.35): C 68.99, H 6.11, N 4.47. Found: C 68.71, H 6.40, N 4.12. 4.1.4.6 2-(3,5-Dimethoxybenzyl)-5-(hydroxymethylidene)-2,5,6,7-tetrahydro-4H-isoindol-4-one (9j). This compound was obtained from reaction of 8j. Yellow oil; yield 63%; Rf=0.25 (CH2Cl2); IR cm1
: 2935 (OH), 1637 (CO); 1H NMR (CDCl3, 200 MHz, ppm): δ 2.49 (t, 2H, J = 6.6 Hz, CH2), 2.64
ACCEPTED MANUSCRIPT (t, 2H, J = 6.6 Hz, CH2), 3.76 (s, 6H, 2 x CH3), 4.94 (s, 2H, CH2), 6.31 (s, 2H, Ar), 6.38–6.42 (m, 2H, H-1 and Ar), 7.27 (s, 1H, H-3), 7.49 (s, 1H, CH), 14.36 (bs, 1H, OH);
13
C NMR (CDCl3, 50
MHz, ppm): δ 21.1 (t), 25.8 (t), 54.1 (t), 55.4 (2 x q), 99.7 (d), 105.7 (2 x d), 109.4 (s), 117.4 (d), 121.0 (s), 122.7 (d), 125.9 (s), 138.6 (s), 161.3 (2 x s), 165.5 (d), 186.8 (s). Anal calcd for
4.1.4.7
RI PT
C18H19NO4 (313.35): C 68.99, H 6.11, N 4.47. Found: C 68.77, H 6.34, N 4.15.
5-(Hydroxymethylidene)-2-(3,4,5-trimethoxybenzyl)-2,5,6,7-tetrahydro-4H-isoindol-4-one
(9k). This compound was obtained from reaction of 8k. Brown solid; yield 64%; Rf=0.29 (CH2Cl2);
SC
m.p.: 105–106 °C; IR cm-1: 2931 (OH), 1631 (CO); 1H NMR (CDCl3, 200 MHz, ppm): δ 2.49 (t, 2H, J = 7.4 Hz, CH2), 2.65 (t, 2H, J = 7.4 Hz, CH2), 3.83 (s, 6H, 2 x CH3), 3.84 (s, 3H, CH3), 4.96
M AN U
(s, 2H, CH2), 6.40 (s, 2H, H-2’ and H-6’), 6.43–6.45 (m, 1H, H-1), 7.27–7.29 (m, 1H, H-3), 7.47– 7.50 (s, 1H, CH), 14.35 (s, 1H, OH); 13C NMR (CDCl3, 50 MHz, ppm): δ 21.1 (t), 25.9 (t), 54.3 (t), 56.2 (2 x q), 60.9 (q), 104.7 (2 x d), 109.4 (s), 117.3 (d), 121.0 (s), 122.5 (d), 125.9 (s), 131.7 (s), 137.8 (s), 153.6 (2 x s), 165.3 (d), 187.0 (s). Anal calcd for C19H21NO5 (343.37): C 66.46, H 6.16, N
TE D
4.08. Found: C 66.30, H 5.95, N 4.20.
4.1.4.8 5-(hydroxymethylidene)-2-(4-methoxyphenyl)-2,5,6,7-tetrahydro-4H-isoindol-4-one (9m). This compound was obtained from reaction of 8m. Light brown solid; yield 97%; Rf=0.38
EP
(CH2Cl2); m.p.: 126–127 °C; IR cm-1: 2928 (OH), 1635 (CO); 1H NMR (CDCl3, 200 MHz, ppm): δ
AC C
2.54 (t, 2H, J = 6.0 Hz, CH2), 2.73 (t, 2H, J = 6.0 Hz, CH2), 3.84 (s, 3H, CH3), 6.75–6.73 (m, 1H, H-1), 6.96 (d, 2H, J = 9.0 Hz, H-3’ and H-5’), 7.31 (d, 2H, J = 9.0 Hz, H-2’ and H-6’), 7.54–7.59 (m, 2H, H-3 and CH), 14.40 (bs, 1H, OH);
13
C NMR (CDCl3, 50 MHz, ppm): δ 21.0 (t), 25.7 (t),
55.6 (q), 109.4 (s), 114.8 (2 x d), 116.4 (d), 120.6 (d), 122.1 (s), 122.4 (2 x d), 126.4 (s), 133.3 (s), 158.5 (s), 166.4 (d), 186.5 (s). Anal calcd for C16H15NO3 (269.30): C 71.36, H 5.61, N 5.20. Found: C 71.49, H 5.72, N 5.07. 4.1.5 General procedure for the Synthesis of [1,2]oxazolo[5,4-e]isoindoles (6). To a solution of the suitable hydroxymethylideneketones 9 (5.0 mmol) in ethanol (15 mL) hydroxylamine hydrochloride (0.38 g, 5.5 mmol) was added and the reaction mixture was heated
ACCEPTED MANUSCRIPT under reflux for 50 min. After cooling, the solvent was evaporated at reduced pressure. The crude product was purified by chromatography column using dichloromethane as eluent. 4.1.5.1 7-Methyl-5,7-dihydro-4H-[1,2]oxazolo[5,4-e]isoindole (6a). This compound was obtained from reaction of 9a. Yellow oil; yield 64%; Rf=0.19 (CH2Cl2); 1H NMR (DMSO-d6, 200 MHz,
RI PT
ppm): δ 2.60–2.68 (m, 4H, 2 x CH2), 3.62 (s, 3H, CH3), 6.64 (s, 1H, H-6), 7.08 (s, 1H, H-8), 8.36 (s, 1H, H-3); 13C NMR (DMSO-d6, 50 MHz, ppm): δ 20.0 (t), 20.5 (t), 36.3 (q), 108.6 (s), 110.3 (s),
5.79, N 16.08. Found: C 69.16, H 5.43, N 16.21.
SC
116.8 (d), 119.7 (d), 120.4 (s), 149.5 (d), 163.1 (s). Anal calcd for C10H10N2O (174.20): C 68.95, H
4.1.5.2 7-Benzyl-5,7-dihydro-4H-[1,2]oxazolo[5,4-e]isoindole (6b). This compound was obtained
M AN U
from reaction of 9b. White solid; yield 64%; Rf=0.36 (CH2Cl2); m.p.: 88–89 °C; 1H NMR (DMSOd6, 200 MHz, ppm): δ 2.55–2.80 (m, 4H, 2 x CH2), 5.10 (s, 2H, CH2), 6.76 (s, 1H, H-6), 7.26–7.39 (m, 6H, H-8 and Ar), 8.37 (s, 1H, H-3); 13C NMR (DMSO-d6, 50 MHz, ppm): δ 19.4 (t), 20.0 (t), 52.4 (t), 108.3 (s), 110.0 (s), 115.7 (d), 118.4 (d), 120.2 (s), 127.4 (2 x d), 127.5 (d), 128.6 (2 x d),
TE D
138.4 (s), 149.0 (d), 162.5 (s). Anal calcd for C16H14N2O (250.30): C 76.78, H 5.64, N 11.19. Found: C 76.52, H 5.35, N 11.50.
4.1.5.3 7-(4-Methylbenzyl)-5,7-dihydro-4H-[1,2]oxazolo[5,4-e]isoindole (6c). This compound was
EP
obtained from reaction of 9c. White solid; yield72%; Rf=0.38 (CH2Cl2); m.p.: 141–142 °C; 1H
AC C
NMR (DMSO-d6, 200 MHz, ppm): δ 2.27 (s, 3H, CH3), 2.63–2.66 (m, 4H, 2 x CH2), 5.03 (s, 2H, CH2), 6.73 (s, 1H, H-6), 7.08 (d, 2H, J = 8.6 Hz, H-3’ and H-5’), 7.20 (d, 2H, J = 8.6 Hz, H-2’ and H-6’), 7.22 (s, 1H, H-8), 8.36 (s, 1H, H-3); 13C NMR (DMSO-d6, 50 MHz, ppm): δ 19.4 (t), 20.1 (t), 20.6 (q), 52.2 (t), 108.3 (s), 110.0 (s), 115.6 (d), 118.3 (d), 120.1 (s), 127.5 (2 x d), 129.1 (2 x d), 135.3 (s), 136.8 (s), 149.0 (d), 162.5 (s). Anal calcd for C17H16N2O (264.32): C 77.25, H 6.10, N 10.60. Found: C 77.52, H 6.40, N 10.38. 4.1.5.4 7-(4-Methoxybenzyl)-5,7-dihydro-4H-[1,2]oxazolo[5,4-e]isoindole (6d). This compound was obtained from reaction of 9d. White solid; yield 60%; Rf=0.39 (CH2Cl2); m.p.: 102–103 °C; 1H
ACCEPTED MANUSCRIPT (DMSO-d6, 200 MHz, ppm): δ 2.50–2.64 (m, 4H, 2 x CH2), 3.73 (s, 3H, CH3), 5.00 (s, 2H, CH2), 6.73 (s, 1H, H-6), 6.91 (d, 2H, J = 8.5 Hz, H-3’ and H-5’), 7.21 (s, 1H, H-8), 7.25 (d, 2H, J = 8.5 Hz, H-2’ and H-6’), 8.36 (s, 1H, H-3);
C NMR (DMSO-d6, 50 MHz, ppm): δ 19.4 (t), 20.1 (t),
13
51.9 (t), 55.0 (q), 108.3 (s), 110.0 (s), 113.9 (2 x d), 115.4 (d), 118.2 (d), 120.1 (s), 129.0 (2 x d),
RI PT
130.3 (s), 149.0 (d), 158.7 (s), 162.5 (s). Anal calcd for C17H16N2O2 (280.32): C 72.84, H 5.75, N 9.99. Found: C 72.57, H 5.93, N 9.69.
4.1.5.5 7-(3-Methoxybenzyl)-5,7-dihydro-4H-[1,2]oxazolo[5,4-e]isoindole (6e). This compound
SC
was obtained from reaction of 9e. White solid; yield 78%; Rf=0.46 (CH2Cl2); m.p.: 83–84 °C; 1H (DMSO-d6, 200 MHz, ppm): δ 2.64–2.67 (m, 4H, 2 x CH2), 3.73 (s, 3H, CH3), 5.05 (s, 2H, CH2),
M AN U
6.77 (s, 1H, H-6), 6.81–6.87 (m, 3H, Ar), 7.22–7.30 (m, 2H, H-8 and Ar), 8.37 (s, 1H, H-3);
13
C
NMR (DMSO-d6, 50 MHz, ppm): δ 19.4 (t), 20.0 (t), 52.3 (t), 55.0 (q), 108.3 (s), 110.0 (s), 112.7 (d), 113.3 (d), 115.7 (d), 118.4 (d), 119.6 (d), 120.1 (s), 129.7 (d), 139.9 (s), 149.0 (d), 159.4 (s), 162.5 (s). Anal calcd for C17H16N2O2 (280.32): C 72.84, H 5.75, N 9.99. Found: C 72.69, H 5.87, N
TE D
9.71.
4.1.5.6 7-(2-Methoxybenzyl)-5,7-dihydro-4H-[1,2]oxazolo[5,4-e]isoindole (6f). This compound was obtained from reaction of 9f. White solid; yield 75%; Rf=0.39 (CH2Cl2); m.p.: 126–127 °C; 1H
EP
NMR (DMSO-d6, 200 MHz, ppm): δ 2.64–2.67 (m, 4H, 2 x CH2), 3.84 (s, 3H, CH3), 5.05 (s, 2H,
AC C
CH2), 6.73 (s, 1H, H-6), 6.87–7.05 (m, 3H, Ar), 7.19 (s, 1H, H-8), 7.25–7.34 (m, 1H, Ar), 8.37 (s, 1H, H-3);
13
C NMR (DMSO-d6, 50 MHz, ppm): δ 19.4 (t), 20.1 (t), 47.5 (t), 55.4 (q), 108.3 (s),
109.8 (s), 110.8 (d), 115.8 (d), 118.6 (d), 119.9 (s), 120.4 (d), 126.1 (s), 128.6 (d), 129.2 (d), 149.1 (d), 156.4 (s), 162.5 (s). Anal calcd for C17H16N2O2 (280.32): C 72.84, H 5.75, N 9.99. Found: C 73.02, H 5.47, N 10.08. 4.1.5.7
7-(2,3-Dimethoxybenzyl)-5,7-dihydro-4H-[1,2]oxazolo[5,4-e]isoindole
(6g).
This
compound was obtained from reaction of 9g. White solid; yield 65%; Rf=0.29 (CH2Cl2); m.p.: 131– 132 °C; 1H NMR (DMSO-d6, 200 MHz, ppm): δ 2.60–2.70 (m, 4H, 2 x CH2), 3.73 (s, 3H, CH3),
ACCEPTED MANUSCRIPT 3.80 (s, 3H, CH3), 5.07 (s, 2H, CH2), 6.67–6.74 (m, 2H, H-6 and Ar), 6.99–7.08 (m, 2H, Ar), 7.19 (s, 1H, H-8), 8.37 (s, 1H, H-3); 13C NMR (DMSO-d6, 50 MHz, ppm): δ 19.4 (t), 20.0 (t), 47.5 (t), 55.7 (q), 60.1 (q), 108.3 (s), 109.8 (s), 112.6 (d), 115.8 (d), 118.5 (d), 119.9 (s), 120.5 (d), 124.1 (d), 131.5 (s), 146.1 (s), 149.1 (d), 152.3 (s), 162.5 (s). Anal calcd for C18H18N2O3 (310.35): C 69.66, H
4.1.5.8
RI PT
5.85, N 9.03. Found: C 69.50, H 5.99, N 9.15.
7-(2,5-Dimethoxybenzyl)-5,7-dihydro-4H-[1,2]oxazolo[5,4-e]isoindole
(6h).
This
compound was obtained from reaction of 9h. White solid; yield 75%; Rf=0.35 (CH2Cl2); m.p.: 119–
SC
120 °C; 1H NMR (DMSO-d6, 200 MHz, ppm): δ 2.60–2.69 (m, 4H, 2 x CH2), 3.66 (s, 3H, CH3), 3.79 (s, 3H, CH3), 5.01 (s, 2H, CH2), 6.65 (s, 1H, H-6), 6.72–6.75 (m, 1H, Ar), 6.81–6.87 (m, 1H, 13
M AN U
Ar), 6.93–6.97 (m, 1H, Ar), 7.19 (s, 1H, H-8), 8.37 (s, 1H, H-3);
C NMR (DMSO-d6, 50 MHz,
ppm): δ 19.4 (t), 20.1 (t), 47.5 (t), 55.3 (q), 55.8 (q), 108.3 (s), 109.8 (s), 111.7 (d), 112.8 (d), 115.4 (d), 115.8 (d), 118.5 (d), 119.9 (s), 127.2 (s), 149.1 (d), 150.6 (s), 153.1 (s), 162.5 (s). Anal calcd for C18H18N2O3 (310.35): C 69.66, H 5.85, N 9.03. Found: C 69.55, H 5.98, N 9.21.
TE D
4.1.5.9 7-(3,4-Dimethoxybenzyl)-5,7-dihydro-4H-[1,2]oxazolo[5,4-e]isoindole (6i). This compound was obtained from reaction of 9i. Yellow oil; yield 65%; Rf=0.23 (CH2Cl2); 1H NMR (DMSO-d6, 200 MHz, ppm): δ 2.59–2.66 (m, 4H, 2 x CH2), 3.72 (s, 3H, CH3), 3.74 (s, 3H, CH3), 4.99 (s, 2H,
EP
CH2), 6.76 (s, 1H, H-6), 6.80–6.94 (m, 2H, H-5’ and H-6’), 7.01 (s, 1H, H-2’), 7.25 (s, 1H, H-8),
AC C
8.37 (s, 1H, H-3); 13C NMR (DMSO-d6, 50 MHz, ppm): δ 19.4 (t), 20.1 (t), 52.2 (t), 55.4 (q), 55.5 (q), 108.2 (s), 109.8 (s), 111.7 (d), 111.8 (d), 115.5 (d), 118.2 (d), 120.0 (d), 120.1 (s), 130.6 (s), 148.3 (s), 148.6 (s), 149.0 (d), 162.6 (s). Anal calcd for C18H18N2O3 (310.35): C 69.66, H 5.85, N 9.03. Found: C 69.74, H 5.61, N 8.84. 4.1.5.10
7-(3,5-Dimethoxybenzyl)-5,7-dihydro-4H-[1,2]oxazolo[5,4-e]isoindole
(6j).
This
compound was obtained from reaction of 9j. Yellow oil; yield 74%; Rf=0.28 (CH2Cl2); 1H NMR (DMSO-d6, 200 MHz, ppm): δ 2.64–2.67 (m, 4H, 2 x CH2), 3.72 (s, 6H, 2 x CH3), 5.00 (s, 2H, CH2), 6.42–6.46 (m, 3H, Ar), 6.77 (s, 1H, H-6), 7.26 (s, 1H, H-8), 8.37 (s, 1H, H-3);
13
C NMR
ACCEPTED MANUSCRIPT (DMSO-d6, 50 MHz, ppm): δ 19.4 (t), 20.0 (t), 52.4 (t), 55.2 (2 x q), 98.9 (d), 105.7 (2 x d), 108.3 (s), 109.9 (s), 115.8 (d), 118.5 (d), 120.1 (s), 140.6 (s), 149.1 (d), 160.6 (2 x s), 162.5 (s). Anal calcd for C18H18N2O3 (310.35): C 69.66, H 5.85, N 9.03. Found: C 69.80, H 5.55, N 8.79. 4.1.5.11 7-(3,4,5-Trimethoxybenzyl)-5,7-dihydro-4H-[1,2]oxazolo[5,4-e] isoindole (6k). This
RI PT
compound was obtained from reaction of 9k. White solid; yield 61%; Rf=0.13 (CH2Cl2); m.p.: 77– 78 °C; 1H NMR (DMSO-d6, 200 MHz, ppm): δ 2.65 (s, 4H, 2 x CH2), 3.63 (s, 3H, CH3), 3.76 (s, 6H, 2 x CH3), 4.98 (s, 2H, CH2), 6.71 (s, 2H, H-2’ and H-6’), 6.81 (s, 1H, H-6), 7.29 (s, 1H, H-8), 13
C NMR (DMSO-d6, 50 MHz, ppm): δ 19.4 (t), 20.1 (t), 52.7 (t), 55.9 (2 x q),
SC
8.37 (s, 1H, H-3);
59.9 (q), 105.4 (2 x d), 108.3 (s), 109.9 (s), 115.6 (d), 118.3 (d), 120.0 (s), 133.8 (s), 136.9 (s),
Found: C 66.92, H 5.84, N 8.39.
M AN U
149.1 (d), 152.9 (2 x s), 162.5 (s). Anal calcd for C19H20N2O4 (340.37): C 67.05, H 5.92, N 8.23.
4.1.5.12 7-Phenyl-5,7-dihydro-4H-[1,2]oxazolo[5,4-e]isoindole (6l). This compound was obtained from reaction of 9l. White solid; yield 58%; Rf=0.48 (CH2Cl2); m.p.: 123–124 °C; 1H NMR
1H, H-8), 8.47 (s, 1H, H-3);
TE D
(DMSO-d6, 200 MHz, ppm): δ 2.69–2.82 (m, 4H, 2 x CH2), 7.24–7.67 (m, 6H, H-6 and Ar), 7.80 (s, 13
C NMR (DMSO-d6, 50 MHz, ppm): δ 19.8 (t), 20.5 (t), 110.3 (s),
EP
112.7 (s), 113.7 (d), 117.0 (d), 119.8 (2 x d), 122.7 (s), 126.1 (d), 130.2 (2 x d), 139.9 (s), 149.8 (d), 162.2 (s). Anal calcd for C15H12N2O (236.27): C 76.25, H 5.12, N 11.86. Found: C 76.03, H 5.39, N
AC C
11.58.
4.1.5.13 7-(4-Methoxyphenyl)-5,7-dihydro-4H-[1,2]oxazolo[5,4-e]isoindole (6m). This compound was obtained from reaction of 9m. White solid; yield 62%; Rf=0.33 (CH2Cl2); m.p.: 134–135 °C; 1H NMR (DMSO-d6, 200 MHz, ppm): δ 2.68–2.81 (m, 4H, 2 x CH2), 3.79 (s, 3H, CH3), 7.03 (d, 2H, J = 9.0 Hz, H-3’ and H-5’), 7.25 (s, 1H, H-6), 7.55 (d, 2H, J = 9.0 Hz, H-2’ and H-6’), 7.67 (s, 1H, H8), 8.45 (s, 1H, H-3); 13C NMR (DMSO-d6, 50 MHz, ppm): δ 19.3 (t), 20.0 (t), 55.4 (q), 109.4 (s), 111.8 (s), 113.4 (d), 114.7 (2 x d), 116.8 (d), 120.9 (2 x d), 121.7 (s), 133.0 (s), 149.2 (d), 157.2 (s),
ACCEPTED MANUSCRIPT 161.9 (s). Anal calcd for C16H14N2O2 (266.29): C 72.16, H 5.30, N 10.52. Found: C 72.32, H 5.19, N 10.38. 4.1.5.14 Ethyl 7,8-dimethyl-5,7-dihydro-4H-[1,2]oxazolo[5,4-e]isoindol-6-carboxylate (6n). This compound was obtained from reaction of 9n. White solid; yield 72%; Rf=0.35 (CH2Cl2); m.p.: 84–
RI PT
85 °C; IR cm-1: 1689 (CO); 1H NMR (DMSO-d6, 200 MHz, ppm): δ 1.30 (t, 3H, J = 7.1 Hz, CH3), 2.46 (s, 3H, CH3), 2.69 (t, 2H, J = 8.1 Hz, CH2), 3.01 (t, 2H, J = 8.1 Hz, CH2), 3.76 (s, 3H, CH3), 4.23 (q, 2H, J = 7.1 Hz, CH2), 8.42 (s, 1H, H-3); 13C NMR (DMSO-d6, 50 MHz, ppm): δ 11.2 (q),
SC
13.5 (q), 18.6 (t), 21.6 (t), 32.5 (q), 59.6 (t), 108.3 (s), 109.0 (s), 118.5 (s), 128.3 (s), 131.4 (s),
Found: C 64.93, H 6.47, N 10.60.
M AN U
148.8 (d), 160.8 (s), 162.0 (s). Anal calcd for C14H16N2O3 (260.29): C 64.60, H 6.20, N 10.76.
4.1.5.15 Ethyl 7-benzyl-8-methyl-5,7-dihydro-4H-[1,2]oxazolo[5,4-e]isoindol-6-carboxylate (6o). This compound was obtained from reaction of 9o. White solid; yield 65%; Rf=0.45 (CH2Cl2); m.p.: 120–121 °C; IR cm-1: 1687 (CO); 1H NMR (DMSO-d6, 200 MHz, ppm): δ 1.23 (t, 3H, J = 7.1 Hz,
TE D
CH3), 2.41 (s, 3H, CH3), 2.74 (t, 2H, J = 8.1 Hz, CH2), 3.09 (t, 2H, J = 8.1 Hz, CH2), 4.17 (q, 2H, J = 7.1 Hz, CH2), 5.62 (s, 2H, CH2), 6.97 (d, 2H, J = 6.6 Hz, H-2’ and H-6’), 7.20–7.37 (m, 3H, H-3’, H-4’ and H-5’), 8.46 (s, 1H, H-3); 13C NMR (DMSO-d6, 50 MHz, ppm): δ 11.7 (q), 14.6 (q), 19.1
EP
(t), 22.2 (t), 48.1 (t), 60.2 (t), 109.3 (s), 110.2 (s), 118.8 (s), 126.2 (2 x d), 127.4 (d), 129.1 (2 x d),
AC C
129.6 (s), 132.0 (s), 138.5 (s), 149.4 (d), 161.1 (s), 162.3 (s). Anal calcd for C20H20N2O3 (336.38): C 71.41, H 5.99, N 8.33. Found: C 71.75, H 6.23, N 8.12. 4.1.5.16
Ethyl
7-(4-methoxybenzyl)-8-methyl-5,7-dihydro-4H-[1,2]oxazolo[5,4-e]isoindol-6-
carboxylate (6p). This compound was obtained from reaction of 9p. White solid; yield 63%; Rf=0.35 (CH2Cl2); m.p.: 102–103 °C; IR cm-1: 1689 (CO); 1H NMR (DMSO-d6, 200 MHz, ppm): δ 1.25 (t, 3H, J = 7.1 Hz, CH3), 2.42 (s, 3H, CH3), 2.73 (t, 2H, J = 7.8 Hz, CH2), 3.07 (t, 2H, J = 7.8 Hz, CH2), 3.71 (s, 3H, CH3), 4.19 (q, 2H, J = 7.1 Hz, CH2), 5.54 (s, 2H, CH2), 6.86 (d, 2H, J = 8.0 Hz, H-3’ and H-5’), 6.94 (d, 2H, J = 8.0 Hz, H-2’ and H-6’), 8.45 (s, 1H, H-3); 13C NMR (DMSOd6, 50 MHz, ppm): δ 11.3 (q), 14.1 (q), 18.6 (t), 21.7 (t), 47.0 (t), 55.0 (q), 59.7 (t), 108.7 (s), 109.7
ACCEPTED MANUSCRIPT (s), 114.0 (2 x d), 118.2 (s), 127.2 (2 x d), 129.1 (s), 129.8 (s), 131.5 (s), 148.9 (d), 158.2 (s), 160.7 (s), 161.8 (s). Anal calcd for C21H22N2O4 (366.41): C 68.84, H 6.05, N 7.65. Found: C 68.99, H 5.81, N 7.39. 4.1.5.17 7,8-Dimethyl-5,7-dihydro-4H-[1,2]oxazolo[5,4-e]isoindole (6q). This compound was
RI PT
obtained from reaction of 9q. Yellow oil; yield 58%; Rf=0.42 (CH2Cl2); 1H NMR (DMSO-d6, 200 MHz, ppm): δ 2.37 (s, 3H, CH3), 2.55–2.72 (m, 4H, 2 x CH2), 3.48 (s, 3H, CH3), 6.55 (s, 1H, H-6), 8.34 (s, 1H, H-3);
13
C NMR (DMSO-d6, 50 MHz, ppm): δ 11.2 (q), 20.1 (t), 20.7 (t), 33.58 (q),
SC
107.9 (s), 108.5 (s), 118.3 (s), 118.7 (d), 124.7 (s), 149.2 (d), 164.0 (s). Anal calcd for C11H12N2O (188.23): C 70.19, H 6.43, N 14.88. Found: C 70.07, H 6.64, N 14.54.
M AN U
4.1.5.18 7-Benzyl-8-methyl-5,7-dihydro-4H-[1,2]oxazolo[5,4-e]isoindole (6r). This compound was obtained from reaction of 9r. White solid; yield 56%; Rf=0.53 (CH2Cl2); m.p.: 98–99 °C; 1H NMR (DMSO-d6, 200 MHz, ppm): δ 2.33 (s, 3H, CH3), 2.60–2.82 (m, 4H, 2 x CH2), 5.09 (s, 2H, CH2), 6.71 (s, 1H, H-6), 7.12 (d, 2H, J = 6.8 Hz, H-2’ and H-6’), 7.26–7.38 (m, 3H, H-3’, H-4’ and H-5’),
TE D
8.36 (s, 1H, H-3); 13C NMR (DMSO-d6, 50 MHz, ppm): δ 11.4 (q), 20.0 (t), 20.7 (t), 49.8 (t), 108.3 (s), 109.1 (s), 118.6 (d), 118.9 (s), 124.4 (s), 127.2 (2 x d), 127.8 (d), 129.1 (2 x d), 138.7 (s), 149.3
AC C
6.33, N 10.28.
EP
(d), 163.8 (s). Anal calcd for C17H16N2O (264.32): C 77.25, H 6.10, N 10.60. Found: C 77.59, H
4.2. Biology
4.2.1. Drugs.
For in vitro studies, vinorelbine (purchased from Pierre Fabre Pharma), taxol (Paclitaxel; purchased from Santa Cruz Biotechnology), Purvalanol A (Tocris) and 6j were completely dissolved in 1% dimethylsulfoxide (DMSO), stored at -20°C, and then diluted in complete culture medium immediately before use at the appropriate concentration. For in vivo studies, 6j was dissolved in DMSO 10% and diluted in saline solution.
ACCEPTED MANUSCRIPT 4.2.2. Cell Culture and In Vivo Experiments. The Mycoplasma-free melanoma (JR8 and M14), breast cancer (MCF-7 and MDA-MB-231), castration-resistant prostate carcinoma (PC3 and DU145) and DMPM (STO and MP8), and the normal human lung fibroblast (WI38) and adult breast (MCF10A) cell lines were maintained in the
RI PT
logarithmic growth phase as a monolayer in the appropriate culture medium (Lonza): RPMI-1640 medium supplemented with 10% fetal bovine serum (FBS; JR8, M14, PC3 and DU145), DMEMF12 supplemented with 5% (MCF-7 and MDA-MB-231) or 10% (STO and MP8) FBS, or DMEM
with a supply of 5% CO2/95% air atmosphere.
SC
supplemented with 10% FBS (WI38), or in MEBM (MCF10A), in a humidified incubator at 37°C
M AN U
For in vivo experiments, female nude mice (6-8 weeks-old, purchased from Charles River) were maintained in laminar flow rooms keeping temperature and humidity constant. The mice had free access to food and water. Experimental protocols were approved by the Ethics Committee for Animal Experimentation of the Fondazione IRCCS Istituto Nazionale Tumori of Milan according to
TE D
institutional guidelines that are in compliance with national and international laws and policies. Exponentially growing STO cells were subcutaneously injected into the right flank mice (107 cells/flank). Eight mice for each experimental group were used. 6j was dissolved in a mixture of
EP
DMSO (10%) and saline solution (90%) and delivered i.p. every seven days for four weeks (q7dx4) starting from the tenth day after cell inoculum at a dose of 5 mg/kg. Tumor growth was followed by
AC C
weekly measurements of tumor diameters with a Vernier caliper and tumor volume (TV) was calculated according to the formula: TV (mm3) = d2 × D/2 where d and D are the shortest and the longest diameter, respectively. The efficacy of the drug treatment was assessed as tumor volume inhibition percentage (TVI %) in treated versus control mice, calculated as TVI% = 100 − (mean TV treated/mean TV control × 100). The toxicity of the drug treatment was determined as body weight loss and lethal toxicity. 4.2.3. Cell Proliferation Assay.
ACCEPTED MANUSCRIPT After harvesting in the logarithmic growth phase, 4500 cells/50 µL were plated in 96-well flatbottomed microtiter plates (EuroClone) for 24 h and then treated with increasing concentrations of 6j (0.01-100 µM) for 72 h. Control cells received vehicle alone (0.01% DMSO). Studies were performed in eight replicates and repeated at least three times independently. At the end of drug
RI PT
exposure, the antiproliferative potential was determined with the CellTiter 96 AQueous One Solution Cell Proliferation Assay (MTS, purchased from Promega) according to the manufacturer’s protocols. Optical density was read at 490 nm on a microplate reader (POLARstar OPTIMA, BMG
SC
Labtech GmbH), and the results were expressed as a percentage, relative to DMSO-treated cells. Dose-response curves were created, and IC50 and IC80 values (i.e., concentrations able to inhibit cell
compound. 4.2.4. Tubulin polymerization assays.
M AN U
growth by 50% and 80%, respectively) were determined graphically from the curve for each
Cells were seeded in 6 wells plates (Eppendorf) and were exposed the next day to the 15r
TE D
derivative for 24 h at concentrations corresponding to the IC50 at 72 h. Samples were then processed for the tubulin polymerization assay [67,68]. To separate cytosolic and cytoskeletal-associated
EP
proteins, cells were rinsed twice in PIPES-EGTA-MgCl2 (PEM) buffer (85 mmol/L PIPES, pH 6.94; 10 mmol/L EGTA; 1 mmol/L MgCl2; 2 mol//L glycerol; 1 mmol/L phenylmethylsulfonyl
AC C
fluoride; 0.1 mmol/L leupeptin; 1 µmol/L pepstatin; 2 µg/mL aprotinin), lysed at room temperature for 10 min with PEM buffer supplemented with 0.1% v/v Triton X-100, and rinsed in PEM buffer. The Triton X-100-soluble fractions were then diluted 3:1 with 4 × SDS-PAGE sample buffer. The insoluble material that remained attached to the dish was scraped into SDS-PAGE sample buffer containing protease inhibitors. Proteins were separated by SDS-PAGE, and tubulin distribution was analyzed by immunoblotting using anti α-tubulin antibody (Sigma-Aldrich). 4.2.5. Cell cycle distribution analysis.
ACCEPTED MANUSCRIPT Both adherent and floating cells were fixed in 70% EtOH and incubated at 4°C for 30 min in staining solution containing 50 µg/mL of propidium iodide, 50 mg/mL of RNase, and 0.05% Nonidet-P40 in PBS. Samples were analyzed with BD Accuri c6 flow cytometer (Becton
software according to the Modfit model (Becton Dickinson). 4.2.6. Fluorescence microscopy analysis
RI PT
Dickinson). At least 30000 events were read, and histograms were analyzed using the CellQuest
Cells seeded on glass cover-slips were treated for 72 h with equitoxic (IC50) concentrations of
SC
6j, taxol and vinorelbine, and then fixed in 2% paraformaldehyde for 30 minutes and permeabilized
M AN U
with ice-cold methanol for 20 minutes at -20°C. After blocking with PBS containing 1% bovine serum albumin, cells were probed overnight with a mouse anti-MPM-2 (mitotic protein monoclonal #2; Upstate Biotechnology) at room temperature, followed by a 1-h incubation at room temperature with goat antimouse AlexaFluor594® dye secondary antibody (Life Technologies). Images were acquired using a Leika fluorescence microscope (Leica). The percentage of mitotic cells was
TE D
determined by scoring at least 300 cells for each sample run in duplicate. 4.2.7. Apoptosis analysis. The catalytic activity of caspase-3 was measured in the same cellular
EP
samples as the ability to cleave the specific substrate N-acetyl-Asp-Glu-Val-Asp-pNA (DEVDpNA) by means of the APOPCYTO/caspase-3 kit (MBL), according to manufacturer's instructions.
AC C
Briefly, cells were washed, pelleted, and lyzed according to the manufacturer's instructions. Total protein and the specific fluorogenic substrate N-acetyl-Asp-Glu-Val-Asp-pNA (DEVD-pNA) were mixed for 1 h at 37 °C and transferred to 96-well microtiter plates. The hydrolysis of the specific substrates was monitored by a spectrofluorometer (POLARstar OPTIMA) with 380-nm excitation and 460-nm emission filters. Results were expressed as relative fluorescence units (rfu). 4.2.8. In Vitro Kinase Assay. The effect of 6j on CDK1 activity was evaluated using the OmniaTM Recombinant Kit (Invitrogen) according to the manufacturer’s protocols. Briefly, increasing concentrations (0.1-100 µM) of 6j and Purvalanol A were mixed with recombinant CDK1/cyclin B
ACCEPTED MANUSCRIPT complex (Invitrogen) in 1× kinase buffer containing a peptide substrate, ATP, and dithiothreitol, and the kinase reaction was performed at 30°C for 30 min. Fluorescences were measured upon excitation at 360 nm and emission at 485 nm.
Acknowledgements
SC
test. Ps < 0.05 was considered statistically significant.
RI PT
4.2.9. Statistical Analysis. Statistical evaluation of data was done with the two-tailed Student’s t
This work was financially supported by Ministero dell’Istruzione dell’Università e della Ricerca
AC C
EP
TE D
M AN U
(MIUR). DC is recipient of an AIRC fellowship (#16360).
ACCEPTED MANUSCRIPT
5. References [1]
A. Carbone, B. Parrino, G. Di Vita, A. Attanzio, V. Spanò, A. Montalbano, P. Barraja, L.
thiazolyl-bis-pyrrolo[2,3-b]pyridines
and
indolyl-thiazolyl-pyrrolo[2,3-c]pyridines,
nortopsentin analogues, Marine Drugs 13 (2015) 460−492. [2]
RI PT
Tesoriere, M. A. Livrea, P. Diana, G. Cirrincione, Synthesis and antiproliferative activity of
V. Spanò, B. Parrino, A. Carbone, A. Montalbano, A. Salvador, P. Brun, D. Vedaldi, P.
SC
Diana, G. Cirrincione, P. Barraja, Pyrazolo[3,4-h]quinolines promising photosensitizing
[3]
M AN U
agents in the treatment of cancer, Eur. J. Med. Chem. 102 (2015) 334−351. B. Parrino, A. Carbone, M. Muscarella, V. Spanò, A. Montalbano, P. Barraja, A. Salvador, D. Vedaldi, G. Cirrincione, P. Diana, 11H‑Pyrido[3′,2′:4,5]pyrrolo[3,2‑c]cinnoline and Pyrido[3′,2′:4,5]pyrrolo[1,2‑c][1,2,3]benzotriazine: two new ring systems with antitumor
[4]
TE D
activity, J. Med. Chem. 57 (2014) 9495−9511.
A. Carbone, M. Pennati, P. Barraja, A. Montalbano, B. Parrino, V. Spanò, A. Lopergolo, S. Sbarra, V. Doldi, N. Zaffaroni, G. Cirrincione, P. Diana, Synthesis and antiproliferative of
substituted
3[2-(1H-indol-3-yl)-1,3-thiazol-4-yl]-1H-pyrrolo[3,2-b]pyridines,
EP
activity
marine alkaloid nortopsentin analogues, Curr. Med. Chem. 21 (2014) 1654−1666. A. Lauria, P. Diana, P. Barraja, A. M. Almerico, G. Cirrincione, G. Dattolo, Pyrrolo[3,4-
AC C
[5]
e][1,2,3]triazolo[1,5-a]pyrimidine and pyrrolo[3,4-d][1,2,3]triazolo[1,5-a]pyrimidine. New tricyclic ring systems of biological interest, J. Heterocycl. Chem. 37 (2000) 747−750. [6]
C. Dumontet, M. A. Jordan, Microtubule-binding agents: a dynamic field of cancer therapeutics, Nat. Rev. Drug Discov. 9 (2010) 790–803.
[7]
K. Nepali, R. Ojha, H. Y. Lee, J. P. Liou, Early investigational tubulin inhibitors as novel cancer therapeutics, Expert Opin. Investig. Drugs 25 (2016) 917–936.
ACCEPTED MANUSCRIPT [8]
S. Florian, T. J. Mitchison, Anti-Microtubule Drugs, Methods Mol. Biol. 1413 (2016) 403– 421.
[9]
G. A. Orr, P. Verdier-Pinard, H. McDaid, S. B. Horwitz, Mechanisms of Taxol resistance related to microtubules, Oncogene 22 (2003) –7295.
RI PT
[10] R. Kaur, G. Kaur, R. K. Gill, R. Soni, J. Bariwal, Recent developments in tubulin polymerization inhibitors: An overview, Eur. J. Med. Chem. 87 (2014) 89–124.
[11] L. M. Greene, M. J. Meegan, D. M. Zisterer, Combretastatins: more than just vascular
SC
targeting agents?, J. Pharmacol. Exp. Ther. 355 (2015) 212–227.
[12] G. R. Pettit, J. W., Lippert III; D. L. Herald, E. Hamel, R. K. Pittit, Antineoplastic agents
M AN U
440. Asymmetric synthesis and evaluation of the combretastatin A-1 SAR probes (1S,2S)and
(1R,2R)1,2-dihydroxy-1-(2′,3′′-dihydroxy-4′-methoxyphenyl)-2-
(3′′,4′′,5′′trimehoxyphenyl)ethane, J. Nat. Prod. 63 (2000) 969–974. [13] K. Ohsumi, R. Nakagawa, Y. Fukuda, T. Hatanaka, Y. Morinaga, Y. Nihei, K. Ohishi, Y.
TE D
Suga, Y. Akiyama, T. Tsuji, Novel Combretastatin analogues effective against murine solid tumors: design and structure-activity relationships, J. Med. Chem. 41 (1998) 3022–3032. [14] G. R. Pettit, M. R. Rhodes. D. L. Herald, D. J. Chaplin, M. R. L. Stratford, E. Hamel, R. K.
EP
Pettit, J.-C. Chapuis, D. Oliva, Antineoplastic agents 393. Synthesis of the trans isomer of combretastatin A-4 prodrug, Anti-Cancer Drug Des. 13 (1998) 981–993.
AC C
[15] R. Kaur, G. Kaur, R. K.Gill, R. Soni, J. Bariwal, Recent developments in tubulin polymerization inhibitors: an overview, Eur. J. Med. Chem. 87 (2014) 89–124. [16] J. Zhou, J. Jin, Y. Zhang, Y. Yin, X. Chen, B. Xu, Synthesis and antiproliferative evaluation of novel benzoimidazole-contained oxazole-bridged analogs of combretastatin A-4, Eur. J. Med. Chem. 68 (2013) 222–232; [17] S. Zheng, Q. Zhong, M.Mottamal, Q. Zhang, C. Zhang, E. LeMelle, H. McFerrin, G. Wang, Design, synthesis, and biological evaluation of novel pyridine-bridged analogues of Combretastatin-A4 as anticancer agents, J. Med. Chem. 57 (2014) 3369−3381.
ACCEPTED MANUSCRIPT [18] L. Wang, K. W. Woods, Q. Li, K. J. Barr, R. W. McCroskey, S. M. Hannick, L. Gherke, R. B. Credo, Y.-H. Hui, K. Marsh, R. Warner, J. Y. Lee, N. Zielinski-Mozng, D. Frost, S. H. Rosenberg, H. L. Sham, Potent, orally active heterocycle-based Combretastatin A-4
activity evaluation, J. Med. Chem. 45 (2002) 1697−1711.
RI PT
analogues: synthesis, structure-activity relationship, rharmacokinetics, and in vivo antitumor
[19] Q. Li, K. W. Woods, A. Claiborne, S. L. Gwaltney, II, K. J. Barr, G. Liu, L. Gehrke, R. B. Credo, Y. H. Hui, J. Lee, R. B. Warner, P. Kovar, M.A. Nukkala, N. A. Zielinski, S. K. Tahir,
SC
M. Fitzgerald, K. H. Kim, K. Marsh, D. Frost, S.-C. Ng, S. Rosenberg, H, L. Sham, Synthesis and biological evaluation of 2-indolyloxazolines as a new class of tubulin polymerization
M AN U
inhibitors. Discovery of A-289099 as an orally active antitumor agent, Bioorg. Med. Chem. Lett. 12 (2002) 465–469.
[20] A. B. S. Maya, C. Pérez-Melero, N. Salvador, R. Peláez, E. Caballero, M. Medarde, New naphthylcombretastatins. Modifications on the ethylene bridge, Bioorg. Med. Chem. 13
TE D
(2005) 2097–2107.
[21] S. L. Gwaltney II, H. S. Jae, D. M. Kalvin, G. Liu, H. Sham, Q. Li, A. Claiborne, L. Wang, K. J. Barr, K. W. Woods, Oxazoline antiproliferative agents, US Patent 6228868;. Chem. Abstr.
EP
134 (2001) 326523.
[22] K. Yamamoto, K. Noda, A. Yoshimura, M. Fukuoka, K. Furuse, H Niitani, Phase I study of
AC C
E7010, Cancer Chemother. Pharmacol. 42 (1998) 127–134. [23] B. Parrino, A. Carbone, C. Ciancimino, V. Spanò, A. Montalbano, P. Barraja, G. Cirrincione, P. Diana, C. Sissi, M. Palumbo, O. Pinato, M. Pennati, G. Beretta, M. Folini, P. Matyus, B. Balogh,
N.
Zaffaroni,
Water-soluble
isoindolo[2,1-a]quinoxalin-6-imines:
in
vitro
antiproliferative activity and molecular mechanism(s) of action, Eur. J. Med. Chem. 94 (2015) 149–162.
ACCEPTED MANUSCRIPT [24] A. Montalbano, B. Parrino, P. Diana, P. Barraja, A. Carbone, V. Spanò, G. Cirrincione, Synthesis of the new oligopeptide pyrrole derivative isonetropsin and its one pyrrole unit analogue, Tetrahedron 69 (2013) 2550−2554. [25] B. Parrino, C. Ciancimino, A. Carbone, V. Spanò, A. Montalbano, P. Barraja, G. Cirrincione,
RI PT
P. Diana, Synthesis of isoindolo[1,4]benzoxazinone and isoindolo[1,5]benzoxazepine: two new ring systems of pharmaceutical interest, Tetrahedron 71 (2015) 7332−7338.
[26] B. Parrino, V. Spanò, A. Carbone, P. Barraja, P. Diana, G. Cirrincione, A. Montalbano,
deaza analogue, Molecules 19 (2014) 13342−13357.
SC
Synthesis of the new ring system bispyrido[4',3':4,5]pyrrolo[1,2-a:1',2'-d]pyrazine and its
M AN U
[27] B. Parrino, V. Spanò, A. Carbone, A. Montalbano, P. Barraja, P. Màtyus, G. Cirrincione, P. Diana, ‘Interrupted’ diazotization of 3-aminoindoles and 3-aminopyrroles, Tetrahedron 70 (2014) 7318−7321.
[28] B. Parrino, A. Carbone, V. Spanò, A. Montalbano, D. Giallombardo, P. Barraja, A. Attanzio,
TE D
L. Tesoriere, C. Sissi, M. Palumbo, G. Cirrincione, P. Diana, Aza-isoindolo and isoindoloazaquinoxaline derivatives with antiproliferative activity, Eur. J. Med. Chem. 94 (2015) 367−377.
EP
[29] P. Diana, A. Stagno, P. Barraja, A. Montalbano, A. Carbone, B. Parrino, G. Cirrincione, Synthesis of the new ring system pyrrolizino[2,3-b]indol-4(5H)-one, Tetrahedron 67 (2011)
AC C
3374−3379.
[30] S. Mahboobi, H. Pongratz, H. Hufsky, J. Hockemeyer, M. Frieser, A. Lyssenko, D. H. Paper, J. Burgermeister, F. –D. Bohmer, H. –H. Fiebig, A. M. Burger, S. Baasner, T. Beckers, Synthetic 2-aroylindole derivatives as a new class of potent tubulin-inhibitory antimitotic agents, J. Med. Chem. 44 (2001) 4535–4553. [31] T. Beckers, T. Reissmann, M. Schmidt, A. M. Burger, H. –H. Fiebig, U. Vanhoefer, H. Pongratz, H. Hufsky, J. Hockemeyer, M. Frieser, S. Mahboobi, 2-Aroylindoles, a novel class of potent, orally active small molecule tubulin inhibitors, Cancer Res. 62 (2002) 3113–3119.
ACCEPTED MANUSCRIPT [32] A. Putey, F. Popowycz, Q. –T. Do, P. Bernard, S. K. Talapatra, F. Kozielski, C. M. Galmarini, B. Joseph, Indolobenzazepin-7-ones and 6-, 8-, and 9-Membered ring derivatives as tubulin polymerization inhibitors: synthesis and structure−activity relationship studies, J. Med. Chem. 52 (2009) 5916–5925.
RI PT
[33] B. Parrino, A. Carbone, G. Di Vita, C. Ciancimino, A. Attanzio, V. Spanò, A. Montalbano, P. Barraja, L. Tesoriere, M. A. Livrea, P. Diana, G. Cirrincione, 3-[4-(1H-Indol-3-yl)-1,3thiazol-2-yl]-1H-pyrrolo[2,3-b]pyridines,
nortopsentin
with
antiproliferative
SC
activity, Marine Drugs 13 (2015) 1901–1924.
analogues
[34] S. Inatsuki, T. Noguchi, H. Miyachi, S. Oda, T. Iguchi, M. Kizaki, Y. Hashimoto, H.
Chem. Lett. 15 (2005) 321–325.
M AN U
Kobayashia, Tubulin-polymerization inhibitors derived from thalidomide, Bioorg. Med.
[35] V. Spanò, A. Montalbano, A. Carbone, B. Parrino, P. Diana, G. Cirrincione, I. Castagliuolo, P. Brun, O. –G. Issinger, S. Tisi, I. Primac, D. Vedaldi, A. Salvador, P. Barraja, Synthesis of a
(2014) 340−357.
TE D
new class of pyrrolo[3,4-h]quinazolines with antimitotic activity, Eur. J. Med. Chem. 74
[36] D. Giomi, F. M. Cordero, F. Machetti in Comprehensive Heterocyclic Chemistry III-
EP
Isoxazoles, Vol. 4, (Eds.: A. R. Katritzky, C. A. Ramsden, E. F. V. Scriven, R. J. K. Taylor), Elsevier, Oxford, UK, 2008, pp. 365–485.
AC C
[37] H. Yoon, Y. Kwak, S. Choi, H. Cho, N. D. Kim, T. Sim, A pyrazolo[3,4-d]pyrimidin-4-amine derivative containing an isoxazole moiety is a selective and potent inhibitor of RET gatekeeper mutants, J. Med. Chem. 59 (2016) 358−373. [38] A. Carbone, M. Pennati, B. Parrino, A. Lopergolo, P. Barraja, A. Montalbano, V. Spanò, S. Sbarra, V. Doldi, M. De Cesare, G. Cirrincione, P. Diana, N. Zaffaroni, Novel 1Hpyrrolo[2,3-b]pyridine derivative nortopsentin analogues: synthesis and antitumor activity in peritoneal mesothelioma experimental models, J. Med. Chem. 56 (2013) 7060−7072.
ACCEPTED MANUSCRIPT [39] M. Caldarelli, I. Beria, N. Colombo, C. Piutti, M. Salsa, G. Traquandi, Isoxazolo-quinazolines as modulators of protein kinase activity, PCT Int. Appl. WO 2012013557; Chem. Abstr. 156 (2012) 203319. [40] R. Sharma, A. Jain, B. Sahu, D. Singh, S. Mali, Heterocyclyl compounds and uses thereof,
RI PT
PCT Int. Appl. WO 2014181287; Chem. Abstr. 161 (2014) 715759.
[41] L. –F. Yu, W. Tuckmantel, J. B. Eaton, B. Caldarone, A. Fedolak, T. Hanania, D. Brunner, R. J. Lukas, A. P. Kozikowski, Identification of novel α4β2-Nicotinic Acetylcholine Receptor
activity, J. Med. Chem. 55 (2012) 812−823.
SC
(nAChR) agonists based on an isoxazole ether scaffold that demonstrate antidepressant-like
M AN U
[42] M. V. Yermolina, J. Wang, M. Caffrey, L. L. Rong, D. J. Wardrop, Discovery, synthesis, and biological evaluation of a novel group of selective inhibitors of filoviral entry, J. Med. Chem. 54 (2011) 765−781.
[43] B. –L. Deng, T. L. Hartman, R. W. Buckheit, C. Pannecouque, E. De Clercq, M. Cushman,
TE D
Replacement of the metabolically labile methyl esters in the alkenyldiarylmethane series of non-nucleoside reverse transcriptase inhibitors with isoxazolone, isoxazole, oxazolone, or cyano substituents, J. Med. Chem. 49 (2006) 5316−5323.
EP
[44] C. M. Bastos, B. Munoz, B. Tait, Compounds, compositions, and methods for increasing cftr activity, PCT Int. Appl. WO 2015138934; Chem. Abstr. 163 (2015) 427383.
AC C
[45] N. C. Patel, J. Schwarz, X. J. Hou, D. J. Hoover, L. Xie, A. J. Fliri, R. J. Gallaschun, J. T. Lazzaro, D. K. Bryce, W. E. Hoffmann, A. N. Hanks, D. McGinnis, E. S. Marr, J. L. Gazard, M. Hajos, R. J. Scialis, R. S. Hurst, C. L. Shaffer, J. Pandit, C. J. O'Donnell, Discovery and characterization of a novel dihydroisoxazole class of α-amino-3-hydroxy-5-methyl-4isoxazolepropionic acid (AMPA) receptor potentiators, J. Med. Chem. 56 (2013) 9180−9191. [46] S. Velaparthi, M. Brunsteiner, R. Uddin, B. Wan, S. G. Franzblau, P. A. Petukhov, 5-tertButyl-N-pyrazol-4-yl-4,5,6,7-tetrahydrobenzo[d]isoxazole-3-carboxamide
derivatives
as
ACCEPTED MANUSCRIPT novel potent inhibitors of Mycobacterium tuberculosis pantothenate synthetase: initiating a quest for new antitubercular drugs, J. Med. Chem. 51 (2008) 1999−2002. [47] A. Lauria, C. Patella, P. Diana, P. Barraja, A. Montalbano, G. Cirrincione, G. Dattolo, A. M. Almerico,
A
new
tetracyclic
ring
system
of
biological
interest.
Indolo[3,2-
RI PT
e][1,2,3]triazolo[1,5-a]pyrimidines through domino reactions of 2-azidoindole, Heterocycles 60 (2003) 2669−2675.
[48] P. Barraja, L. Caracausi, P. Diana, A. Carbone, A. Montalbano, G. Cirrincione, P. Brun, G.
SC
Palù, I. Castagliuolo, F. Dall'Acqua, D. Vedaldi, Synthesis of pyrrolo[3,2-h]quinolinones with good photochemotherapeutic activity and no DNA damage, Bioorg. Med. Chem. 18 (2010)
M AN U
4830−4843.
[49] P. Barraja, L. Caracausi, P. Diana, A. Montalbano, A. Carbone, A. Salvador, P. Brun, I. Castagliuolo, S. Tisi, F. Dall'Acqua, D. Vedaldi, A. Salvador, G. Cirrincione, Pyrrolo[3,2h]quinazolines as photochemotherapeutic agents, ChemMedChem 6 (2011) 1238−1248.
TE D
[50] P. Barraja, P. Diana, V. Spanò, A. Montalbano, A. Carbone, B. Parrino, G. Cirrincione, An efficient synthesis of pyrrolo[3',2':4,5]thiopyrano[3,2-b]pyridin-2-one: a new ring system of pharmaceutical interest, Tetrahedron 68 (2012) 5087−5094.
EP
[51] A. Carbone, B. Parrino, P. Barraja, V. Spanò, G. Cirrincione, P. Diana, A. Maier, G. Kelter, H. –H. Fiebig, Synthesis and antiproliferative activity of 2,5-bis(3′-indolyl)pyrroles,
AC C
analogues of the marine alkaloid nortopsentin, Marine Drugs 11 (2013) 643−654. [52] P. Barraja, P. Diana, A. Montalbano, G. Dattolo, G. Cirrincione, G. Viola, D. Vedaldi, F. Dall'Acqua, Pyrrolo[2,3-h]quinolinones: a new ring system with potent photoantiproliferative activity, Bioorg. Med. Chem. 14 (2006) 8712−8728. [53] P. Diana, A. Stagno, P. Barraja, A. Carbone, B. Parrino, F. Dall'Acqua, D. Vedaldi, A. Salvador, P. Brun, I. Castagliuolo, O. G. Issinger, G. Cirrincione, Synthesis of triazenoazaindoles: a new class of triazenes with antitumor activity, ChemMedChem 6 (2011) 1291−1299.
ACCEPTED MANUSCRIPT [54] P. Barraja, V. Spanò, P. Diana, A. Carbone, G. Cirrincione, D. Vedaldi, A. Salvador, G. Viola, F. Dall’Acqua, Pyrano[2,3-e]isoindol-2-ones, new angelicin heteroanalogues, Bioorg. Med. Chem. Lett. 19 (2009) 1711–1714. [55] P. Barraja, P. Diana, A. Montalbano, A. Carbone, G. Viola, G. Basso, A. Salvador, D.
RI PT
Vedaldi, F. Dall’Acqua, G. Cirrincione, Pyrrolo[3,4-h]quinolinones a new class of photochemotherapeutic agents, Bioorg. Med. Chem. 19 (2011) 2326−2341.
[56] P. Barraja, V. Spanò, P. Diana, A. Carbone, G. Cirrincione, Synthesis of the new ring system
SC
6,8-dihydro-5H-pyrrolo[3,4-h]quinazoline, Tetrahedron Lett. 50 (2009) 5389−5391.
[57] V. Spanò, A. Montalbano, A. Carbone, B. Parrino, P. Diana, G. Cirrincione, P. Barraja,
M AN U
Convenient synthesis of pyrrolo[3,4-g]indazoles, Tetrahedron 69 (2013) 9839−9847. [58] P. Barraja, V. Spanò, D. Giallombardo, P. Diana, A. Montalbano, A. Carbone, B. Parrino, G. Cirrincione, Synthesis of [1,2]oxazolo[5,4-e]indazoles as antitumor agents, Tetrahedron 69 (2013) 6474–6477.
TE D
[59] P. Barraja, L. Caracausi, P. Diana, V. Spanò, A. Montalbano, A. Carbone, B. Parrino, G. Cirrincione, Synthesis and antiproliferative activity of the ring system [1,2]Oxazolo[4,5g]indole, ChemMedChem 7 (2012) 1901–1904.
EP
[60] A. M. Starosotnikov, D. V. Khakimo, M. A. Bastrakov, S. Y. Pechenkin, S. A. Shevelev, T. S. Pivina, Special features of 1,3-dipolar cycloaddition of n-methylazomethinylid to
AC C
nitrobenzazoles, Chem. Heterocycl. Compd. 47 (2011) 215−221. [61] A. M. Starosotnikov, M. A. Bastrakov, S. Y. Pechenkin, M. A. Leontieva, V. V. Kachala, S. A. Shevelev, 1,3-Dipolar cycloaddition of unstabilized N-methyl azomethine ylide to nitrobenzene annelated with azoles, J. Het. Chem. 48 (2011) 824−828. [62] N. Zaffaroni, A. Costa, M. Pennati, C. De Marco, E. Affini, M. Madeo, R. Erdas, A. Cabras, S. Kusamura, D. Baratti, M. Deraco, M. G. Daidone, Survivin is highly expressed and promotes cell survival in malignant peritoneal mesothelioma, Cell Oncol. 29 (2007) 453−466.
ACCEPTED MANUSCRIPT [63] M. De Cesare, D. Cominetti, V. Doldi, A. Lopergolo, M. Deraco, P. Gandellini, S. Friedlander, Y. Landesman, M. G. Kauffman, S. Shacham, M. Pennati, N. Zaffaroni, Antitumor activity of selective inhibitors of XPO1/CRM1-mediated nuclear export in diffuse malignant peritoneal mesothelioma: the role of survivin, Oncotarget 6 (2015) 13119−13132.
RI PT
[64] V. Spanò, M. Pennati, B. Parrino, A. Carbone, A. Montalbano, V. Cilibrasi, V. Zuco, A. Lopergolo, D. Cominetti, P. Diana, G. Cirrincione, P. Barraja, N. Zaffaroni, Preclinical activity of new [1,2]oxazolo[5,4-e]isoindole derivatives in diffuse malignant peritoneal
SC
mesothelioma, J Med Chem. 59 (2016) 7223−7238.
Perspect Biol. 4 (2012) a005942.
M AN U
[65] N. Rhind, P. Russell, Signaling pathways that regulate cell division, Cold Spring Harb
[66] M. Pennati, A. J. Campbell, M. Curto, M. Binda, Y. Cheng, L. Z. Wang, N. Curtin, B. T. Golding, R. J. Griffin, I. R. Hardcastle, A. Henderson, N. Zaffaroni, D. R. Newell, Potentiation of paclitaxel-induced apoptosis by the novel cyclin-dependent kinase inhibitor
1337.
TE D
NU6140: a possible role for survivin down-regulation, Mol. Cancer Ther. 4 (2005) 1328–
[67] M. V. Blagosklonny, P. Giannakakou, W. S. El-Deiry, D. G. I. Kingston, P. I. Higgs, L.
EP
Neckers, T Fojo, Raf-1/bcl-2 phosphorylation: a step from microtubule damage to cell death, Cancer Res 57 (1997) 130−135.
AC C
[68] V. Appierto, P. Tiberio, E. Cavadini, P. Casalini, G. Cappelletti, F. Formelli, Antimitotic effect of the retinoid 4-oxo-fenretinide through inhibition of tubulin polymerization: a novel mechanism of retinoid growth-inhibitory activity, Mol. Cancer Ther. 8 (2009) 3360–3368.
ACCEPTED MANUSCRIPT
Tables Table 1.
Sbt
R
R1
R2
Yieldsa (%)
6a
9a
Me
H
H
64
6b
9b
Bn
H
H
64
6c
9c
4-MeBn
H
H
72
6d
9d
4-OMeBn
H
H
60
6e
9e
3-OMeBn
H
H
78
6f
9f
2-OMeBn
H
H
75
6g
9g
2,3-(OMe)2Bn
H
H
65
6h
9h
2,5-(OMe)2Bn
H
H
75
6i
9i
3,4-(OMe)2Bn
H
H
65
6j
9j
3,5-(OMe)2Bn
H
H
74
6k
9k
3,4,5-(OMe)3Bn
H
H
61
6l
9l
Ph
H
H
58
9m
4-OMePh
H
H
62
9n
Me
COOEt Me
72
9o
Bn
COOEt Me
65
9p
4-OMeBn
COOEt Me
63
6q
9q
Me
H
Me
58
6r
9r
Bn
H
Me
56
6n 6o
a
AC C
EP
6p
M AN U
TE D
6m
SC
[1,2]Oxazoles
RI PT
[1,2]Oxazolo[5,4-e]isoindoles 6a-r.
Figures represent the yield obtained at the final reaction step.
ACCEPTED MANUSCRIPT
Table 2.
IC50 (µM)[a] cpd
JR8
M14
MCF7
MDA-MB-231
PC3
RI PT
Cytotoxic activity of [1,2]Oxazolo[5,4-e]isoindoles 6a-r in human cell lines.
DU145
STO
MP8
6a
58.37±2.36 65.85±3.65 86.64±1.95
55.37±3.52
77.14±4.99 69.32±3.51 28.13±4.08 26.69±1.72
6b
5.14±1.19
6.88±1.17
11.26±3.21
19.64±2.44
13.47±3.54 15.19±2.32
6c
>100
>100
>100
>100
6d
9.15±1.81
12.45±3.20
7.89±1.56
15.56±2.25
18.63±3.09 15.21±3.65
6e
0.96±0.12
1.02±0.17
1.17±0.22
0.85±0.07
0.96±0.11
6f
16.21±3.48 20.47±2.89 19.37±3.65
14.38±2.56
>100
>100
>100
>100
n.a.
n.a.
0.69±0.22
>100
>100
>100
>100
n.a.
n.a.
>100
>100
>100
>100
1.37±0.24
0.46±0.06
0.51±0.07
>100
>100
18.57±4.08 17.54±2.99
0.79±0.14
1.52±0.34
>100
>100
>100
>100
n.a.
n.a.
>100
>100
>100
>100
>100
SC
0.79±0.13
>100
M AN U
6g
WI38 MCF10A
>100
>100
19.16±4.44 16.83±3.85 15.15±2.82
19.63±1.89
23.65±3.93 19.47±2.87
6i
0.85±0.11
0.96±0.09
1.03±0.04
0.88±0.02
0.79±0.12
0.87±0.04
0.64±0.16
0.51±0.11
>100
>100
6j
0.23±0.04
0.31±0.07
0.33±0.09
0.32±0.02
0.27±0.10
0.31±0.06
0.06±0.01
0.07±0.02
>100
>100
6k
0.63±0.05
0.89±0.08
0.92±0.03
0.75±0.04
0.87±0.03
0.99±0.01
0.62±0.04
0.73±0.03
>100
>100
6l
59.55±3.97 63.38±4.12 81.15±5.32
6m
61.55±4.90 58.46±2.62 79.64±2.56
EP
TE D
6h
70.45±2.08 76.44±3.33 29.47±3.11 25.33±1.96
n.a.
n.a.
57.49±3.29
79.87±4.17 81.13±2.77 25.46±3.22 21.19±2.13
n.a.
n.a.
AC C
68.44±3.64
6n
>100
>100
>100
>100
>100
>100
>100
>100
n.a.
n.a.
6o
>100
>100
>100
>100
>100
>100
>100
>100
n.a.
n.a.
6p
>100
>100
>100
>100
>100
>100
>100
>100
n.a.
n.a.
6q
>100
>100
>100
>100
>100
>100
>100
>100
n.a.
n.a.
6r
>100
>100
>100
>100
>100
>100
>100
>100
n.a.
n.a.
ACCEPTED MANUSCRIPT
[a]
Data are reported as IC50 values (concentration of drug required to inhibit growth by 50%) determined by MTS assay after 72 h of continuous
exposure of both human tumor (JR8, M14, MCF7, MDA-MB-231, PC3, DU145, STO and MP8) and normal (WI38 and MCF10A) cell lines to each
AC C
EP
TE D
M AN U
SC
RI PT
compound. Data represent mean values±SD of three independent experiments. n.a.: not assessed.
ACCEPTED MANUSCRIPT Figure Legends Figure 1. Structures of pyrano[2,3-e]isoindoles (1), pyrrolo[3,4-h]quinolines (2), pyrrolo[3,4h]quinazolines
(3),
[1,2]oxazolo[5,4-e]indazoles
(4),
[1,2]oxazolo[4,5-g]indoles
(5),
RI PT
[1,2]oxazolo[5,4-e]isoindoles (6). Figure 2. Effect of 6j derivative on DMPM growth. (A) Cytotoxic activity of 6j in DMPM cell lines. STO (●) and MesoII (■) cells were cultured for 72 h in the presence of increasing concentrations of the compound and the cytotoxic activity was assessed by MTS assay. Data are
SC
expressed as percentage values with respect to untreated cells (only DMSO) and represent the mean
M AN U
values±SD (standard deviation) of three independent experiments. (B) Antitumor activity of 6j on STO cells xenotransplanted on athymic nude mice. Drugs were administered ip at 5 mg/kg, q7dx4, starting 10 days after cell inoculum. Data are expressed as mean values±SE. Figure 3. Effect of 6j derivative on tubulin polymerization. Representative western blot showing
TE D
the soluble (S) or polymerized (P) tubulin fraction in STO and MP8 cells after 24 h of exposure to 6j at the concentrations corresponding to the IC50 at 72 h. Vinorelbine (Vin: 2 and 13 nM for STO and MP8, respectively) and taxol (Tx: 1 and 18 nM for STO and MP8, respectively) were selected
EP
as reference drugs due to their opposite mechanism of action on tubulin polymerization.
AC C
Figure 4. Effect of 6j derivative on cell cycle progression. (A) Flow cytometric analysis of the cell cycle distribution in STO and MP8 cells exposed for 48 and 72 h to 1% (v/v) DMSO (control cells; Ctr) and 6j (IC50 and IC80) or taxol (Tx) and vinorelbine (Vin) (IC50). Data are reported as the percentage of cells in G1, S , and G2/M phases and represent the mean values of three independent experiments; SDs were always within 5%. (B) Percentage of mitotic cells upon the exposure of STO and MP8 cells to equitoxic concentrations (IC50) of 6j, vinorelbine (Vin; 2 and 13 nM for STO and MP8, respectively) and taxol (Tx; 1 and 18 nM for STO and MP8, respectively). Data are expressed as mean values±SD of three independent experiments.
ACCEPTED MANUSCRIPT Figure 5. Effect of 6j derivative on apoptosis induction. The catalytic activity of caspase-3 was assessed by the in vitro hydrolysis of the fluorogenic substrates (DEVD-pNA) after exposure of STO and MP8 cells to 1% (v/v) DMSO (control cells; Ctr) or to 6j derivative. Data are expressed as
AC C
EP
TE D
M AN U
SC
RI PT
mean values±SD of three independent experiments. ***P<0.001, **P<0.01, *P< 0.05.
Figure 1
ACCEPTED MANUSCRIPT
O
O N
RI PT
N
NR1
O
NR
NR
1
SC
NR
N O
2
R N
N
8
3
TE D 5
EP
R
1
R2 7
R2
AC C
N
3
N O
N O
4
M AN U
2
NR 4
5
6
6
R1
Figure 2
ACCEPTED MANUSCRIPT
600
75
6j
25
0.06
0.08
EP
Concentrations (M)
0.10
TE D
0.04
AC C
0.02
400
M AN U
50
0 0.00
Ctr
SC
Tumor Volume (mm3)
STO MP8
100
RI PT
B
% DMSO-treated cells
A
200
0 0
10
20
30
40
50
Days after cells injection
60
Figure 3
Ctr
6j
Tx
Vin
STO
RI PT
ACCEPTED MANUSCRIPT
MP8 P
S
P
S
EP
P
M AN U
S
TE D
P
AC C
S
SC
α-Tubulin
Figure 4
ACCEPTED MANUSCRIPT
A
B STO
STO
RI PT
Vn
75 50 G1
25
M AN U
% Cells
100
S
G2/M
48
72
48
72
48
IC50
72
IC80
48
72
IC50
6j
Tx
50 25 0
G1 S G2/M
48
72
48
72
IC50
48
72
IC80
48
75 50 25
0
Ctr
6j
Tx
Vin
Tx
Vin
MP8
EP AC C
% Cells
75
100
Vn
100
hours
72
IC50
MP8 Ctr
48
TE D
0
hours
% MPM-2-positive cells
Tx
72
IC50
48
72
IC50
% MPM-2-positive cells
6j
SC
Ctr
100 75 50 25 0
Ctr
6j
Figure 5
ACCEPTED MANUSCRIPT
6j
50
**
**
25
M AN U
**
*
0
hours
24
48
72
24
48
***
SC
Caspase-3 activity
75
RI PT
STO Ctr
72
* 24
TE D
IC50
48
72
IC80
MP8
50
***
0
24
48
**
**
25
hours
6j
EP
75
AC C
Caspase-3 activity
Ctr
* 72
24
* 48
IC50
* 72
24
48
IC80
72
ACCEPTED MANUSCRIPT HIGHLIGHTS • A new series of [1,2]oxazolo[5,4-e]isoindoles were prepared by a versatile sequence • Remarkable and selective antiproliferative activity against DMPM
RI PT
• 6j impairs tubulin polymerization in a vinca alkaloid-like manner • 6j causes cell cycle arrest at G2/M phase and apoptosis in DMPM cells
AC C
EP
TE D
M AN U
SC
• 6j determined tumor volume inhibition (51%) without any appreciable sign of toxicity