Abstract
New N,N’-bis(5-arylidene-4-oxo-4,5-dihydrothiazoline-2-yl)diamine derivatives 5 were prepared in two steps from rhodanine and piperazine, or 1,4-bis(3-amino-propyl)piperazine, under microwave reaction conditions with retention of configuration. Some of these compounds were tested for in vitro antiproliferative activities and for their kinase inhibitory potencies towards six kinases (CDK5/p25, GSK3α/β, DYRK1A, DYRK2, CLK1, and CLK2). The compound 5d showed nanomolar activity towards DYRK1A kinase (IC50 = 0.041 μM).
Keywords: 5-Arylidene rhodanine; 2,2’-(Piperazine-1,4-diyl)bis(5-benzylidene-1,3-thiazol-4(5H)-one); Microwave irradiation; Cytotoxity; Protein kinase; Diamines
Introduction
The 2-amino-5-arylidene-5H-thiazol-4-ones and their 5-arylidene rhodanine precursors are a class of five-membered heterocyclic rings (FMHRs) considered as “privileged scaffolds” in the medicinal chemistry community [1]. Considerable work has been published over decades about their chemistry and biology. A number of compounds containing the 2-amino-5-arylidenethiazol-4(5H)-one moiety have been shown to exhibit anti-inflammatory [2], antimicrobial [3], and antitumor [4] effects. Among these compounds, Darbufelone® A[5] (Figure 1) is orally active in animal models of inflammation [6] and DBPT B is under clinical trials for colon cancer [7]. 5-Arylidene rhodanines have also proven to be attractive for the discovery of new candidates. A series of rhodanine-based hits C (Figure 1) were found as potent and selective inhibitors of the “atypical” dual-specificity phosphatase (DSP) family member-JNK-stimulating phophatase-1 (JSP-1). Compounds of this class may be useful for the treatment of inflammatory and proliferative disorders [8]. As the last example, epalrestat D reduced the symptoms of diabetic neuropathy [9].
Fig. 1.
Structures of Darbufelone (A), DBPT (B), inhibitor of DSP (C), and epalrestat (D).
Protein kinases are the enzymes which control the phosphorylation of protein in cellular life [10] which is frequently deregulated in human diseases. For the pharmaceutical industry, the protein kinases represent interesting targets for new therapeutic agents [11] and this interest was boosted by the approval of the first marketed inhibitor Gleevec™ used in myeloid leukemia [12]. Due to the biological activity associated with the 2-amino-thiazolidinone moiety, we decided in this paper to explore the synthesis of N,N’-bis(5-arylidene-4-oxo-4,5-dihydrothiazolidin-2-yl)diamines derived from piperazine or 1,4-bis(3-aminopropyl)piperazine as linkers, and to study their effects on cells and protein kinases.
Results and Discussion
Chemistry
The strategy used for the synthesis of the symmetric derivatives 5 is outlined in Scheme 1. The reactions have been realized under microwave irradiation [13]. The main benefits of microwave irradiation technology are the significant rate-enhancements and sometimes elevated product yields enabling the rapid synthesis of molecules of potential value in medicinal chemistry [14]. The synthesis started by the solution-phase Knoevenagel condensation of aryl aldehydes 2 and commercial rhodanine 1a. The expected compounds 3a–c were prepared in yields ranging from 65 to 88% with a reaction time of 10 min. under microwave irradiation (MWI) at 65°C in the presence of sodium acetate. In a similar approach, the 5-arylidene rhodanine propanoic derivatives 3d–g were easily synthesized using solvent-less reaction conditions under microwave irradiation (130°C, 10 min.). The geometric double bond of 3 was attributed as being Z by the shielding effect of the carbonyl group C-4 on the olefinic proton H-5 (δ 7.5 ppm) in the 1H NMR spectra.
Sch. 1.
a) for 3a–c: MeOH, AcONa 3 eq., MWI, 65°C, 10 min. and for 3d–g: MWI, 130°C, 10 min. b) MWI, 80–120°C, 30 min.
Transformation of 5-arylidene rhodanine 3 into 2-amino-5-arylidene-5H-thiazol-4-one after addition of a primary amine [15] usually involves activation of the C=S bond of rhodanine via the thioether intermediate that subsequently undergoes a thioalkyl/nitrogen displacement. In order to be able to carry out such sulfur/nitrogen displacement in a faster and more efficient way – avoiding the preparation of the thioether intermediate – we examined the influence of microwave irradiation on the reaction between compound 3 and the symmetric diamino linkers 4a,b. The experiments revealed that optimal reaction conditions were obtained at 80–120°C after 30 minutes. It is noteworthy that sulfur/nitrogen displacement reactions at 120°C under microwave irradiation have been realized in solution with hexane to avoid decomposition of compound 5. The desired compounds 5a–e were prepared in poor to moderate yields (13–26%) and their structures were substantiated by 1H, 13C NMR, and HRMS analyses and only the more thermo-dynamically stable Z-isomers were obtained.
Biology
To evaluate the potency of the compounds 3a–c, 5a, and 5c for their in vitro antiproliferative activities, we used six representative tumor cell lines of liver (Huh7), colon (Caco2, HCT 116), breast (MDA-MB 231), prostate (PC3), lung (NCI), and one normal cell line (fibroblats) and measured survival. Results are reported in Table 1. The 5-arylidene rhodanines 3a–c and two compounds 5a and 5c showed measurable, albeit very poor, cytotoxic activity. No clear tendency for higher activity of these compounds can be deduced from these results. Since the log P values of all tested compounds are in the same range, influences of lipophilicity on cytotoxic activity cannot be deduced as well.
Tab. 1.
Cell effects of the products and calculated partition coefficients log P (calculated with Chem Draw Pro, Cambridge Soft)
| Cpd. |
Cell lines IC50 (μM)
|
|||||||
|---|---|---|---|---|---|---|---|---|
| Huh7 | Caco2 | MDA-MB 231 | HCT 116 | PC3 | NCI | Fibroblats | Log Pcalc. | |
| 3a | > 25 | > 25 | > 25 | > 25 | > 25 | > 25 | > 25 | 2.0 |
| 3b | > 25 | > 25 | > 25 | > 25 | > 25 | > 25 | > 25 | 1.9 |
| 3c | > 25 | > 25 | > 25 | > 25 | > 25 | > 25 | > 25 | 1.8 |
| 5a | > 25 | 25 | > 25 | > 25 | > 25 | > 25 | > 25 | 4.8 |
| 5c | > 25 | 20 | > 25 | > 25 | 40 | > 25 | 40 | 5.3 |
| Roscovitine | 10 | 10 | 15 | 8 | 8 | 20 | > 25 | – |
| DMSO | > 25 | > 25 | > 25 | > 25 | > 25 | > 25 | > 25 | – |
The kinase inhibitory potencies of compounds 3a–c and 5d were evaluated as IC50 values towards six protein kinases (CDK5/p25, GSK3α/β, DYRK1A, DYRK2, CLK1 and CLK2) and the results are reported in Table 2. The 5-arylidene rhodanines 3a,b appeared to be inactive towards these six protein kinases, but interesting results were obtained with compounds 5d and 3c. The 1,4-bis[(5Z)-5-(4-methoxybenzylidene)methylene-4-oxo-4,5-dihydrothiazol-2-yl]piperazine 5b and the 5-arylidene rhodanine 3c are very active on DYRK1A [16] and a noteworthy IC50 = 41 nM was measured for 5b. The compound 5d has also shown submicromolar inhibition potencies towards DYRK2 (IC50 = 0.6 μM) and CLK1 (IC50 = 0.5 μM). Regarding these results, the potential of the (5Z) 5-arylidene-4-oxo-4,5-dihydro-thiazolidine-2-yl moiety appended on 1,4-bis(3-aminopropyl)piperazine 4b as a linker could be highly interesting in the development of a new class of inhibitors of DYRK1A kinase which is known to be involved in Alzheimer’s disease/Down syndrome [17].
Tab. 2.
CDK5/p25, GSK3α/β, DYRK1A, DYRK2, CLK1, CLK3 inhibitions.
| Cpd. |
IC50 (μM)
|
|||||
|---|---|---|---|---|---|---|
| CDK5/p25 | GSK3α/β | DYRK1A | DYRK2 | CLK1 | CLK2 | |
| 3a | > 10 | > 10 | > 10 | > 10 | > 10 | > 10 |
| 3b | > 10 | > 10 | > 10 | > 10 | > 10 | > 10 |
| 3c | > 10 | 8.5 | 0.070 | – | – | – |
| 5d | > 10 | > 10 | 0.041 | 0.6 | 0.5 | 7.7 |
Conclusion
In conclusion, we worked out a short and practical synthesis under microwave irradiation of N,N’-(5-arylidene-4-oxo-4,5-dihydrothiazolidin-2-yl)diamines 5a–e derived from piperazine 4a[18, 19] and 1,4-bis(3-aminopropyl)piperazine 4b. The in vitro anti-proliferative activities are extremely weak and this could be due to a lower cellular penetration of compounds 3 and 5 or a lower interaction with the cellular targets. Surprisingly, the compound 5d has shown nanomolar inhibition potency towards DYRK1A and this interesting inhibition led us to expand our efforts in the synthesis of diversely disubstituted N,N’-bispiperazine derivatives with the 5-arylidene thiazolidinone moiety as new potential inhibitors of this kinase. Work is in progress to gain deeper insight into the structure-activity relationships (SAR) of this new interesting class of diamines.
Experimental
General
Elemental analysis: Flash EA1112 CHN/O Thermo Electron; HRMS (MS/MS ZABSpec Tof Micromass, EBE TOF geometry, IP 8 eV); NMR: BRUKER AC 300P (1H: 300 MHz, 13C: 75 MHz); melting points: Leica System Kofler VMHB Melting Point apparatus (not corrected); microwave reactor: Monowave 300 Anton-Paar (850 W), monowave software package.
General procedure I: Preparation of 5-arylidene-2-thioxo-thiazolidine-4-one 3a–c under microwave irradiation
A mixture of rhodanine 1a (1 g, 7.5 mmol), aldehyde 2 (8.95–9 mmol), sodium acetate (1.85 g, 22.55 mmol), and methanol (5 ml) was placed in a borosilicate glass vial (10 ml) with a Teflon® magnetic stir bar and sealed with a snap cap. The glass tube was then introduced into the Monowave 300 Anton-Paar microwave cavity (P = 850 Watt) and the stirred mixture was irradiated at 65°C (with a power of 10 Watt) for 30 min. After microwave dielectric heating, the crude reaction mixture was allowed to cool down to room temperature, and then was filtered on a Buchner funnel and the insoluble compound 3 was washed with methanol (2x5 ml). The crude compound 3 was purified by recrystallization from methanol and further dried under high vacuum (10−2 Torr) for 1 hour, which gave the desired compound 3 as a powder.
General procedure II: Preparation of 3-(5-arylidene-4-oxo-2-thioxo-thiazolidine-3-yl)propanoic acid 3d–g under solvent-free microwave irradiation
A mixture of rhodanine 1b (0.5 g, 2.44 mmol) and aldehyde 2 (2.44 mmol) was placed in a borosilicate glass vial (10 ml) with a Teflon® magnetic stir bar and sealed with a snap cap. The glass tube was then introduced into the Monowave 300 Anton-Paar microwave cavity (P = 850 Watt) and the stirred mixture was irradiated at 130°C for 10 min. After microwave dielectric heating, the crude reaction mixture was allowed to cool down to room temperature and 5 ml of a mixture of ethanol/hexane (1:1) were added directly to the glass vial. The resulting precipitated product 3 was filtered on a Buchner funnel and the insoluble compound 3 was washed with the same mixture (2x5 ml). The crude compound 3 was further dried under high vacuum (10−2 Torr) for 1 hour, which gave the desired compound 3 as a powder.
General procedure III: Synthesis of N,N’-bis(5-arylidene-4-oxo-4,5-dihydro-thiazolidine-2-yl)piperazine derivatives 5a–e under microwave irradiation
A mixture of 5-arylidene-2-thioxo-thiazolidine-4-one 3 (4 mmol), piperazine 4a (172 mg, 2 mmol) or 1,4-bis(3-aminopropyl)piperazine 4b (400 mg, 2 mmol) was placed in a borosilicate glass vial (10 ml) with a Teflon® magnetic stir bar and sealed with a snap cap. The glass tube was then introduced into the Monowave 300 Anton-Paar microwave cavity (P = 850 Watt) and the stirred mixture was irradiated at 80–120°C (with a power of 5–200 Watt) for 30 min. After microwave dielectric heating, the crude reaction mixture was allowed to cool down to room temperature and 10 ml of cooled ethanol (4°C) were added directly in the glass vial. The resulting precipitated product 5 was filtered off, washed with 2 × 5 ml of ethanol and dried under high vacuum (10−2 Torr) at room temperature for 1 hour. After 1H NMR analysis, the product 5 was purified by recrystallization from methanol, which gave the desired compound 3 as a powder.
(5Z)-5-Benzylidene-2-thioxo-1,3-thiazolidin-4-one (3a)
Prepared following General procedure I from benzaldehyde 2a (0.95 g, 8.95 mmol) to give 1.08 g (65%) as an orange powder. Mp = 208–210°C. 1H NMR (300 MHz, DMSO-d6) δ (ppm) = 7.44–7.55 (m, 5H, Ar), 7.57 (s, 1H, =CH), 13.12 (br s, 1H, NH). 13C NMR (75 MHz, DMSO-d6) δ (ppm) = 126.88 (C-1), 129.20 (C-2), 130.30 (C-3), 130.37 (C-4), 130.45 (C=, C-5), 133.22 (=CH), 171.28 (C=S, C-2); 196,64 (C=O, C-4). HRMS, m/z = 243.9868 found (calculated for C10H7NONaS2, [M+Na]+ requires 243.9867). Anal. Calcd. for C10H7NOS2: C, 54.27; H, 3.19. Found: C, 54.23; H, 3.16.
(5Z)-5-(4-Methoxybenzylidene)-2-thioxo-1,3-thiazolidin-4-one (3b)
Prepared following General procedure I from 4-methoxybenzaldehyde 2b (1.23 g, 9 mmol) to give 1.36 g (74%) as an orange powder. Mp = 250–252°C. 1H NMR (300 MHz, DMSO-d6) δ (ppm) = 3.82 (s, 3H, OCH3), 7.10 (d, J = 8.8Hz, 2H, H-3, Ar), 7.55 (d, J = 9.1 Hz, 2H, H-2, Ar), 7.57 (s, 1H; =CH), 13.76 (br s, 1H, NH). 13C NMR (75 MHz, DMSO-d6) δ (ppm) = 55.45 (OCH3), 114.92 (C-3’), 124.90 (C=, C-5), 126.02 (C-1’), 129.71 (=CH), 132.28 (C-2’), 160.82 (C-4’), 173.024 (C=S, C-2), 197.25 (C=O, C-4). HRMS, m/z = 295.9792 found (calculated for C11H9NO2Na2S2, [M-H+2Na]+ requires 295.9792). Anal. Calcd. for C11H9NO2S2: C, 52.57; H, 3.61. Found: C, 52.65; H, 3.67.
(5Z)-5-(1,3-Benzodioxol-5-ylmethylidene)-2-thioxo-1,3-thiazolidin-4-one (3c)
Prepared following General procedure I from piperonaldehyde 2c (1.35 g, 9 mmol) to give 1.74 g (88%) as an orange powder. Mp > 260°C. 1H NMR (300 MHz, DMSO-d6) δ (ppm) = 6.13 (s, 2H; OCH2O), 7.11–7.45 (m, 3H; H-5’, H-6’, Ar), 7.52 (s, 1H, =CH), 7.55 (s, 1H, H-2’; Ar), 13.74 (br s, 1H, NH). 13C NMR (75 MHz, DMSO-d6) δ (ppm) = 102.10 (OCH2O), 109.26 (C=, C-5), 109.45 (C-6’), 123.02 (C-1’), 126.65 (C-5’), 127.16 (=CH), 131.81 (C-2’), 148.27 (C-4’), 149.60 (C-3’), 169.58 (C=S, C-2), 195.51 (C=O, C-4). HRMS, m/z = 287.9765 found (calculated for C11H7NO3NaS2, [M+Na]+ requires 287.9765). Anal. Calcd. for C11H7NO3S2: C, 49.80; H, 2.66. Found: C, 49.91; H, 2.69.
3-[(5Z)-5-Benzylidene-4-oxo-2-thioxo-1,3-thiazolidin-3-yl]propanoic acid (3d)
Prepared following General procedure II from benzaldehyde 2a (0.256 g, 2.44 mmol) to give 3d in 60% yield as a brown powder. Mp = 224–226°C. 1H NMR (300 MHz, DMSO-d6) δ (ppm) = 2.33 (m, 2H, CH2), 4.15 (m, 2H, CH2), 7.51–7.65 (m, 5H, H-2, H-3, H-4, Ar), 7.80 (s, 1H, =CH), 13.16 (br s, 1H, OH). 13C NMR (75 MHz, DMSO-d6) δ (ppm) = 33.30 (CH2), 41.78 (CH2), 122.6 (C=, C-5), 129.46 (C-2’), 130.57 (C-3’), 130.84 (C-4’), 132.63 (=CH), 133.02 (C-1), 166.76 (C=O, C-4), 171.94 (CO2H), 193.15 (C=S, C-2). HRMS, m/z = 337.9904 found (calculated for C13H10NO3Na2S2, [M-H+2Na]+ requires 337.9898).
3-[(5Z)-5-(4-Methoxybenzylidene)-4-oxo-2-thioxo-1,3-thiazolidin-3-yl]propanoic acid (3e)
Prepared following General procedure II from 4-methoxybenzaldehyde 2b (0.332 g, 2.44 mmol) to give 3e in 60% as a brown powder. Mp > 260°C. 1H NMR (300 MHz, DMSO-d6) δ (ppm) = 2.24 (t, 2H, J = 8.2 Hz, CH2), 3.83 (s, 3H, OCH3), 4.12 (t, 2H, J = 8.2 Hz, CH2), 7.10 (d, 2H, J = 8. 8 Hz, H-3, Ar), 7.64 (d, 2H, J = 8.8 Hz, H-2, Ar), 7.86 (s, 1H, =CH), 13.26 (br s, 1H, OH). 13C NMR (75 MHz, DMSO-d6) δ (ppm) = 34.11 (CH2), 42.20 (CH2), 55.56 (OCH3), 115.11 (C-3’), 119.26 (C=, C-5), 125.58 (C-1’); 129.90 (=CH), 132.84 (C-2’), 161.41 (C-4’), 166.86 (C=O, C-4), 171.98 (CO2H), 192.86 (C=S, C-2). HRMS, m/z = 346.0185 found (calculated for C14H13NO4NaS2, [M+Na]+ requires 346.0184).
3-[(5Z)-5-(1,3-Benzodioxol-5-ylmethylidene)-4-oxo-2-thioxo-1,3-thiazolidin-3-yl]propanoic acid (3f)
Prepared following General procedure II from piperonal 2c (0.366 g, 2.44 mmol) to give 3f in 63% as a brown powder. Mp = 220–224°C. 1H NMR (300 MHz, DMSO-d6) δ (ppm) = 2.61 (t, 2H, J = 3.9 Hz, CH2), 4.21 (t, 2H, J = 4.7 Hz, CH2), 6.15 (s, 2H, OCH2O), 7.09–7.23 (m, 3H, H-2, H-5, H-6, Ar), 7.74 (s, 1H, =CH), 12.50 (br s, 1H, OH). 13C NMR (75 MHz, DMSO-d6) δ (ppm) = 30.74 (CH2), 40.30 (CH2), 102.21 (OCH2O), 109.35 (C-2’), 109.61 (C-5’), 119.71 (C=, C-5), 126.98 (C-6’), 127.14 (C-1’), 133.21 (=CH), 148.35 (C-4’), 149.88 (C-3’), 166.67 (C=O, C-4), 171.72 (CO2H), 192.97 (C=S, C-2). HRMS, m/z = 346.0185 found (calculated for C14H13NO4NaS2, [M+Na]+ requires 346.0184).
3-[(5Z)-5-(2,3-Dihydro-1,4-benzodioxin-6-ylmethylidene)-4-oxo-2-thioxo-1,3-thiazolidin-3-yl]propanoic acid (3g)
Prepared following General procedure II from 1,4-benzodioxane-6-carboxaldehyde (2d) (0.401 g, 2.44 mmol) to give 3g in 54% yield as a brown powder. Mp = 226–228°C. 1H NMR (300 MHz, DMSO-d6) δ (ppm) = 2.55 (t, 2H, J = 7.3 Hz, CH2), 4.18 (t, 2H, J = 7.4 Hz, CH2), 4.30 (m, 4H, OCH2CH2O), 7.02 (d, 1H, J = 8.6 Hz, H-5, Ar), 7.12 (d, 1H, J = 2.2 Hz, H-6, Ar), 7.15 (s, 1H, H-2, Ar), 7.70 (s, 1H; =CH), 12.46 (br s, 1H, OH). 13C NMR (75 MHz, DMSO-d6) δ (ppm) = 31.30 (CH2), 40.30 (CH2), 64.01 (OCH2CH2O), 118.22 (C-2’), 119.47 (C-5’), 119.79 (C=, C-5), 124.46 (C-6’), 126.31 (C-1’), 132.96 (=CH), 143.82 (C-4’), 146.25 (C-3’); 166.67 (C=O, C-4), 171.80 (CO2H), 192.93 (C=S, C-2). HRMS, m/z = 374.0138 found (calculated for C15H13NO5NaS2, [M+Na]+ requires 374.0133).
(5Z,5’Z)-2,2’-(Piperazine-1,4-diyl)bis(5-benzylidene-1,3-thiazol-4(5H)-one) (1,4-Bis[(5Z)-5-benzylidene-4-oxo-4,5-dihydro-1,3-thiazol-2-yl]piperazine, 5a)
Prepared following General procedure III from 5-benzylidene-2-thioxo-thiazolidin-4-one (3a) (885 mg, 4 mmol) and piperazine 4a (172 mg, 2 mmol) in hexane (2 ml) at 120°C to give 5a in 13% yield as a brown powder. Mp > 260°C. 1H NMR (300 MHz, DMSO-d6) δ (ppm) = 3.34 (br s, 8H, N(CH2)4N), 7.28–7.7 (m, 12H, H-2’, H-3’, H-4’, Ar, =CH). 13C NMR (75 MHz, DMSO-d6) δ (ppm) = 42.45 (N(CH2)4N), 129.08 (C-2’), 129.16 (=CH), 129.22 (C=), 129.55 (C-3’), 129.82 (C-4’), 129.94 (C-1’); 130.41 (C=N), 159.56 (C=O). HRMS, m/z = 483.0918 found (calculated for C24H20N4O2NaS2, [M+Na]+ requires 483.0925). Anal. Calcd. for C24H20N4O2S2: C, 62.59; H, 4.38. Found: C, 62.56; H, 4.41.
(5Z,5’Z)-2,2’-(Piperazine-1,4-diyl)bis[5-(4-methoxybenzylidene)-1,3-thiazol-4(5H)-one] (1,4-Bis[(5Z)-5-(4-methoxybenzylidene)-4-oxo-4,5-dihydro-1,3-thiazol-2-yl]piperazine, 5b)
Prepared following General procedure III from 5-(4-methoxybenzylidene)-2-thioxothiazolidin-4-one (3b) (1.005 g, 4 mmol) and piperazine 4a (172 mg, 2 mmol) at 80°C to give 5b in 36% yield as a yellow powder. Mp > 260°C. 1H NMR (300 MHz, DMSO-d6) δ (ppm) = 2.99 (s, 8H, N(CH2)4N), 3.79 (s, 6H, OCH3), 7.04 (d, J = 5 Hz, 4H, H-3’, Ar), 7.16 (s, 2H, =CH), 7.43 (d, J = 8.7 Hz, 4H, H-2’, Ar). 13C NMR (75 MHz, DMSO-d6) δ (ppm) = 42.99 (N(CH2)4N), 55.27 (OCH3), 114.54 (C-3’), 114.73 (C-1’), 124.13 (=CH), 127.55 (C=, C-5), 131.29 (C-2’), 131.38 (C-4’), 132.14 (C=N, C-2), 159.56 (C=O, C-4). HRMS, m/z = 521.1317 found (calculated for C26H25N4O4S2, [M+H]+ requires 521.1310). Anal. Calcd. for C26H24N4O4S2: C, 59.98; H, 4.65. Found: C, 59.87; H, 4.64.
(5Z,5’Z)-2,2’-(Piperazine-1,4-diyl)bis[5-(1,3-benzodioxol-5-ylmethylene)-1,3-thiazol-4(5H)-one]: (1,4-Bis[(5Z)-5-(1,3-benzodioxol-5-ylmethylene)-4-oxo-4,5-dihydro-1,3-thiazol-2-yl]piperazine, 5c)
Prepared following General procedure III from 5-(1,3-benzodioxol-5-ylmethylidene)-2-thioxothiazolidin-4-one (3c) (1.061 g, 4 mmol) and piperazine 4a (172 mg, 2 mmol) in hexane (2 ml) at 120°C to give 5a in 15% yield as a brown powder. Mp > 260°C. 1H NMR (300 MHz, DMSO-d6) δ (ppm) = 3.02 (s, 8H, N(CH2)4N), 6.11 (s, 4H, OCH2O), 7.02–7.18 (m, 6H, H-2’, H-5’, H-6’, Ar), 7.58 (s, 2H, =CH). 13C NMR (75 MHz, DMSO-d6) δ (ppm) = 52.72 (N(CH2)4N), 101.88 (OCH2O), 108.71 (C-2’), 125.13 (C-5’), 126.04 (C=, C-5), 127.86 (C-1’), 130.41 (C-6’), 144.21 (=CH), 146.02 (C=N, C-2), 148.11 (C-4’), 148.78 (C-3’), 182.22 (C=O, C-4). HRMS, m/z = 571.0726 found (calculated for C26H20N4O6NaS2, [M+Na]+ requires 571.0722). Anal. Calcd. for C26H20N4O6S2: C, 56.92; H, 3.67. Found: C, 56.96; H, 3.71.
(5Z,5’Z)-2,2’-[Piperazine-1,4-diylbis(propane-3,1-diylimino)]bis[5-(4-methoxybenzylidene)-1,3-thiazol-4(5H)-one]: (1,4-Bis(3-{[(5Z)-5-(4-methoxybenzylidene)-4-oxo-4,5-dihydro-1,3-thiazol-2-yl]amino}propyl)piperazine, 5d)
Prepared following General procedure III from 5-(4-methoxybenzylidene)-2-thioxothiazolidin-4-one (3b) (1.005 g, 4 mmol) and 1,4-bis(3-aminopropyl)piperazine (4b) (0.4 g, 2 mmol) in hexane (2 ml) at 120°C to give 5d in 25% yield as a yellow powder. Mp = 220–222°C. 1H NMR (300 MHz, DMSO-d6) δ (ppm) = 1.68 (quint, J = 6.9 Hz, 4H, NCH2CH2CH2N(CH2)4N), 2.37 (t, J = 6.7 Hz; 4H, NCH2CH2CH2N(CH2)4N), 2.57 (s, 8H, NCH2CH2CH2N(CH2)4N), 2.84 (t, J = 7.1 Hz, 4H; NCH2CH2CH2N(CH2)4N), 3.79 (s, 6H, OCH3), 7.02 (d, J = 8.8 Hz, 4H, H-3’, Ar), 7.18 (s, 2H; =CH), 7.44 (d, J = 8.8 Hz, 4H, H-2’, Ar). 13C NMR (75 MHz, DMSO-d6) δ (ppm) = 23.39 (NCH2CH2CH2N(CH2)4N), 37.41 (NCH2CH2CH2N(CH2)4N), 52.03 (NCH2CH2CH2N(CH2)4N), 54.42 (NCH2CH2CH2N(CH2)4N), 55.27 (OCH3), 114.58 (C-3’), 125.04 (=CH), 127.28 (C-1’), 130.99 (C=), 131.44 (C-2’), 159.77 (C-4’), 201.61 (C=N, C-2), 206.74 (C=O, C-4). HRMS, m/z = 635.2396 found (calculated for C32H39N6O4S2, [M+H]+ requires 635.1819). Anal. Calcd. for C32H38N6O4S2: C, 60.54; H, 6.03. Found: C, 60.57; H, 6.07.
(5Z,5’Z)-2,2’-[Piperazine-1,4-diylbis(propane-3,1-diylimino)]bis[5-(1,3-benzodioxol-5-ylmethylene)-1,3-thiazol-4(5H)-one]: (1,4-Bis(3-{[(5Z)-5-(1,3-benzodioxol-5-ylmethylene)-4-oxo-4,5-dihydro-1,3-thiazol-2-yl]amino}propyl)piperazine, 5e)
Prepared following General procedure III from 5-(1,3-benzodioxol-5-ylmethylidene)-2-thioxothiazolidin-4-one (3c) (1.061 g, 4 mmol) and 1,4-bis(3-aminopropyl)piperazine (4b) (0.4 g, 2 mmol) in hexane (2 ml) at 120°C to give 5e in 15% yield as a brown powder. Mp = 222–224°C. 1H NMR (300 MHz, DMSO-d6) δ (ppm) = 1,69 (quint, J = 6.9 Hz, 4H, NCH2CH2CH2N(CH2)4N), 2.37 (t, J = 6.6 Hz, 4H, NCH2CH2CH2N(CH2)4N), 2.37 (s, 8H, NCH2CH2CH2N(CH2)4N), 2.83 (t, J = 7.1 Hz, 4H, NCH2CH2CH2N(CH2)4N), 6.07 (s, 4H, OCH2O), 7.02 (m, 6H, H-2’, H-5’, H-6’, Ar), 7.14 (s, 2H, =CH). 13C NMR (75 MHz, DMSO-d6) δ (ppm) = 23.47 (NCH2CH2CH2N(CH2)4N), 37.45 (NCH2CH2CH2N(CH2)4N), 52.22 (NCH2CH2CH2N(CH2)4N), 54.54 (NCH2CH2CH2N(CH2)4N), 101.52 (OCH2O), 108.74 (C-2’), 108.82 (C-5’), 124.21 (C-6’), 124.84 (=CH), 147.72 (C-4’), 147.89 (C-3’), 182.66 (C=, C-5), 202.14 (C=N, C-2), 206.72 (C=O, C-4). HRMS, m/z = 663.1981 found (calculated for C32H35N6O6S2, [M+H]+ requires 663.7790). Anal. Calcd. for C32H34N6O6S2: C, 57.99; H, 5.17. Found: C, 57.92; H, 5.16.
In vitro kinase inhibition assays
Buffer A: 10 mM MgCl2, 1 mM EGTA, 1 mM DTT, 25 mM Tris-HCl pH 7.5, 50 μg heparin/ml. Buffer C: 60 mM ß-glycerophosphate, 15 mM p-nitrophenylphosphate, 25 mM Mops (pH 7.2), 5 mM EGTA, 15 mM MgCl2, 1 mM DTT, 1 mM sodium vanadate, 1 mM phenylphosphate. Kinase activities were assayed in Buffer A or C, at 30°C, at a final ATP concentration of 15 μM. Blank values were subtracted and activities expressed in % of the maximal activity, i.e. in the absence of inhibitors. Controls were performed with appropriate dilutions of DMSO. The kinase peptide substrates were obtained from Millegen (Labege, France). DYRK1A and DYRK2 (human, recombinant, expressed in E. coli as a GST fusion protein) were purified by affinity chromatography on glutathione-agarose and assayed in buffer A (+ 0.5 mg BSA /ml) using Woodtide (KKISGRLSPIMTEQ) (1.5 µg/assay) as a substrate, in the presence of 15 µM [γ-33P] ATP (3,000 Ci/mmol; 10 mCi/ml) in a final volume of 30 μl. After 30 min incubation at 30°C, the reaction was stopped by harvesting onto P81 phosphocellulose papers (Whatman) using a FilterMate harvester (Packard) and were washed in 1% phosphoric acid. Scintillation fluid was added and the radioactivity was measured in a Packard counter. CLK1 and CLK3 (human, recombinant, expressed in E. coli as GST fusion proteins) were assayed in buffer A (+ 0.15 mg BSA /ml) with RS peptide (GRSRSRSRSRSR) (1μg/assay). CDK5/p25 (human, recombinant) was prepared as previously described [26]. Its kinase activity was assayed in buffer C, with 1 μg histone H1 /ml. GSK-3α/β (porcine brain, native) was assayed in Buffer A and using a GSK-3 specific substrate (GS-1: YRRAAVPPSPSLSRHSSPHQSpEDEEE) (pS stands for phosphorylated serine) [20].
Cell culture and survival assays
Skin diploid fibroblastic cells were provided by BIOPREDIC International Company (Rennes, France). Caco-2 cells and Huh7 cells were obtained from the ECAC collection. Cells were grown according to ECAC recommendations. The RLEC-F1 clone was derived from an established rat biliary epithelial cell line as previously described [21]. The toxicity test of the compounds on these cells was as follows: 4 × 103 cells were seeded in 96 multiwell plates and left for 24 h for attachment, spreading, and growing. Then they were exposed for 48 h to increasing concentrations of the compounds, ranging from 0.1 to 25 μL in a final volume of 80 μL of culture medium. They were then fixed with 4% paraform-aldehyde solution and the nuclei were stained with Hoechst 3342 and counted using automated imaging analysis (Simple PCI software).
Acknowledgments
One of us (K. W. C.) wishes to thank the “Agence Universitaire de la Francophonie AUF, contract N°0486” for a research fellowship. Financial support of this program carried out under the French National Cancer Institute “Cancéropôle Grand Ouest” by contracts PRIR 04-8390 and ACI 04-2254, is gratefully acknowledged.
Footnotes
This article is available from: http://dx.doi.org/10.3797/scipharm.1206-04
Authors’ Statement
Competing Interests
The authors declare no conflict of interest.
References
- [1].Mentgen T, Steuer C, Klein CD. Privileged Scaffolds or Promiscuous Binders: A comparative Study on Rhodanines and Related Heterocycles in Medicinal Chemistry. J Med Chem. 2012;55:743–753. doi: 10.1021/jm201243p. http://dx.doi.org/10.1021/jm201243p. [DOI] [PubMed] [Google Scholar]
- [2].Nasr MNA, Said SA. Novel 3,3a,4,5,6,7-Hexahydroindazole and Arylthiazolylpyrazoline derivatives as Anti-inflammatory Agents. Arch Pharm. 2003;336:551–559. doi: 10.1002/ardp.200300796. http://dx.doi.org/10.1002/ardp.200300796. [DOI] [PubMed] [Google Scholar]
- [3].Hu Y, Helm J-S, Chen L, Ginsberg C, Gross B, Kraybill B, Tiyanont K, Fang X, Wu T, Walker S. Identification of selective inhibitors for the glycosyltransferase MurG via high-throughput screening. Chem Biol. 2004;11:703–711. doi: 10.1016/j.chembiol.2004.02.024. http://dx.doi.org/10.1016/j.chembiol.2004.02.024. [DOI] [PubMed] [Google Scholar]
- [4].Martin L, Rabasseda X, Castaner J. Darbufelone Mesilate. Drugs Future. 1999;24:853–857. http://dx.doi.org/10.1358/dof.1999.024.08.534526. [Google Scholar]
- [5].Johnson A-R, Marletta M-A, Dyer RD. Slow-binding inhibition of human prostaglandin endoperoxide synthase-2 with darbufelone, an isoform-selective antiinflammatory di-tert-butyl phenol. Biochemistry. 2001;40:7736–7745. doi: 10.1021/bi002343f. http://dx.doi.org/10.1021/bi002343f. [DOI] [PubMed] [Google Scholar]
- [6].Inagaki M, Tsuri T, Jyoyama H, Ono T, Yamada K, Kobayashi M, Hori Y, Arumura A, Yasui K, Ohno K, Kakudo S, Koizumi K, Suzuki R, Kato M, Kawai S, Matsumoto S. Novel antiarthritic agents with 1,2-isothiazolidine-1,1-dioxide (γ-sultam) skeleton: cytokine suppressive dual inhibitors of cyclooxygenase-2 and 5-lipoxygenase. J Med Chem. 2000;43:2040–2048. doi: 10.1021/jm9906015. http://dx.doi.org/10.1021/jm9906015. [DOI] [PubMed] [Google Scholar]
- [7].Teraishi F, Wu S, Zhang L, Guo W, Davis J-J, Dong F, Fang B. Identification of a novel synthetic thiazolidin compound capable of inducing c-Jun NH2-terminal kinase-dependent apoptosis in human colon cancer cells. Cancer Res. 2005;65:6380–6387. doi: 10.1158/0008-5472.CAN-05-0575. http://dx.doi.org/10.1158/0008-5472.CAN-05-0575. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [8].Cutshall N-S, O’Day C, Prezhdo M. Rhodanine derivatives as inhibitors of JSP-1. Bioorg Med Chem Lett. 2005;15:3374–3382. doi: 10.1016/j.bmcl.2005.05.034. http://dx.doi.org/10.1016/j.bmcl.2005.05.034. [DOI] [PubMed] [Google Scholar]
- [9].Hotta N, Akanuma Y, Kawamori R, Matsuoka K, Oka Y, Shichiri M, Toyata T, Nakoshima M, Yoshimura I, Sakamoto N, Shigeta Y. Long-Term clinical effects of Epalrestat, an aldose reductase inhibitor, on diabetic peripheral neuropathy: The 3-year, multicenter, comparative aldose reductase inhibitor-diabetes complications trial. Diabetes Care. 2006;29:1538–1541. doi: 10.2337/dc05-2370. http://dx.doi.org/10.2337/dc05-2370. [DOI] [PubMed] [Google Scholar]
- [10].Zhang J, Yang P-L, Gray NS. Targeting cancer with small molecule kinase inhibitors. Nat Rev Cancer. 2009;9:28–39. doi: 10.1038/nrc2559. http://dx.doi.org/10.1038/nrc2559. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [11].Galons H, Oumata N, Meijer L. Cyclin-dependent kinase inhibitors: a survey of recent patent literature. Exp Opin Ther Patents. 2010;20:377–404. doi: 10.1517/13543770903524284. http://dx.doi.org/10.1517/13543770903524284. [DOI] [PubMed] [Google Scholar]
- [12].Druker B-J, Talpaz M, Resta D-J, Peng B, Buchdunger E, Ford J-M, Lydon N-B, Kantarjian H, Capdeville R, Ohnojones S, Sawyers C-L. Efficacy and Safety of a Specific Inhibitor of the BCR-ABL Tyrosine Kinase in Chronic Myeloid Leukemia. N Engl J Med. 2001;344:1031–1037. doi: 10.1056/NEJM200104053441401. http://dx.doi.org/10.1056/NEJM200104053441401. [DOI] [PubMed] [Google Scholar]
- [13].Bazureau J-P, Draye M, editors. Ultrasound and Microwave: Recent advances in organic chemistry. Transworld Research Signpost; Kerala: 2011. [Google Scholar]
- [14].Guihéneuf S, Paquin L, Carreaux F, Durieu E, Meijer L, Bazureau JP. An efficient approach to dispacamide A and its derivatives. Org Biomol Chem. 2012;10:978–987. doi: 10.1039/c1ob06161e. http://dx.doi.org/10.1039/c1ob06161e. [DOI] [PubMed] [Google Scholar]
- [15].Bourahla K, Derdour A, Rahmouni M, Carreaux F, Bazureau JP. A practical access to novel 2-amino-5-arylidene-1,3-thiazol-4(5H)-ones via sulfur/nitrogen displacement under solvent-free microwave irradiation. Tetrahedron Lett. 2007;48:5785–5789. http://dx.doi.org/10.1016/j.tetlet.2007.06.078. [Google Scholar]
- [16].Kimura R, Kamino K, Yamamoto M, Nuripa A, Kida T, Kazui H, Hashimoto R, Tanaka T, Kudo T, Yamagata H, Tabara Y, Miki T, Akatsu H, Kosaka K, Funakoshi E, Nishitomi K, Sakaguchi G, Kato A, Hattori H, Uema T, Takeda M. The DYRK1A gene, encoded in chromosome 21 Down syndrome critical region, bridges between β-amyloid production and tau phosphorylation in Alzheimer disease. Hum Mol Genet. 2007;16:15–23. doi: 10.1093/hmg/ddl437. http://dx.doi.org/10.1093/hmg/ddl437. [DOI] [PubMed] [Google Scholar]
- [17].Debdab M, Carreaux F, Renault S, Soundararajan M, Federov O, Lozach O, Babault L, Baratte B, Ogawa Y, Hagiwara M, Einsenreich A, Rauch U, Knapp S, Meijer L, Bazureau JP. Design, synthesis and biological evaluation of leucettines, a class of potent CLK and DYRK kinases inhibitors derived from the marine sponge leucettamine B. Modulation of alternative RNA splicing. J Med Chem. 2011;54:4172–4186. doi: 10.1021/jm200274d. http://dx.doi.org/10.1021/jm200274d. [DOI] [PubMed] [Google Scholar]
- [18].Besyadetskaya EI, Kuz’min Y. Synthesis and properties of bicyclic compounds containing piperazine and thiazolidine rings. Farmatsevt Zhurnal (Kiev) 1973;28:34–36. [PubMed] [Google Scholar]
- [19].Besyadetskaya EI. Synthesis of 5-substituted N,N-bis(2-thiazolidin-4-one-2-yl)piperazines. Farmatsevt Zhurnal (Kiev) 1972;27:30–32. [Google Scholar]
- [20].Primot A, Baratte B, Gompel M, Borgne A, Liabeuf S, Romette J-L, Costantini F, Meijer L. Purification of GSK-3 by affinity chromatography on immobilised axin. Protein Expression Purif. 2000;20:394–404. doi: 10.1006/prep.2000.1321. http://dx.doi.org/10.1006/prep.2000.1321. [DOI] [PubMed] [Google Scholar]
- [21].Nakabayashi H, Taketssa K, Miyano K, Yamane T, Sato J. Growth of Human Hepatoma Cell Lines with Differentiated Functions in Chemically Defined Medium. Cancer Res. 1982;42:3858–3863. http://www.ncbi.nlm.nih.gov/pubmed/6286115. [PubMed] [Google Scholar]


