Next Article in Journal
Thermodynamic Exercises for the Kinetically Controlled Hydrogenation of Carvone
Previous Article in Journal
Aaptamine: A Versatile Marine Alkaloid for Antioxidant, Antibacterial, and Anticancer Therapeutics
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Efficient Two-Step Synthesis of Novel Pyrimido[4,5-d] Pyrimidines with Potent Neuroprotective, Antioxidant, and Aβ Anti-Aggregation Properties

1
Laboratory of Applied Chemistry, Heterocycles, Lipids and Polymers, Faculty of Sciences of Sfax, University of Sfax, B.P. 802, Sfax 3000, Tunisia
2
Laboratoire Matériaux, Traitement et Analyse (LMTA), Institut National de Recherche et d’Analyse Physico-Chimique (INRAP), Ariana 2020, Tunisia
3
Université de Franche-Comté, EFS, INSERM, UMR RIGHT, F-25000 Besançon, France
4
Université de Franche-Comté, UMR INSERM 1322 LINC, F-25000 Besançon, France
5
Laboratory of Medicinal Chemistry (IQOG, CSIC), C/Juan de la Cierva 3, 28006 Madrid, Spain
6
Center for Biomedical Network Research on Rare Diseases (CIBERER), CIBER, ISCIII, 28006 Madrid, Spain
*
Authors to whom correspondence should be addressed.
Chemistry 2024, 6(4), 695-705; https://doi.org/10.3390/chemistry6040041
Submission received: 18 July 2024 / Revised: 1 August 2024 / Accepted: 2 August 2024 / Published: 7 August 2024
(This article belongs to the Section Medicinal Chemistry)

Abstract

:
Eleven new differently substituted N,7-diphenylpyrimido [4,5-d]pyrimidin-4-amines 4ak were synthesized from readily available reagents in a simple and inexpensive two-step procedure with yields up to 57%. Neuroprotective analysis against H2O2 and analysis using ORAC assays identified compounds 4g, 4i and 4j as promising antioxidant compounds. These compounds also showed potent inhibition of Aβ1–42 self-aggregation, and suitable physicochemical properties predicted by Datawarior software V6.1.0, this biological activity and physicochemical property being of great interest for pathologies linked to oxidative stress, such as Alzheimer’s disease.

1. Introduction

Over the past decade, we have been actively involved in the design, synthesis and pharmacological evaluation of diversely functionalized heterocyclic derivatives as multi-target small molecules for the potential treatment of Alzheimer’s disease (AD) [1,2,3]. Our approach has always been motivated by preparing these molecules by using sustainable synthetic processes, either by reducing the number of steps (2 or 3) [4,5] or by favoring multicomponent reactions [2,3].
Among the many neuropathological features of AD (cholinergic deficit, intracellular neurofibrillary tangles and beta-amyloid (Aβ) deposition), oxidative stress (OS) plays a fundamental role in the biological events leading to this neurodegenerative disease [6,7]. In fact, histopathological data obtained from extracellular Aβ deposits have shown that the release of reactive oxygen species (ROS) can severely damage mitochondria and other cellular contents of neurons [8,9,10]. In light of these observations, the search for new and more effective neuroprotective and antioxidant agents for the treatment of AD has been intense in recent years [11,12,13,14].
In this context, we have developed several new compounds based on azaheterocyclic scaffolds [15,16]. In fact, nitrogen-containing heterocycles have been widely used in medicinal chemistry to find new bioactive molecules for AD [17,18,19,20]. In particular, our work reported in 2020 [21] describes the discovery of a new pyridopyrimidine scaffold as dual-target small molecules for AD therapy. Continuing our work in this field, we were interested in exploring the nitrogen bioisosteres of pyridopyrimidines, in particular pyrimidopyrimidines. Indeed, compounds bearing the pyrimidopyrimidine scaffold have been reported to exhibit a wide range of biological activities, such as antibacterial, antiviral, antitumor, antiallergic, antihypertensive and hepatoprotective effects, making this scaffold of particular interest [22,23].
In this report, we describe the synthesis of eleven new pyrimidopyrimidines 4ak (Scheme 1) and the evaluation of their neuroprotective and antioxidant properties using ORAC assays. Our results have identified compounds 4g, 4i and 4j as promising neuroprotective and antioxidant agents, which also exhibit potent inhibition of Aβ self-aggregation.

2. Results

2.1. Synthesis

The synthesis of the new diphenylpyrimido [4,5-d]pyrimidin-4-amines 4ak was carried out in two steps, as shown in Scheme 1, from moderate to good overall results.
The first step involves the reaction of readily available 4-amino-2,6-alkylpyrimidine-5-carbonitrile 1ae with triethylorthoester derivatives at reflux. Next, in the second step, the resulting intermediate imidates 2af were then reacted with various commercially available substituted anilines in refluxing toluene in the presence of a catalytic amount of acetic acid. It is interesting to note that precursors bearing no substituent at position 6 of the pyrimidinopyrimidine nucleus and on the aromatic ring attached at position 8 always gave to the best yields observed in this reaction, ranging between 47% and 57% (Table 1).
A p-tolyl substituent in position 7 (R3 = Me) consistently leads to low yields, as shown by compounds 4d (16%) and 4e (20%). Similarly, the presence of a chlorine or methoxy group in para position of the aromatic ring attached to the amine also results in modest yields, as demonstrated by compounds 4i (22%) and 4j (20%).
All new compounds showed excellent analytical and spectroscopic data, in good agreement with the expected values (see Section 3, and Supplementary Materials).

2.2. Biological Evaluation

2.2.1. In Cellulo Evaluation of the Neuroprotective Activity of Compounds 4ak

The neuroprotective potential of compounds 4ak was evaluated by examining their ability to inhibit cell death in the human neuroblastoma cell line SH-SY5Y induced by hydrogen peroxide (H2O2). H2O2 is known to generate exogenous free radicals, leading to a cytotoxic effect that reduces cell viability to 65% at a concentration of 200 mM compared to untreated cells. Therefore, compounds that can prevent or mitigate these damaging effects are considered to have neuroprotective properties [24].
Before performing the neuroprotection assay, we evaluated the direct toxicity of compounds 4ak over a concentration range from 0.1 to 10 μM. Cell viability was measured using the MTT assay [25], which indicated that there were no cytotoxic effects on SH-SY5Y cells at concentrations up to 5 μM.
Subsequently, compounds to be tested for neuroprotection were evaluated at 0.1, 1 and 5 μM. The results are summarized in Table 2.
These results highlight the significant efficacy of several compounds, particularly molecules 4g, 4hj. Compound 4g demonstrated a significant and strong neuroprotective effect in response to H2O2 with percentages of 47%, 68%, and 77% at concentrations of 0.1 μM, 1 μM, and 5 μM, respectively. Compound 4h exerts its neuroprotective power mainly at 0.1 μM with 66%, but remains significantly neuroprotective, though at lower levels: 39% at 1 μM and 26% at 5 μM. Compound 4i, undoubtedly the best neuroprotectant, completely or almost completely reversed the toxicity of H2O2 (118% at 1 μM and 95% at 5 μM). However, it remains an excellent neuroprotectant even at 0.1 μM with a percentage of 68%. Finally, compound 4j shows a significant neuroprotective effect only at concentrations of 0.1 μM and 1 μM with 67% and 46%, respectively.
From a structure–activity relationship perspective, it is clear that these four compounds share similarities. Indeed, a double substitution at positions 2 and 6 with an alkyl group seems to be essential for the neuroprotective activity, combined with a substituent of whatever nature (alkyl, methoxy or chlorine) in the para position of the aromatic ring attached to the aromatic amine.

2.2.2. Antioxidant Analysis

The antioxidant capacity of compounds 4ak to scavenge peroxyl radicals was evaluated using the ORAC-FL (Oxygen Radical Absorbance Capacity by Fluorescence) assay [26,27]. Fluorescein (FL) was used as the fluorescent probe and 6-hydroxy-2,5,7,8-tetramethylchroman-2-carboxylic acid (Trolox) as the standard compound. Results were expressed in micromoles of Trolox equivalents (TE). Melatonin and Trolox were used as positive controls, with ORAC values of 2.4 TE and 0.99 TE, respectively, in agreement with previously reported values [1,28].
As shown in Table 2, three compounds showed no activity, while the remaining compounds showed a modest ability to trap peroxyl radicals. The ORAC values ranged from 0.07 TE (compound 4a) to 1.64 TE (compound 4j). Among the best neuroprotective compounds, 4g, 4i and 4j showed antioxidant activities with ORAC values of 0.24, 0.34 and 1.64 TE, respectively. In particular, compound 4j is 1.6-fold more active than Trolox and has almost 70% of the antioxidant activity of melatonin, one of the best-known antioxidants.

2.2.3. Inhibition of Self-Induced Aβ1–42 Aggregation by Compounds 4g, 4i and 4j

The accumulation and aggregation of Aβ1–42 in the brain may lead to neurotoxicity and contribute to the development of AD. According to the amyloid cascade hypothesis, Aβ aggregation and deposition are responsible for the formation of senile plaques, cell damage and dementia in AD [29]. Therefore, compounds that can prevent the self-aggregation of Aβ1–42 may slow the progression of AD. Three compounds, selected for their neuroprotective and antioxidant activities, were tested for their ability to inhibit self-induced Aβ1–42 aggregation. The thioflavin T-based fluorometric assay [30] was used to evaluate this inhibitory effect and the results are summarized in Figure 1.
Based on the results of this experiment, compounds 4g, 4i and 4j showed significant inhibition of Aβ1–42 aggregation, with inhibition rates of 16.3%, 25.7% and 56.6%, respectively, compared to 73.3% for curcumin used as a reference standard at a concentration of 2.5 μM. These results suggest that our compounds have multifunctional properties.
The Thioflavin-T fluorescence method was used. The values are expressed as the mean ± SD of at least three independent measurements. All values were obtained at a compound concentration of 2.5 μM.

2.2.4. ADME Studies

In order to predict the physicochemical properties of the compounds, we used ‘Data Warrior V6.1.0’, physical and chemical property visualization and analysis software developed by Idorsia Pharmaceuticals Ltd., 4123 Allschwil, Switzerland. This tool allows the prediction of drug-like properties using various parameters from Lipinski’s rule of five, including molecular weight, LogP, LogS, hydrogen bond acceptors, hydrogen bond donors and topological polar surface area (TPSA).
As shown in Table 3, all compounds had molecular weights below 500 g/mol, a value typical of most oral drugs and used as the basis for Lipinski’s rule of five. Lipophilicity, a critical physicochemical property, determines whether a molecule can cross biological membranes with a predicted CLogP less than 5. Interestingly, almost all the compounds showed suitable lipophilicity, with CLogP values ranging from 3.7393 to 4.8832, except for compound 4h which had a slightly higher CLogP of 5.0367. This value remains well below the upper limit of 6.5 for druggable compounds.
Most compounds had a number of hydrogen bond donors and acceptors in accordance with Lipinski’s rule of five. The number of hydrogen bond donors was below 5 and the number of acceptors was also below 10. Notably, compounds 4j and 4k had slightly more hydrogen bond acceptors than the values suggested by Lipinski’s rule.
TPSA represents the van der Waals surface area of the polar atoms of the molecule. In other words, PSA is the surface area associated with heteroatoms (such as oxygen, nitrogen and phosphorus atoms) and polar hydrogen atoms. According to Veber’s rule, the polar surface area should not exceed 140 Å2. It is noteworthy that all compounds had a TPSA of less than 75 Å2.
Toxicity, particularly cardiac toxicity due to inhibition of the hERG (human Ether-à-go-go-Related Gene) channel, is a significant concern in drug development. The hERG channel plays a crucial role in the repolarization of the cardiac action potential, and its inhibition can lead to potentially fatal arrhythmias, which can result in sudden death [31,32]. In this context, hERG inhibition was predicted by PreADMET, one of several web-based applications developed to meet the need for rapid prediction of drug-likeness and ADME/Tox data, available as open source.
As shown in Table 3, no compounds showed a high risk of hERG inhibition. Three compounds showed a low risk, while eight compounds displayed a medium risk. It is worth noting that compounds with substituents on the aromatic rings attached to the pyrimidopyrimidine core always lead to a medium risk, whereas the absence of substituents leads to a low risk.

3. Materials and Methods

Melting points (°C) were determined using a Kofler hot bench and are uncorrected. Analytical thin-layer chromatography (TLC) was performed on silica gel precoated aluminium sheets (type 60 F254, 0.25 mm thickness, from Merck, Darmstadt, Germany) to monitor the progress of the reactions and to assess the purity and homogeneity of the synthesized products. High-resolution mass spectra (HRMS) were obtained using a Bruker micrOTOF-Q II spectrometer (Bruker Daltonics, Billerica, MA, USA) with positive electrospray ionization time-of-flight at the UCA Clermont Ferrand, France. Nuclear magnetic resonance (NMR) spectra were obtained on a Bruker DRX-400 Avance spectrometer (operating at 400 MHz for 1H and 100 MHz for 13C) using dimethylsulfoxide (DMSO-d6) as solvent and tetramethylsilane (TMS) as internal standard. Chemical shifts are given in parts per million (ppm) and the multiplicities of the 1H NMR signals are given as follows: s (singlet), d (doublet), t (triplet), q (quartet) and m (multiplet). Coupling constants are given in hertz (Hz).

3.1. Synthesis of Compounds 2ak

General experimental procedure for synthesis of imidate (2af)
A mixture of 4-amino-2,6-arylpyrimidine-5-carbonitrile 1ae (1 mmol) and triethylorthoester (4 mmol) was refluxed for 3 h without special atmospheric precautions. After cooling, the solvent was evaporated. The resulting solid was collected by filtration and recrystallized from ether.
Ethyl N-(5-cyano-6-methyl-2-phenylpyrimidin-4-yl)acetimidate 2a
Beige solid; Yield: 74%; mp: 66–68 °C; IR: υ (cm−1) 2210, 1660; 1H NMR (CDCl3, 400 MHz) [δ ppm]: 8.38 (d, 3J = 8 Hz, 2 H), 7.45–7.39 (m, 3 H), 4.32 (q, 3J = 7.1 Hz, 2 H), 2.68 (s, 3 H), 2.07 (s, 3 H), 1.33 (t, 3J = 7.1 Hz, 3 H); 13C NMR (CDCl3, 100 MHz) [δ ppm]: 170.39, 167.42, 165.02, 164.17, 135.39, 130.69, 127.91, 127.46, 114.25, 97.18, 62.68, 22.52, 16.96, 12.83. HRMS (ESI, M+H+) Calcd for C16H17N4O: 281.13969. Found: 281.1396.
Ethyl N-(5-cyano-2-phenylpyrimidin-4-yl)acetimidate 2b
Beige solid; Yield: 94%; mp: 90–92 °C; IR: υ (cm−1): 2210, 1660; 1H NMR (CDCl3, 400 MHz) [δ ppm]: 8.88 (s, 1 H), 8.47 (d, 3J = 8.3 Hz, 2 H), 7.56–7.50 (m, 3 H), 4.43 (q, 3J = 7.1 Hz, 2 H), 2.20 (s, 3 H), 1.44 (t, 3J = 7.1 Hz, 3 H); 13C NMR (CDCl3, 100 MHz) [δ ppm]: 167.88, 166.59, 166.51, 161.11, 136.36, 132.08, 129.06, 128.68, 128.55, 115.07, 99.48, 64, 18.14,13.92. HRMS (ESI, M+H+) Calcd for C15H15N4O: 267.12404. Found: 267.1238.
Ethyl N-(5-cyano-2-phenylpyrimidin-4-yl)Propionimidate 2c
Viscous oil; Yield: 71%; IR: υ (cm−1): 2227, 1650; 1H NMR(CDCl3, 400 MHz) [δ ppm]: 8.87 (s, 1 H), 8.47 (d, 3J = 8.2, 2 H), 7.53 (m, 3 H), 4.42 (q, 3J = 7.1 Hz, 2 H), 2.48 (q, 3J = 7.6 Hz, 2 H), 1.44 (t, 3J = 7.1 Hz, 3 H), 1.26 (t, 3J = 7.6 Hz, 3 H); 13C NMR (CDCl3, 100 MHz) [δ ppm]: 169.65, 167.85, 166.52, 161.04, 136.38, 132.06, 129.05, 128.90, 128.69, 128.58, 115.15, 99.29, 63.89, 25.47, 13.90, 10.67. HRMS (ESI, M+H+) Calcd for C16H17N4O: 281.13969. Found: 281.1397.
Ethyl N-(5-cyano-6-ethyl-2-phenylpyrimidin-4-yl)acetimidate 2d
Beige solid; Yield: 90%; mp: 90–92 °C; IR υ (cm−1):2220, 1625; 1H NMR (CDCl3, 400 MHz) [δ ppm]: 8.50 (d, 3J = 8.1 Hz, 2 H), 7.56–7.48 (m, 3 H), 4.42 (q, 3J = 7.1 Hz, 2 H), 3.07 (q, 3J = 7.5 Hz, 2 H), 2.17 (s, 1 H), 1.46–1.41 (m, 6 H); 13C NMR (CDCl3, 100 MHz) [δ ppm]: 176.00, 168.06, 166.09, 165.51, 136.73, 131.78, 129.10, 128.56, 115.25, 97.58, 63.77, 30.18, 18.06, 13.97, 12.24. HRMS (ESI, M+H+) Calcd for C17H19N4O: 295.15534. Found: 295.1553.
Ethyl N-(5-cyano-2-(p-tolyl)pyrimidin-4-yl)acetimidate 2e
Beige solid; Yield: 53%; mp: 254–256 °C; IR υ (cm−1): 2225, 1641; 1H NMR (CDCl3, 400 MHz) [δ ppm]: 8.85 (s, 1 H), 8.36 (d, 3J = 8.2 Hz, 2 H), 7.32 (d, 3J = 8.2 Hz, 2 H), 4.42 (q, 3J = 7.1 Hz, 2 H), 2.45 (s, 3 H),2.18 (s, 3 H), 1.43 (t, 3J = 7.1 Hz, 3 H); 13C NMR (CDCl3, 100 MHz) [δ ppm]: 167.84, 166.55, 166.51, 161.08, 142.79, 133.66, 129.49, 129.08, 115.23, 99.05, 63.96, 21.62, 18.1, 13.95. HRMS (ESI, M+H+) Calcd for C16H17N4O: 281.13969. Found: 281.1394.
Ethyl N-(5-cyano-6-methyl-2-(p-tolyl)pyrimidin-4-yl)acetimidate 2f
White solid; Yield: 45%; mp: 220–222 °C; IR υ (cm−1): 2212, 1662; 1H NMR (CDCl3, 400 MHz) [δ ppm]: 8.37 (d, 3J = 7.4 Hz, 2 H), 7.30 (d, 3J = 7.4 Hz, 2 H), 4.41 (q, 3J = 7.1 Hz, 2 H), 2.76 (s, 3 H), 2.45 (s, 3 H), 2.15 (s, 3 H), 1.43 (t, 3J = 7.1 Hz, 3 H); 13C NMR (CDCl3, 100 MHz) [δ ppm]: 171.40, 168.53, 166.03, C7 165.38, 142.42, 133.87, 129.37, 129.08, 115.49, 97.92, 63.75, 23.64, 21.59, 18.05, 13.96. HRMS (ESI, M+H+) Calcd for C17H19N4O: 295.15534. Found: 295.1553.

3.2. Synthesis of Compounds 4ak

General experimental procedure for synthesis of pyrimido [4,5-d]pyrimidin-4-amines 4a–k
A mixture of imidate 2 (1 mmol) and aniline derivatives 3 (1 mmol) was refluxed for 6h with acetic acid (1 mmol) as catalyst in anhydrous toluene (3 mL) without special atmospheric precautions. After evaporation of the solvent, the resulting product was filtered and recrystallized from ether to give the desired compounds 4ak.
2-Methyl-N,7-diphenylpyrimido [4,5-d]pyrimidin-4-amine (4a):
Beige solid; Yield: 57%; mp > 260 °C; IR υ (cm−1): 3326, 1610; 1H NMR (DMSO-d6,400 MHz) [δ ppm]: 10.38 (s, 1 H), 10.01 (s, 1 H), 8.53 (d, 3J = 7.2 Hz, 2 H), 7.89 (d, 3J = 7.2 Hz, 2 H), 7.59 (m, 3 H), 7.44 (t, 3J = 7.5 Hz, 2 H), 7.19 (t, 3J = 7.5 Hz, 1 H), 2.55 (s, 3 H); 13C NMR (DMSO-d6, 100 MHz) [δ ppm]: 172.53, 166.05, 163.27, 158.80, 158.19, 138.89, 137.24, 132.16, 129.27, 129.10, 129.01, 124.93, 122.85, 105.07, 27.47. HRMS (ESI, M+H+) Calcd for C19H16N5: 314.14002. Found: 314.1400.
2-Ethyl-N,7-diphenylpyrimido [4,5-d]pyrimidin-4-amine (4b):
Beige solid; Yield: 50%; mp: 264–266 °C; IR υ (cm−1): 3414, 1626; 1H NMR(DMSO-d6, 400 MHz) [δ ppm]: 10.40 (s, 1 H), 10.04 (s, 1 H), 8.55 (d, 3J = 7.8 Hz, 2 H), 7.92 (d, 3J = 7.8 Hz, 2 H), 7.61–7.59 (m, 3 H), 7.45 (t, 3J = 7.7 Hz, 2 H), 7.19 (t, 3J = 7.7 Hz, 1 H), 2.84 (q, 3J = 7.5 Hz, 2 H),1.32 (t, 3J = 7.5 Hz, 3 H); 13C NMR(DMSO-d6, 100 MHz) [δ ppm]: 166.12, 158.91, 158.26, 137.24, 132.21, 129.31, 129.09, 129.04, 124.89, 122.72, 105.33, 33.43, 12.40. HRMS (ESI, M+H+) Calcd for C20H18N5: 328.15567. Found: 328.1555.
5-Ethyl-2-methyl-N,7-diphenylpyrimido [4,5-d]pyrimidin-4-amine (4c):
Beige solid; Yield: 35%; mp: 224–226 °C; IR υ (cm−1): 3484, 1605; 1H NMR (CDCl3, 400 MHz) [δ ppm]: 8.74 (d, 3J = 7.6 Hz, 2 H), 7.77 (d, 3J = 7.6 Hz, 2 H), 7.73 (s, 1 H), 7.54–7.52 (m, 3 H), 7.46 (t, 3J = 7.5 Hz, 2 H), 7.25 (t, 3J = 7.5 Hz, 1 H), 3.45 (q, 3J = 7.0 Hz, 2 H), 2.73 (s, 3 H), 1.67 (t, 3J = 7.0 Hz, 3 H); 13C NMR (CDCl3, 100 MHz) [δ ppm]: 171.42, 169.48, 165.14, 164.91, 158.20, 137.82, 136.90, 131.67, 129.34, 129.17, 128.49, 125.23, 121.91, 104.17, 32.38, 26.85, 11. HRMS (ESI, M+H+) Calcd for C21H20N5: 342.17132. Found: 342.1714.
2-Methyl-N-phenyl-7-(p-tolyl)pyrimido [4,5-d]pyrimidin-4-amine (4d):
White solid; Yield: 16%; mp: 234 °C; IR υ (cm−1): 3462, 1611; 1H NMR (DMSO-d6, 400 MHz) [δ ppm]: 10.37 (s, 1 H), 10.03 (s, 1 H), 8.43 (d, 3J = 7.9 Hz, 2 H), 7.90 (d, 3J = 7.9 Hz, 2 H), 7.44 (t, 3J = 7.7 Hz, 2 H), 7.38 (d, 3J = 7.8 Hz, 2 H), 7.19 (t, 3J = 7.7 Hz, 1 H), 2.56 (s, 3 H), 2.41 (s, 3 H); 13C NMR (DMSO-d6, 100 MHz) [δ ppm]: 172.40, 166.21, 163.20, 158.83, 158.15, 142.23, 138.98, 134.63, 129.89, 129.08, 129.05, 124.93, 122.88, 104.91, 27.38, 21.57. HRMS (ESI, M+H+) Calcd for C20H18N5: 328.15567. Found: 328.1555.
2,5-Dimethyl-N-phenyl-7-(p-tolyl)pyrimido [4,5-d]pyrimidin-4-amine (4e):
Beige solid; Yield: 20%; mp: 226 °C; IR υ(cm−1): 3478, 1607; 1H NMR (CDCl3, 400 MHz) [δ ppm]: 8.58 (m, 2 H); 7.76 (m, 2 H), 7.45 (m, 2 H), 7.32–7.25 (m, 3 H), 3.20 (s, 3 H), 2.71 (s, 3 H), 2.44 (s, 3 H); 13C NMR (CDCl3, 100 MHz) [δ ppm]: 21.58, 26.84, 27.33, 104.46, 121.90, 125.19, 128.90, 129.12, 129.28, 133.94, 137.80, 142.19, 158.47, 164.76, 165.13, 165.44, 171.56. HRMS (ESI, M+H+) Calcd for C21H20N5: 342.17132. Found: 342.1714.
2,5-Dimethyl-N,7-diphenylpyrimido [4,5-d]pyrimidin-4-amine (4f):
Beige solid; Yield: 50%; mp: 230 °C; IR υ (cm−1): 3291, 1606; 1H NMR (CDCl3,400MHz) [δ ppm]: 8.70 (d, 3J = 7.2 Hz, 2 H), 7.78–7.74 (m, 3 H), 7.52 (d, 3J = 7.7 Hz, 2 H), 7.46 (t, 3J= 7.7 Hz, 2 H), 7.25 (t, 3J = 0.77, 1 H), 3.21 (s, 3 H), 2.73(s, 3 H); 13C NMR (CDCl3, 100 MHz) [δ ppm]: 171.77, 165.41, 165.26, 164.76, 158.52, 137.70, 136.63, 131.68, 129.29, 129.15, 128.50, 125.30, 121.95, 104.62, 27.35, 26.90. HRMS (ESI, M+H+) Calcd for C20H18N5: 328.15567. Found: 328.1554.
2,5-Dimethyl-7-phenyl-N-(p-tolyl)pyrimido [4,5-d]pyrimidin-4-amine (4g):
Beige solid; Yield: 28%; mp:176–178 °C; IR υ (cm−1): 3467, 1606; 1H NMR (CDCl3, 400 MHz) [δ ppm]: 8.65 (d, 3J = 6.6 Hz, 2 H), 7.64 (d, 3J= 8.1 Hz, 2 H), 7.54–7.47 (m, 3 H), 7.24 (d, 3J = 8.1 Hz, 2 H), 3.20 (s, 3 H), 2.66 (s, 3 H), 2.41 (s, 3 H). 13C NMR (CDCl3, 100 MHz) [δ ppm]: 171.65, 165.32, 158.51, 136.59, 135.22, 135.06, 131.66, 129.66, 129.26, 128.49, 122.09, 104.60, 27.35, 26.82, 21.02. HRMS (ESI, M+H+) Calcd for C21H20N5: 342.17132. Found: 342.1713.
5-Ethyl-2-methyl-7-phenyl-N-(p-tolyl)pyrimido [4,5-d]pyrimidin-4-amine (4h):
Beige solid; Yield: 30%; mp: 148–150 °C; IR υ (cm−1): 3460, 1604; 1H NMR (CDCl3, 400 MHz) [δ ppm]: 8.71 (d, 3J = 6.8 Hz, 2 H), 7.63 (d, 3J = 8.1 Hz, 2 H), 7.54–7.50 (m, 3 H), 7.25 (d, 3J = 8.1 Hz, 2 H), 3.44 (q, 3J =7.2 Hz, 2 H), 2.69 (s, 3 H), 2.40 (s, 3 H), 1.65 (t, 3J = 7.2 Hz, 3 H); 13C NMR (CDCl3, 100 MHz) [δ ppm]: 171.12, 169.71, 165.05, 158.18, 136.80, 135.14, 131.64, 129.65, 129.32, 128.46, 122.06, 104.15, 32.29, 26.67, 21.02, 11.57. HRMS (ESI, M+H+) Calcd for C22H22N5: 356.18697. Found: 356.1869.
N-(4-Chlorophenyl)-2,5-dimethyl-7-phenylpyrimido [4,5-d]pyrimidin-4-amine (4i):
Beige solid; Yield: 22%; mp: 162–164 °C; IR υ (cm−1): 3472, 1606; 1H NMR (CDCl3, 400 MHz) [δ ppm]: 8.63 (d, 3J = 6.7 Hz, 2 H), 7.71 (d, 3J = 8.4 Hz, 2 H), 7.55–7.46 (m, 4 H), 7.40 (d, 3J = 8.4 Hz, 2 H), 3.20 (s, 3 H), 2.66 (s, 3 H); 13C NMR (CDCl3, 100 MHz) [δ ppm]: 171.54, 165.38, 158.24, 136.42, 131.80, 130.30, 129.25, 129.12, 128.52, 123.27, 104.61, 27.31, 26.59. HRMS (ESI, M+H+) Calcd for C20H17N5Cl: 362.11670. Found: 362.1166.
N-(3,4-Dimethoxyphenyl)-5-ethyl-2-methyl-7-phenylpyrimido [4,5-d]pyrimidin-4-amine (4j):
Beige solid; Yield: 20%; mp: 178–180 °C; IR υ (cm−1): 3420, 1613; 1H NMR (CDCl3, 400 MHz) [δ ppm]: 8.69 (d, 3J = 2.6 Hz, 2 H), 7.64 (s, 1 H), 7.52–7.49 (m, 3 H), 7.10 (d, 3J = 8.5 Hz, 1 H), 6.91 (d, 3J = 8.5 Hz, 1 H), 3.95 (m, 6 H), 3.47 (q, 3J = 6.8 Hz, 2 H), 2.65 (s, 3 H), 1.65 (t, 3J = 6.8 Hz, 3 H); 13C NMR (CDCl3, 100 MHz) [δ ppm]: 165.03, 158.18, 8.95, 146.66, 136.78, 131.80, 131.59, 129.31, 128.59, 128.38, 114.15, 113.97, 111.20, 111, 107.08, 106.84, 104.11, 55.68, 56.33, 32.33, 11.77, 11.27. HRMS (ESI, M+H+) Calcd for C23H24N5O2: 402.19245. Found: 402.1926.
N-(3,5-Dimethoxyphenyl)-2-methyl-7-phenylpyrimido [4,5-d]pyrimidin-4-amine (4k):
Beige solid; Yield: 47%; mp > 266 °C; IR υ (cm−1): 3326, 1620; 1H NMR (DMSO-d6, 400 MHz) [δ ppm]: 10.27 (s, 1 H), 10.06 (s, 1 H), 8.56 (d, 2 H), 7.61 (m, 3 H), 7.27 (d, 2 H), 6.35 (s, 1 H), 3.80 (s, 6 H), 2.61 (s, 3 H); 13C NMR (DMSO-d6, 100 MHz) [δ ppm]: 172.38, 166.11, 163.25, 160.78, 158.77, 158.17, 140.65, 137.27, 132.17, 129.27, 129.03, 105.11, 100.78, 96.84, 55.66, 27.46. HRMS (ESI, M+H+) Calcd for C21H20N5O2: 374.16115. Found: 374.1609.

3.3. Biological Evaluation

Effect of compounds 4a–k on H2O2 (200 µM)-induced cell death in SH-SY5Y cells. SH-SY5Y cells were seeded in 96-well culture plates at a density of 8 × 104 cells per well in DMEM/F12 (1:1) medium supplemented with 10% fetal bovine serum, 1X non-essential amino acids, 100 units/mL penicillin and 10 mg/mL streptomycin (Dutscher, France). After 48 h of incubation, the cultures were treated with 100 µL of the test compounds or DMSO (0.1%) in the same medium. Following 24 h, the cells were co-incubated with H2O2 (200 µM) with or without the tested compounds at noncytotoxic concentrations for an additional period of 24 h. The percent of cell viability was measured using CellTiter 96 AQueous Non-Radioactive Cell Proliferation (MTS) Assay (Promega, France).
Oxygen Radical Absorbance Capacity Assay. The antioxidant capacity of compounds 4a–k was evaluated by the ORAC-FL assay using fluorescein as a probe [26]. Samples were incubated in a black 96-well plate at 37 °C for 15 min followed by the addition of AAPH and fluorescence was measured every minute for 60 min. Each reaction was run in triplicate. The area under the fluorescence decay curve (AUC) was calculated and normalised to a blank. The ORAC value was derived by comparing the net AUC of the sample with that of Trolox, with results expressed as Trolox equivalents (µmol Trolox equivalent/µmol adduct). Data are expressed as mean ± SD.
Inhibition of Aβ1–42 aggregation. Aβ1–42 was stocked in ultra pure water ([Aβ1–42] = 50 μM). Experiments were performed by incubating the peptide in 50 mM phosphate buffer (pH 7.4) at 37 °C for 24 h (final Aβ concentration = 25 μM) with and without inhibitor (25 μM). The inhibitor was dissolved in DMSO (50 μM) and diluted in the essay buffer (final concentration = 25 μM). Blanks containing inhibitor and ThT were also prepared and evaluated to account for quenching and fluorescence properties. To quantify amyloid fibril formation, the ThT fluorescence method was used.
After incubation, samples were diluted to a final volume of 400 μL ([Aβ1–42] = [inhibitor] = 2.5 μM) with 50 mM glycine-NaOH buffer (pH 8) containing 5 μM ThT. A 300 scan of fluorescence intensity was carried out (λexc = 450 nm; λem = 485 nm) and values at plateau were averaged after subtracting the background fluorescence of 5 μm ThT solution. The fluorescence intensities were compared, and the % inhibition was calculated.

4. Conclusions

In this study we have successfully designed and synthesized eleven new N,7-diphenylpyrimido [4,5-d]pyrimidin-4-amines 4ak by a simple two-step process from moderate to good overall chemical yields. Based on the biological and physicochemical results, compounds 4g, 4i and 4j were identified as multifunctional agents with neuroprotective and antioxidant activities and the ability to inhibit Aβ self-aggregation. In addition, these compounds exhibited suitable physicochemical properties as predicted by the DataWarrior software V6.1.0. Our results suggest that compounds 4g, 4i and 4j hold promise for further research in the treatment of AD. Ongoing work in our laboratories is aimed at developing analogues with improved pharmacological profiles and the results will be reported in due course.

Supplementary Materials

The following supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/chemistry6040041/s1, The NMR, IR and HRMS spectras.

Author Contributions

G.B.A. carried out the synthesis of the molecules. A.-S.J., performed the neuroprotective study and ORAC assay. P.J.B. provided the Physicochemical properties. N.B. carried out the Self-Induced Aβ1–42 Aggregation assay. H.M. supervised the biological assays. E.M. supervised the synthesis of compounds. F.C. and J.M.-C., supervised the project and edited the manuscript. L.I. supervised the project and wrote the manuscript. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by the Regional Council of Franche-Comté (2022Y-13659 and 13660 ACCURATE PROJECT), The PHC Hubert Curien “Utique” of the French Ministry of Europe and Foreign Affairs and the Tunisian Ministry of Higher Education and Scientific Research.

Data Availability Statement

Samples of the compounds are available from the authors.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Benchekroun, M.; Pachón-Angona, I.; Luzet, V.; Martin, H.; Oset-Gasque, M.-J.; Marco-Contelles, J.; Ismaili, L. Synthesis, Antioxidant and Aβ Anti-Aggregation Properties of New Ferulic, Caffeic and Lipoic Acid Derivatives Obtained by the Ugi Four-Component Reaction. Bioorganic Chem. 2019, 85, 221–228. [Google Scholar] [CrossRef] [PubMed]
  2. Ismaili, L.; Bernard, P.J.; Refouvelet, B. Latest Progress in the Development of Multitarget Ligands for Alzheimer’s Disease Based on the Hantzsch Reaction. Future Med. Chem. 2022, 14, 943–946. [Google Scholar] [CrossRef]
  3. Bernard, P.J.; Bellili, D.; Ismaili, L. Calcium Channel Blockers’ Contribution to Overcoming Current Drug Discovery Challenges in Alzheimer’s Disease. Expert Opin. Drug Discov. 2024, 19, 21–32. [Google Scholar] [CrossRef]
  4. Mkaouar, K.; Iriepa, I.; Diez-Iriepa, D.; Marco-Contelles, J.; Ismaili, L.; Chabchoub, F. Synthesis of (±)-cis-1-Aryl-3-Oxo-2,3-Dihydro-1H-benzo[f]chromene-2-carbonitriles and (±)-trans-4-Aryl-2-oxo-3,4-Dihydro-2H-benzo[h]chromene-3-carbonitriles. ChemistrySelect 2019, 4, 12902–12905. [Google Scholar] [CrossRef]
  5. Choura, E.; El Ghali, F.; Bernard, P.J.; Marco-Contelles, J.; Aifa, S.; Ismaili, L.; Chabchoub, F. Synthesis and Biological Evaluation of New Benzochromenopyrimidines for the Therapy of Colon and Lung Cancer. ChemistrySelect 2024, 9, e202303632. [Google Scholar] [CrossRef]
  6. Querfurth, H.W.; LaFerla, F.M. Alzheimer’s Disease. N. Engl. J. Med. 2010, 362, 329–344. [Google Scholar] [CrossRef] [PubMed]
  7. Christen, Y. Oxidative Stress and Alzheimer Disease. Am. J. Clin. Nutr. 2000, 71, 621s–629s. [Google Scholar] [CrossRef] [PubMed]
  8. Mancuso, M.; Coppede, F.; Migliore, L.; Siciliano, G.; Murri, L. Mitochondrial Dysfunction, Oxidative Stress and Neurodegeneration. J. Alzheimers Dis. 2006, 10, 59–73. [Google Scholar] [CrossRef] [PubMed]
  9. von Bernhardi, R.; Eugenín, J. Alzheimer’s Disease: Redox Dysregulation as a Common Denominator for Diverse Pathogenic Mechanisms. Antioxid. Redox Signal. 2011, 16, 974–1031. [Google Scholar] [CrossRef]
  10. Giasson, B.I.; Ischiropoulos, H.; Lee, V.M.-Y.; Trojanowski, J.Q. The Relationship between Oxidative/Nitrative Stress and Pathological Inclusions in Alzheimer’s and Parkinson’s Diseases. Free. Radic. Biol. Med. 2002, 32, 1264–1275. [Google Scholar] [CrossRef]
  11. McBean, G.J.; López, M.G.; Wallner, F.K. Redox-Based Therapeutics in Neurodegenerative Disease. Br. J. Pharmacol. 2017, 174, 1750–1770. [Google Scholar] [CrossRef] [PubMed]
  12. Guzior, N.; Bajda, M.; Skrok, M.; Kurpiewska, K.; Lewiński, K.; Brus, B.; Pišlar, A.; Kos, J.; Gobec, S.; Malawska, B. Development of Multifunctional, Heterodimeric Isoindoline-1,3-Dione Derivatives as Cholinesterase and β-Amyloid Aggregation Inhibitors with Neuroprotective Properties. Eur. J. Med. Chem. 2015, 92, 738–749. [Google Scholar] [CrossRef] [PubMed]
  13. Albertini, C.; Salerno, A.; de Sena Murteira Pinheiro, P.; Bolognesi, M.L. From Combinations to Multitarget-Directed Ligands: A Continuum in Alzheimer’s Disease Polypharmacology. Med. Res. Rev. 2021, 41, 2606–2633. [Google Scholar] [CrossRef] [PubMed]
  14. Pathak, C.; Kabra, U.D. A Comprehensive Review of Multi-Target Directed Ligands in the Treatment of Alzheimer’s Disease. Bioorganic Chem. 2024, 144, 107152. [Google Scholar] [CrossRef] [PubMed]
  15. Dgachi, Y.; Ismaili, L.; Knez, D.; Benchekroun, M.; Martin, H.; Szałaj, N.; Wehle, S.; Bautista-Aguilera, O.M.; Luzet, V.; Bonnet, A.; et al. Synthesis and Biological Assessment of Racemic Benzochromenopyrimidinimines as Antioxidant, Cholinesterase, and Aβ1–42 Aggregation Inhibitors for Alzheimer’s Disease Therapy. ChemMedChem 2016, 11, 1318–1327. [Google Scholar] [CrossRef] [PubMed]
  16. Dgachi, Y.; Bautista-Aguilera, O.M.; Benchekroun, M.; Martin, H.; Bonet, A.; Knez, D.; Godyń, J.; Malawska, B.; Gobec, S.; Chioua, M.; et al. Synthesis and Biological Evaluation of Benzochromenopyrimidinones as Cholinesterase Inhibitors and Potent Antioxidant, Non-Hepatotoxic Agents for Alzheimer’s Disease. Molecules 2016, 21, 634. [Google Scholar] [CrossRef] [PubMed]
  17. Kumari, S.; Maddeboina, K.; Bachu, R.D.; Boddu, S.H.S.; Trippier, P.C.; Tiwari, A.K. Pivotal Role of Nitrogen Heterocycles in Alzheimer’s Disease Drug Discovery. Drug Discov. Today 2022, 27, 103322. [Google Scholar] [CrossRef]
  18. Martorana, A.; Giacalone, V.; Bonsignore, R.; Pace, A.; Gentile, C.; Pibiri, I.; Buscemi, S.; Lauria, A.; Piccionello, A.P. Heterocyclic Scaffolds for the Treatment of Alzheimer’s Disease. Curr. Pharm. Des. 2016, 22, 3971–3995. [Google Scholar] [CrossRef]
  19. Rastegari, A.; Nadri, H.; Mahdavi, M.; Moradi, A.; Mirfazli, S.S.; Edraki, N.; Moghadam, F.H.; Larijani, B.; Akbarzadeh, T.; Saeedi, M. Design, Synthesis and Anti-Alzheimer’s Activity of Novel 1,2,3-Triazole-Chromenone Carboxamide Derivatives. Bioorganic Chem. 2019, 83, 391–401. [Google Scholar] [CrossRef]
  20. Xu, M.; Peng, Y.; Zhu, L.; Wang, S.; Ji, J.; Rakesh, K.P. Triazole Derivatives as Inhibitors of Alzheimer’s Disease: Current Developments and Structure-Activity Relationships. Eur. J. Med. Chem. 2019, 180, 656–672. [Google Scholar] [CrossRef]
  21. Zribi, L.; Pachòn-Angona, I.; Bautista-Aguilera, Ò.M.; Diez-Iriepa, D.; Marco-Contelles, J.; Ismaili, L.; Iriepa, I.; Chabchoub, F. Triazolopyridopyrimidine: A New Scaffold for Dual-Target Small Molecules for Alzheimer’s Disease Therapy. Molecules 2020, 25, 3190. [Google Scholar] [CrossRef] [PubMed]
  22. Bansal, Y.; Silakari, O. Multifunctional Compounds: Smart Molecules for Multifactorial Diseases. Eur. J. Med. Chem. 2014, 76, 31–42. [Google Scholar] [CrossRef]
  23. Khandelwal, S.; Tailor, Y.K.; Rushell, E.; Kumar, M. 9—Use of Sustainable Organic Transformations in the Construction of Heterocyclic Scaffolds. In Green Approaches in Medicinal Chemistry for Sustainable Drug Design; Banik, B.K., Ed.; Advances in Green and Sustainable Chemistry; Elsevier: Amsterdam, The Netherlands, 2020; pp. 245–352. ISBN 978-0-12-817592-7. [Google Scholar]
  24. González-Muñoz, G.C.; Arce, M.P.; López, B.; Pérez, C.; Romero, A.; Barrio, L.d.; Martín-de-Saavedra, M.D.; Egea, J.; León, R.; Villarroya, M.; et al. N-Acylaminophenothiazines: Neuroprotective Agents Displaying Multifunctional Activities for a Potential Treatment of Alzheimer’s Disease. Eur. J. Med. Chem. 2011, 46, 2224–2235. [Google Scholar] [CrossRef] [PubMed]
  25. Denizot, F.; Lang, R. Rapid Colorimetric Assay for Cell Growth and Survival. Modifications to the Tetrazolium Dye Procedure Giving Improved Sensitivity and Reliability. J. Immunol. Methods 1986, 89, 271–277. [Google Scholar] [CrossRef]
  26. Dávalos, A.; Gómez-Cordovés, C.; Bartolomé, B. Extending Applicability of the Oxygen Radical Absorbance Capacity (ORAC–Fluorescein) Assay. J. Agric. Food Chem. 2004, 52, 48–54. [Google Scholar] [CrossRef]
  27. Ou, B.; Hampsch-Woodill, M.; Prior, R.L. Development and Validation of an Improved Oxygen Radical Absorbance Capacity Assay Using Fluorescein as the Fluorescent Probe. J. Agric. Food Chem. 2001, 49, 4619–4626. [Google Scholar] [CrossRef]
  28. Benchekroun, M.; Bartolini, M.; Egea, J.; Romero, A.; Soriano, E.; Pudlo, M.; Luzet, V.; Andrisano, V.; Jimeno, M.-L.; López, M.G.; et al. Novel Tacrine-Grafted Ugi Adducts as Multipotent Anti-Alzheimer Drugs: A Synthetic Renewal in Tacrine-Ferulic Acid Hybrids. ChemMedChem 2015, 10, 523–539. [Google Scholar] [CrossRef]
  29. Kam, T.-I.; Gwon, Y.; Jung, Y.-K. Amyloid Beta Receptors Responsible for Neurotoxicity and Cellular Defects in Alzheimer’s Disease. Cell. Mol. Life Sci. 2014, 71, 4803–4813. [Google Scholar] [CrossRef]
  30. Levine III, H. Thioflavine T Interaction with Synthetic Alzheimer’s Disease β-Amyloid Peptides: Detection of Amyloid Aggregation in Solution. Protein Sci. 1993, 2, 404–410. [Google Scholar] [CrossRef]
  31. Aronov, A.M. Predictive in Silico Modeling for hERG Channel Blockers. Drug Discov. Today 2005, 10, 149–155. [Google Scholar] [CrossRef]
  32. Aronov, A.M. Ligand Structural Aspects of hERG Channel Blockade. Curr. Top. Med. Chem. 2008, 8, 1113–1127. [Google Scholar] [CrossRef] [PubMed]
Scheme 1. Synthesis of pyrimidopyrimidines 4ak.
Scheme 1. Synthesis of pyrimidopyrimidines 4ak.
Chemistry 06 00041 sch001
Figure 1. Inhibition of Aβ1–42 self-aggregation (%) by selected compounds.
Figure 1. Inhibition of Aβ1–42 self-aggregation (%) by selected compounds.
Chemistry 06 00041 g001
Table 1. Synthesis of diphenylpyrimido [4,5-d] pyrimidin-4-amines derivatives 4ak using 2 steps.
Table 1. Synthesis of diphenylpyrimido [4,5-d] pyrimidin-4-amines derivatives 4ak using 2 steps.
CompoundR1R2R3R4R5R6Yield (%)
4aCH3HHHHH57
4bC2H5HHHHH50
4cCH3C2H5HHHH35
4dCH3HCH3HHH16
4eCH3CH3CH3HHH20
4fCH3CH3HHHH50
4gCH3CH3HHCH3H28
4hCH3C2H5HHCH3H30
4iCH3CH3HHClH22
4jCH3C2H5HOCH3OCH3H20
4kCH3HHOCH3HOCH347
Table 2. Neuroprotective activity on H2O2 -induced cell death in SH-SY5Y cells and ORAC values of compounds 4ak.
Table 2. Neuroprotective activity on H2O2 -induced cell death in SH-SY5Y cells and ORAC values of compounds 4ak.
CompoundNeuroprotection (%) against H2O2 a at 0.1, 1 and 5µMORAC c (TE)
0.1 µM1 µM5 µM
4a- b- b- b0.07 ± 0.00
4b6.17 ± 0.014.95 ± 0.0114.66 ± 0.04- b
4c6.96 ± 0.023.09 ± 0.28.64 ± 0.050.24 ± 0.02
4d9.40 ± 0.01- b- b0.14 ± 0.01
4e- b34.00 ± 0.09 *8.45 ± 0.20.17 ± 0.01
4f4.75 ± 0.016.98 ± 0.0224.30 ± 0.02 *0.19 ± 0.02
4g47.03 ± 0.12 **68.06 ± 0.22 **77.55 ± 0.21 **0.24 ± 0.02
4h66.13 ± 0.17 **38.77 ± 0.07 **26.12 ± 0.16 **- b
4i68.35 ± 0.23 **117.89 ± 0.04 ***94.54 ± 0.33 ***0.34 ± 0.01
4j66.94 ± 0.23 **45.89 ± 0187 **11.14 ± 0.151.64 ± 0.02
4k0.13 ± 0.01- b- b- b
Trolox- d- d- d0.99 ± 0.01
Melatonin- d- d- d2.45 ± 0.09
a Data expressed as % neuroprotection ± SEM of quadruplicates from at least three different cultures; * p  <  0.05, ** p < 0.01 and *** p < 0.001, as compared to the control cultures (one-way ANOVA). b not active, c Data are expressed as Trolox equivalents and are the mean (n = 3) ± SEM. d Not determined.
Table 3. Physicochemical properties of the synthesized compounds calculated by Data Warrior and hERG inhibition predicted by PreAdmet.
Table 3. Physicochemical properties of the synthesized compounds calculated by Data Warrior and hERG inhibition predicted by PreAdmet.
CompoundMolweight (g/mol)CLogPH-AcceptorsH-DonorsTopological Polar Surface Area (Å2)hERG
Inhibition
4a312.3753.87934051.56low_risk
4b326.4024.29494051.56low_risk
4c340.4294.69284051.56medium_risk
4d326.4024.22324051.56medium_risk
4e340.4294.62114051.56medium_risk
4f326.4024.27724051.56low_risk
4g340.4294.62114051.56medium_risk
4h354.4565.03674051.56medium_risk
4i360.8474.88324051.56medium_risk
4j400.4814.55286070.02medium_risk
4k372.4273.73936070.02medium_risk
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Ben Ameur, G.; Maalej, E.; Martin, H.; Jacquinot, A.-S.; Barbanneau, N.; Bernard, P.J.; Marco-Contelles, J.; Chabchoub, F.; Ismaili, L. Efficient Two-Step Synthesis of Novel Pyrimido[4,5-d] Pyrimidines with Potent Neuroprotective, Antioxidant, and Aβ Anti-Aggregation Properties. Chemistry 2024, 6, 695-705. https://doi.org/10.3390/chemistry6040041

AMA Style

Ben Ameur G, Maalej E, Martin H, Jacquinot A-S, Barbanneau N, Bernard PJ, Marco-Contelles J, Chabchoub F, Ismaili L. Efficient Two-Step Synthesis of Novel Pyrimido[4,5-d] Pyrimidines with Potent Neuroprotective, Antioxidant, and Aβ Anti-Aggregation Properties. Chemistry. 2024; 6(4):695-705. https://doi.org/10.3390/chemistry6040041

Chicago/Turabian Style

Ben Ameur, Ghada, Emna Maalej, Helene Martin, Anne-Sophie Jacquinot, Nadine Barbanneau, Paul J. Bernard, José Marco-Contelles, Fakher Chabchoub, and Lhassane Ismaili. 2024. "Efficient Two-Step Synthesis of Novel Pyrimido[4,5-d] Pyrimidines with Potent Neuroprotective, Antioxidant, and Aβ Anti-Aggregation Properties" Chemistry 6, no. 4: 695-705. https://doi.org/10.3390/chemistry6040041

Article Metrics

Back to TopTop