Next Article in Journal
Coumarins as Powerful Photosensitizers for the Cationic Polymerization of Epoxy-Silicones under Near-UV and Visible Light and Applications for 3D Printing Technology
Next Article in Special Issue
Molecular Structure Modulated Trap Distribution and Carrier Migration in Fluorinated Epoxy Resin
Previous Article in Journal
Cooperative Analysis of Structural Dynamics in RNA-Protein Complexes by Single-Molecule Förster Resonance Energy Transfer Spectroscopy
Previous Article in Special Issue
Fluorescent Submicron-Sized Poly(heptafluoro-n-butyl methacrylate) Particles with Long-Term Stability
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Efficient Access to 3,5-Disubstituted 7-(Trifluoromethyl)pyrazolo[1,5-a]pyrimidines Involving SNAr and Suzuki Cross-Coupling Reactions

1
Laboratoire de Physico-Chimie des Matériaux et des Electrolytes pour l’Energie (PCM2E), EA 6299, Avenue Monge, Faculté des Sciences, Université de Tours, Parc de Grandmont, 37200 Tours, France
2
Laboratoire de Chimie Moléculaire et Substances Naturelles, Faculté des Sciences, Université Moulay Ismail, B.P. 11201, Zitoune, Meknès 50050, Morocco
3
Laboratoire de Chimie Physique & de Chimie Bioorganique, URAC 22, Université Hassan II de Casablanca, B.P. 146, Mohammedia 28800, Morocco
4
Institut de Chimie Organique et Analytique (ICOA), Université d’Orléans, UMR CNRS 7311, BP 6759, Rue de Chartres, CEDEX 2, 45067 Orléans, France
*
Author to whom correspondence should be addressed.
Molecules 2020, 25(9), 2062; https://doi.org/10.3390/molecules25092062
Submission received: 30 March 2020 / Revised: 17 April 2020 / Accepted: 23 April 2020 / Published: 28 April 2020
(This article belongs to the Special Issue Fluorine Chemistry)

Abstract

:
An efficient and original synthesis of various 3,5-disubstituted 7-(trifluoromethyl)pyrazolo[1,5-a]pyrimidines is reported. A library of compounds diversely substituted in C-3 and C-5 positions was easily prepared from a common starting material, 3-bromo-7-(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-5-one. In C-5 position, a SNAr type reaction was achieved by first activating the C–O bond of the lactam function with PyBroP (Bromotripyrrolidinophosphonium hexafluorophosphate), followed by the addition of amine or thiol giving monosubstituted derivatives, whereas in C-3 position, arylation was performed via Suzuki–Miyaura cross-coupling using the commercially available aromatic and heteroaromatic boronic acids. Moreover, trifluoromethylated analogues of potent Pim1 kinase inhibitors were designed following our concise synthetic methodology.

Graphical Abstract

1. Introduction

Pyrimidine fused heterocycles constitute an important class of heterocyclic compounds, having various applications in many fields owing to their chemical and biological properties. In particular, pyrazolo[1,5-a]pyrimidines are one of the most interesting pyrimidine derivatives displaying a wide range of pharmacological activities, as antimicrobial [1,2], antitumor [3], anticancer [4,5,6,7,8], anxiolytic [9,10], antidepressant [11,12,13], and antiepileptic agents [14]. Moreover, pyrazolo[1,5-a]pyrimidines are also known as antagonists of serotonin 5-HT6 receptors [15], and also act as inhibitors of histone lysine demethylase 4D (KDM4D) [16] as well as several kinases such as Pim kinase [17,18], threonine tyrosine kinase (TTK) [19], and cyclin-dependent kinases (CDKs) [20]. In addition, some pyrazolo[1,5-a]pyrimidine derivatives were found to have an antiviral effect against the respiratory syncytial virus (RSV) [21] and hepatitis C virus [22], and are inhibitors of phosphodiesterase (PDE) 2A [23] and 10A [24,25,26], promising new targets for the treatment of cognitive disorders and schizophrenia, respectively.
The incorporation of a fluorine atom into potential pharmaceutical substances has been successfully used as a key strategy for enhancing the activity of drugs or drug candidates [27,28]. The introduction of fluorine atoms leads to a change in the solubility and lipophilicity of compounds, thus increasing their bioavailability and their cell membranes permeability. As a consequence, the combination between the pyrazolo[1,5-a]pyrimidine scaffold and the interesting properties of the fluorine atom could give rise to new heterocycles displaying potent biological activities. Synthetically, although many approaches have been reported for the synthesis of non-fluorinated pyrazolo[1,5-a]pyrimidines, the incorporation of fluorinated groups in this heterocycle remains limited [29,30,31,32,33,34,35,36]. Recently, we described the condensation of 5-substituted 3-aminopyrazoles with ethyl 4,4,4-trifluoro-2-butynoate, affording regioselectively the 2-substituted 7-(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-5-ones in fair to good yields [37]. Continuing our research directed towards the development of new heterocyclic compounds containing the fluorine atoms [38,39,40,41], we report herein an efficient approach for synthesizing 3,5-disubstituted 7-(trifluoromethyl)pyrazolo[1,5-a]pyrimidines in good yields through SNAr and Suzuki–Miyaura cross-coupling reactions, using 3-bromo-7-(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-5-one as a starting material.

2. Results and Discussion

Our synthesis starts with the generation of 7-(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-5-one core 3 following our reported procedure [41]. The reaction between 3-aminopyrazole 1 and ethyl 4,4,4-trifluoro-2-butynoate 2 in 1,4-dioxane at 110 °C under microwave irradiation for 2 h, followed by the addition of NaOMe (2 equiv.) and stirring for 12 h at room temperature, gave the only regioisomer 3 in 63% yield (Scheme 1). Selective bromination of 3 with N-bromosuccinimide in dichloromethane afforded the bromo derivative 4 in 94% yield. The 3-bromo-7-(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-5-one 4 served as a building block for the synthesis of 3,5-disubstituted 7-(trifluoromethyl)pyrazolo[1,5-a]pyrimidines having two new points of diversity.
Recently, we reported one pot SNAr reaction strategy to functionalize the pyrazolo[1,5-a]pyrimidine moiety at C-5 position through C-O activation of the lactam function with PyBroP (bromotripyrrolidinophosphonium hexafluorophosphate) as the activating reagent [41]. We extended this approach to the brominated compound 4 in order to substitute the pyrazolo[1,5-a]pyrimidine ring differently at C-3 and C-5 positions. The C–O bond activation was carried out with PyBroP (Bromotripyrrolidinophosphonium hexafluorophosphate) (1.3 equiv.) in the presence of Et3N (3 equiv.) in 1,4-dioxane at room temperature for 2 h. Then, the addition of various amines (1.5 equiv.) in 1,4-dioxane at 110 °C for 12 h led to the desired 5-amino-7-(trifluoromethyl)pyrazolo[1,5-a]pyrimidines 5af in 74–98% yields (Scheme 2). A one-pot reaction using benzylamine and 4-methoxybenzylamine as the nucleophilic reagents provided the corresponding trifluoromethylated 5-aminopyrazolo[1,5-a]pyrimidines 5a and 5b in the same 90% yield. Under the same conditions, aliphatic amines bearing protected or unprotected functions such as NHBoc, free hydroxyl group and triple bond, reacted well and afforded the expected heterocycles 5ce in 74%, 84%, and 96% yields, respectively (Scheme 2). When morpholine was used as a secondary amine, this SNAr reaction was also efficiently performed and the compound 5f was isolated in 98%. In view of the success of amine addition, this reaction was extended to sulfur compounds such as p-methoxythiophenol (PMPSH) giving successfully the desired product 5g in 73% yield (Scheme 2).
In order to prepare a library of new 3,5-disubstituted 7-(trifluoromethyl) pyrazolo[1,5-a]pyrimidines, bromine in C-3 position of 5a–f was subjected to a Suzuki–Miyaura cross-coupling. To optimize the coupling conditions, the reaction between compound 5f and 4-methoxyphenylboronic acid was studied as a model (Table 1). First, this coupling was carried out with p-methoxyphenylboronic acid (2 equiv.) in the presence of PdCl2(PPh3)2 (10 mol%) as the catalyst and sodium carbonate (2 equiv.) as the base in a mixture of dioxane/water (4/1) at 110 °C. Under these conditions, the starting material was completely consumed after 12 h, and the analysis of the crude 1H NMR spectrum (see Supplementary Materials) showed 10% of the coupled product 6a along with 90% of debrominated compound 7 as a byproduct (Table 1, entry 1). As a proper combination between catalyst, base, and solvent is very important for the success of the reaction, the influence of these parameters was then examined. Using PdCl2dppf instead of PdCl2(PPh3)2 improved slightly the 6a/7 ratio from (10:90) to (20:80) (Table 1, entries 1–2). Under the same conditions, replacing Na2CO3 by K2CO3 resulted in a similar 6a/7 ratio (Table 1, entry 3). The replacement of PdCl2dppf with XPhosPdG2/XPhos tandem at 135 °C affected slightly the formation of byproduct 7 since its yield decreased to 70% and the yield of the desired product 6a increased to 30% (Table 1, entry 4). To reduce the reaction time and to improve the yield of 6a at the expense of 7, conventional thermal heating was replaced by microwave irradiation (MW) to promote the reaction. This heating mode seems beneficial for the reaction since the time was reduced to only 40 min and the percentage of 6a in the mixture increased to 45% (Table 1, entry 5). Gratifyingly, when adding ethanol as an organic solvent instead of dioxane, the starting material was fully converted to the desired compound 6a which was obtained in excellent 93% isolated yield, after purification by silica-gel column chromatography (Table 1, entry 6). A significant effect of the base on the formation of the undesired product 7 was observed when Na2CO3 was used instead of K2CO3 (Table 1, entry 7). When the reaction was conducted without ligand, both products 6a and 7 were formed equally, indicating that the Xphos ligand plays a critical role in avoiding the formation of byproduct 7 (Table 1, entry 8). By reducing the amounts of catalyst and ligand to 5 mol%, a decrease in the yield of coupling product 6a was observed (Table 1, entry 9).
On analyzing the synthesis of compound 6a, which involved a C–O bond activation reaction using morpholine as amine at C-5 followed by the Suzuki–Miyaura cross-coupling at C-3 (Scheme 3, Path A), the overall yield was found to be 91% over two steps. This sequence could be reversed by conducting the Suzuki–Miyaura coupling first at C-3 followed by SNAr reaction at C-5 (Scheme 3, Path B). Following the synthetic route B, the overall yield of 6a decreased to 77% and some problems were encountered during the purification of the intermediate 8 because of the presence of unprotected lactam function (Scheme 3, Path B). Therefore, the efficient, convergent, and interesting synthetic pathway A was applied to generate several new 3,5-disubstituted 7-(trifluoromethyl)pyrazolo[1,5-a]pyrimidines. Encouraged by the good yield of 6a obtained in two sequential steps (path A), this synthesis was attempted according to a one-pot strategy. In a sealed vial and reproducing the same conditions as those of the first and second steps (Path A), without isolating intermediate 5f, the desired product 6a was not observed since only 61% of debrominated by-product 7 was isolated. This debromination was likely induced by the presence of triethylamine and morpholine in the reaction medium.
After establishing the best conditions to carry out the Suzuki coupling at C-3 of 5f, we extended this coupling to monosubstituted substrates 5ae using commercially available aromatic and heteroaromatic boronic acids. In all cases, direct arylation at C-3 produced the corresponding 3,5-bifunctionnalized 7-(trifluoromethyl)pyrazolo[1,5-a]pyrimidines 6am in good to excellent isolated yields whatever the nature of the amine at C-5. The results are summarized in Scheme 4.
As illustrated in Scheme 4, unsubstituted aryl such as phenyl, biphenyl, and naphthyl were successfully coupled under optimized conditions, and the desired products 6b, 6d, 6f, 6k, and 6m were isolated in yields ranging from 74% and 91%. A phenylboronic acid bearing a methoxy group, as an electron-donating group, at the para position was easily coupled with 5f, giving the desired compound 6a in 93% yield. In addition, a free alcohol moiety was tolerated at the para position of phenylboronic acid providing compound 6g in 82% yield. A phenylboronic acid with CF3, as an electron-withdrawing group, was also tolerated, yielding the expected product 6l in 77% yield (Scheme 4).
After the good results obtained with different aryls, this coupling was extended to heteroaryl boronic acids. Interestingly, using 2-thienyl as an heteroarylboronic acid in coupling reaction with 7-(trifluoromethyl)pyrazolo[1,5-a]pyrimidines containing morpholino or benzylamino group at C-5 position, worked well and the desired heterocycles were isolated in good yields (6c (79%) and 6e (76%)). Similarly, the cross-coupling between 3-pyridinylboronic acid or 4-dibenzothienylboronic acid and 5a, provided the coupled products in excellent yields (6i (91%) and 6j (92%)). However, the isolated yield obtained with the coupling of 1H-pyrazole-4-boronic acid and 5a did not exceed 50% which could be explained by the presence of a free NH-pyrazole bond (Scheme 4).
To demonstrate the synthetic potential of our method, we turned our attention to the synthesis of biologically active compounds. With the introduction of a suitable aryl at C-3 and amine group at C-5 of the pyrazolo[1,5-a]pyrimidine core, we were able to design the fluorinated pyrazolo[1,5-a]pyrimidines 6k and 6l, analogues of compounds 10 and 11, which have a nanomolar activities against Pim1 kinase (10, IC50 = 18 nM) and (11, IC50 = 27 nM), respectively (Scheme 5) [17,42]. In order to prepare the trifluoromethylated analogue of the potent Pim1 kinase inhibitor 12 (IC50 = 1.5 nM), the Boc protected amine 6m was treated with an excess of trifluoroacetic acid at room temperature, yielding the desired diamine 9 in 95% (Scheme 5). It is noteworthy that there is a correlation between prostate cancer disease and Pim-1 since the DNA microarray analyses showed that this kinase was overexpressed in human prostate cancer [43].

3. Materials and Methods

3.1. General Methods

All reagents were purchased from commercial suppliers and were used without further purification. Triethylamine and DCM were distilled over calcium hydride. 1,4-Dioxane was distilled over sodium and benzophenone. Reactions were monitored by thin-layer chromatography (TLC) analysis using silica gel (60 F254) plates. The compounds were visualized by longwave (365 nm) or shortwave (254 nm) UV light. Column chromatography was performed using silica gel 60 (230–400 mesh, 0.040–0.063 mm) to purify the product.
All 1H NMR, 13C NMR, and 19F NMR spectra were recorded with a 300 MHz Bruker Avance FT-NMR spectrometer (300 MHz, 75 MHz, or 282 MHz, respectively, (Billerica, MA, USA).
All chemical shifts are given in ppm and they are referenced to tetramethylsilane (TMS) as an internal standard. 1H NMR assignment abbreviations are indicated as follows: singlet (s), doublet (d), triplet (t), quartet (q), broad singlet (br s), doublet of doublets (dd), triplet of doublets (td), doublet of a triplets (dt), and multiplet (m). Coupling constants (J) are reported in Hertz (Hz). Electrospray ionization high-resolution mass spectrometry experiments were performed with a hybrid tandem quadrupole/time-of flight (Q-TOF) instrument, equipped with a pneumatically assisted electrospray (Zspray) ion source (Micromass, Manchester, UK) operated in positive mode. Melting points were measured using banc Kofler.

Microwave Assisted Reactions

The microwave-assisted reactions were performed using a Monowave 300 (microwave synthesis reactor: Anton Paar, 300 W maximum power, Graz, Austria). Microwave irradiation was carried out in sealed 10–30 mL vessels (borosilicate glass) with a PTFE (polytetrafluoroethylene)-coated silicon septum and closed with a snap cap made of PEEK (polyetheretherketone). The temperatures were measured on the surface of the vial with an IR sensor (measuring range: 30 to 300 °C; uncertainty: ±5 °C) and were measured with high precision in the reaction mixture with a ruby thermometer (measuring range: 30 to 300 °C; uncertainty: ±2 °C) that could be read directly from the instrument screen. The reaction time was measured from when the reaction mixture reached the stated temperature for temperature controlled experiments. The pressure was measured with a non-invasive pressure sensor located in the swiveling cover of the Monowave 300 (measuring range: 0 to 30 bar; uncertainty: ±1.5 bar).

3.2. Synthesis of 3-Bromo-7-(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-5-one 4: Bromination of 3-bromo-7-(trifluoromethyl)pyrazolo[1,5-a]pyrimdin-5-one 3

NBS (1.05 equiv.) was added to a solution of pyrazolo[1,5-a]pyrimdin-5-one 3 (1 equiv.) in 8 mL of CH2Cl2 and the reaction was stirred under an argon atmosphere at room temperature for 12 h. The progress of the reaction was monitored by TLC (PE/EtOAc, 7/3). After completion of the reaction, evaporation of the solvent under reduced pressure provided the crude product, which was purified by column chromatography (PE/EtOAc, 7.5/2.5) to afford the final product 4 as a white solid in 94% yield; melting point: 209–211 °C. 1H NMR (300 MHz, DMSO-d6): δ 13.00 (s, 1H), 8.07 (s, 1H), 6.74 (s, 1H). 13C NMR (75 MHz, DMSO-d6): δ 159.7, 144.8, 140.5, 135.7 (q, J = 36.8 Hz), 119.2 (q, J = 274.6 Hz), 108.0, 75.1. 19F NMR (282 MHz, DMSO-d6): δ −66.65. HRMS (ESI) m/z [M + H]+ calcd for C7H4BrF3N3O: 281.9484; found: 281.9474.

3.3. General Procedure for the Synthesis of 5-Amino (or Mercapto) 3-bromo-7-(trifluoromethyl)pyrazolo[1,5-a]pyrimidines 5a–g by the Direct Amination or Thiolation via C-OH Bond Activation with PyBroP

In a sealed tube, PyBroP (1.44 mmol, 621 mg, 1.3 equiv.) and Et3N (3.33 mmol, 0.46 mL, 3 equiv.) were added successively to a solution of 4 (1.11 mmol, 150 mg, 1 equiv.) in 1.4-dioxane (4 mL). The mixture was degassed by argon bubbling and then stirred at room temperature for 2 h. The corresponding amine or thiol (1.5 equiv.) was then added, and the mixture was stirred for 12 h at 110 °C. After cooling, the solvents were evaporated under reduced pressure, and the crude residue was purified by silica gel column chromatography to give desired 5-aminated (thiolated) 3-bromo-7-(trifluoromethyl)pyrazolo[1,5-a]pyrimdines 5ag.
3-Bromo-5-[N-(4-methoxybenzyl)amino]-7-(trifluoromethyl)pyrazolo[1,5-a]pyrimidine (5a). The purification of the crude product by chromatography on silica gel is carried out using (PE/EtOAc: 9/1) to afford 5a as a yellow solid in 90% yield. m.p. 149–151 °C; 1H NMR (300 MHz, CDCl3): δ 7.97 (s, 1H), 7.35 (d, J = 8.7 Hz, 2H), 6.92 (d, J = 8.7 Hz, 2H), 6.44 (s, 1H), 5.36 (br s, 1H), 4.66 (q, J = 5.3 Hz, 2H), 3.83 (s, 3H); 13C NMR (75 MHz, Acétone-d6): δ 159.2, 154.9, 146.5, 144.4, 133.9 (q, J = 36.5 Hz), 130.3, 129.5 (2C), 119.4 (q, J = 273.3 Hz), 113.8 (2C), 100.9, 79.3, 54.6, 44.3; 19F NMR (282 MHz, CDCl3): δ −68.47; HRMS (ESI) m/z [M + H]+ calcd for C15H13BrF3N4O: 401.0219; found: 104.0224.
5-(N-Benzylamino)-3-bromo-7-(trifluoromethyl)pyrazolo[1,5-a]pyrimidine (5b). The purification of the crude product by chromatography on silica gel is carried out using (PE/EtOAc: 9/1) to afford 5b as a yellow solid in 90% yield. m.p. 147–149 °C; 1H NMR (300 MHz, CDCl3): δ 7.97 (s, 1H), 7.42–7.35 (m, 5H), 6.46 (s, 1H), 5.43 (br s, 1H), 4.75 (d, J = 5.3 Hz, 2 H); 13C NMR (75 MHz, Acetone-d6): δ 155.0, 146.5, 144.4, 138.5, 133.9 (q, J = 36.8 Hz), 128.4, 128.1, 127.3, 119.4 (q, J = 273.3 Hz), 100.8, 79.3, 44.7. 19F NMR (282 MHz, CDCl3): δ −68.46; HRMS (ESI) m/z [M + H]+ calcd for C14H11BrF3N4: 371.0114; found: 371.0119.
3-Bromo-5-[2-(tert-butoxycarboxylamino)-N-ethylamino]-7-(trifluoromethyl)pyrazolo[1,5-a]pyrimidine (5c). The purification of the crude product by chromatography on silica gel is carried out using (PE/EtOAc: 8/2) to afford 5c as a yellow solid in 74% yield. m.p. 155–157 °C; 1H NMR (300 MHz, CDCl3): δ 7.93 (s, 1H), 6.48 (s, 1H), 6.43 (br s, 1H), 5.12 (br s, 1H), 3.73–3.63 (m, 2H), 3.51–3.41 (m, 2H), 1.45 (s, 9H); 13C NMR (75 MHz, Acetone-d6): δ 156.2, 165.4, 146.5, 144.4, 133.8 (q, J = 37.4 Hz), 119.4 (q, J = 273.3 Hz), 101.0, 79.2, 78.0, 41.5, 39.4, 27.7 (3C); 19F NMR (282 MHz, CDCl3): δ −68.41; HRMS (ESI) m/z [M + H]+ calcd for C14H18BrF3N5O2: 424.0590; found: 424.0593.
3-Bromo-5-N-[4-(hydroxycyclohexyl)amino]-7-(trifluoromethyl)pyrazolo[1,5-a]pyrimidine (5d). The purification of the crude product by chromatography on silica gel is carried out using (PE/EtOAc: 6/4) to afford 5d as a yellow solid in 84% yield. m.p. 195–197 °C; 1H NMR (300 MHz, Acetone-d6): δ 7.93 (s, 1H), 7.21 (d, J = 6.32 Hz, 1H), 4.07–4.03 (m, 1H), 3.82 (d, J = 4.5 Hz, 1H), 3.63–3.57 (m, 1H), 2.96–2.93 (m, 2H), 2.10–2.05 (m, 2H), 1.46–1.37 (m, 4H); 13C NMR (75 MHz, Acetone-d6): δ 154.5, 146.6, 144.4, 133.7 (q, J = 37.1 Hz), 119.4 (q, J = 273.3 Hz), 101.0, 79.0, 68.8, 49.3, 33.8 (2C), 29.8 (2C); 19F NMR (282 MHz, Acetone-d6): δ −68.97; HRMS (ESI) m/z [M + H]+ calcd for C13H15BrF3N4O: 379.0376; found: 379.0378.
3-Bromo-N-[(prop-2-ynyl)amino]-7-(trifluoromethyl)pyrazolo[1,5-a]pyrimidine (5e). The purification of the crude product by chromatography on silica gel is carried out using (PE/EtOAc: 8.5/1.5) to afford 5e as a yellow solid in 96% yield. m.p. 149–151 °C; 1H NMR (300 MHz, CDCl3): δ 7.99 (s, 1H), 6.51 (s, 1H), 5.33 (br s, 1H), 4.39 (dd, J = 5.2, 2.5 Hz, 2H), 2.34 (t, J = 2.5 Hz, 1H); 13C NMR (75 MHz, Acetone-d6): δ 154.5, 146.2, 144.6, 134.1 (q, J = 36.9 Hz), 119.4 (q, J = 273.4 Hz), 100.5, 79.7, 79.6, 72.1, 30.2; 19F NMR (282 MHz, CDCl3): δ −68.49; HRMS (ESI) m/z [M + H]+ calcd for C10H7BrF3N4: 318.9801; found: 318.9802.
3-Bromo-5-morpholino-7-(trifluoromethyl)pyrazolo[1,5-a]pyrimidine (5f). The purification of the crude product by chromatography on silica gel is carried out using (PE/EtOAc: 8/2) to afford 5f as a yellow solid in 98% yield. m.p. 182–184 °C; 1H NMR (300 MHz, CDCl3): δ 7.99 (s, 1 H), 6.74 (s, 1H), 3.86–3.79 (m, 8H); 13C NMR (75 MHz, Acetone-d6): δ 155.0, 146.0, 144.9, 134.5 (q, J = 36.7 Hz), 119.6 (q, J = 273.8 Hz), 97.4 (q, J = 4.6 Hz), 79.1, 66.1 (2C), 45.2 (2C); 19F NMR (282 MHz, CDCl3): δ −68.43; HRMS (ESI) m/z [M + H]+ calcd for C11H11BrF3N4O: 351.0063; found: 351.0063.
3-Bromo-5-((4-methoxyphenyl)thio)-7-(trifluoromethyl)pyrazolo[1,5-a]pyrimidine (5g). The purification of the crude product by chromatography on silica gel is carried out using (PE/EtOAc: 9.5/0.5) to afford 5g as a yellow solid in 73% yield. m.p. 135–137 °C; 1H NMR (300 MHz, CDCl3): δ 8.12 (s, 1H), 7.58 (d, J = 8.8 Hz, 2H), 7.05 (d, J = 8.8 Hz, 2H), 6.73 (s, 1H), 3.91 (s, 3H); 13C NMR (75 MHz, Acetone-d6): δ 163.9, 161.7, 145.8, 145.5, 137.2 (2C), 133.4 (q, J = 37.5 Hz), 119.3 (q, J = 274.1 Hz), 117.7, 115.5 (2C), 105.6 (q, J = 4.4 Hz), 83.6, 55.1; 19F NMR (282 MHz, CDCl3): δ −68.59; HRMS (ESI) m/z [M + H]+ calcd for C14H10BrF3N3OS: 403.9675; found: 403.9678.

3.4. General Procedure for C-3 Suzuki–Miyaura Cross-Coupling: Synthesis of 3,5-Disubstituted 7-(trifluoromethyl)pyrazolo[1,5-a]pyrimdines 6am

A mixture of 5-aminated 3-bromo-7-(trifluoromethyl)pyrazolo[1,5-a]pyrimdines 5ag (1 equiv.), boronic acid (2 equiv.), and K2CO3 (2 equiv.) in EtOH (4 mL) and water (1 mL) was thoroughly degassed with a stream of argon. Then, XPhos (10 mol%) and XPhosPdG2 (10 mol%) were added and the microwave vial containing the mixture was capped and inserted into microwave reactor. The reaction mixture was irradiated at 135 °C for 40 min. After that, the mixture was filtered through a Celite, washed with EtOAc, and concentrated under reduced pressure. Purification of the crude via column chromatography afforded desired 3,5-disubstituted 7-(trifluoromethyl)pyrazolo[1,5-a]pyrimidines 6am, which was characterized by 1H, 13C NMR, 19F NMR and HRMS.
3-(4-Methoxyphenyl)-5-morpholine-7-(trifluoromethyl)pyrazolo[1,5-a]pyrimidine (6a). The purification of the crude product by chromatography on silica gel is carried out using (PE/EtOAc: 8/2) to afford 6a as a yellow solid in 93% yield. m.p. 243–245 °C; 1H NMR (300 MHz, CDCl3): δ 8.31 (s, 1H), 7.90 (d, J = 8.8 Hz, 2H), 7.00 (d, J = 8.8 Hz, 2H), 6.74 (s, 1H), 3.90–3.88 (m, 4H), 3.87 (s, 3H), 3.79–3.76 (m, 4H); 13C NMR (75 MHz, CDCl3): δ 157.85, 154.1, 143.7, 135.2 (q, J = 36.1 Hz), 126.9 (2C), 124.9, 120.5, 119.5 (q, J = 274.5 Hz), 114.2 (2C), 107.0, 95.0 (q, J = 4.5 Hz), 66.45 (2C), 55.3, 45.3 (2C); 19F NMR (282 MHz, CDCl3): δ −68.74. HRMS (ESI) m/z [M + H]+ calcd for C18H18F3N4O2: 379.1376; found: 379.1374.
5-Morpholino-3-phenyl-7-(trifluoromethyl)pyrazolo[1,5-a]pyrimidine (6b). The purification of the crude product by chromatography on silica gel is carried out using (PE/EtOAc: 8/2) to afford 6b as a yellow solid in 80% yield. m.p. 230–232 °C; 1H NMR (300 MHz, CDCl3): δ 8.37 (s, 1H), 7.98 (dd, J = 8.4, 1.2 Hz, 2H), 7.45 (t, J = 7.7 Hz, 2H), 7.25 (tt, J = 7.7, 1.2 Hz, 1H), 6.76 (s, 1H), 3.91–3.87 (m, 4H), 3.81–378 (m, 4H); 13C NMR (75 MHz, CDCl3): δ 154.3, 145.35, 144.0, 135.3 (q, J = 36.6 Hz), 132.3, 128.7 (2C), 125.8, 125.6 (2C), 119.5 (q, J = 274.6 Hz), 107.0, 95.1 (q, J = 4.5 Hz), 66.4 (2 C), 45.25 (2C); 19F NMR (282 MHz, CDCl3): δ −68.67; HRMS (ESI) m/z [M + H]+ calcd for C17H16F3N4O: 349.1271; found: 349.12680.
5-Morpholino-3-(thien-2-yl)-7-(trifluoromethyl)pyrazolo[1,5-a]pyrimidine (6c). The purification of the crude product by chromatography on silica gel is carried out using (PE/EtOAc: 8/2) to afford 6c as a yellow solid in 79% yield. m.p. 233–235 °C; 1H NMR (300 MHz, CDCl3): δ 8.27 (s, 1H), 7.42 (dd, J = 3.5, 1.1 Hz, 1H), 7.23 (dd, J = 5.1, 1.1 Hz, 1 H), 7.10 (dd, J = 5.1, 3.5 Hz, 1H), 6.74 (s, 1H), 3.91–3.87 (m, 4H), 3.83–3.80 (m, 4H). 13C NMR (75 MHz, CDCl3): δ 154.3, 144.6, 143.2, 135.3 (q, J = 36.9 Hz), 134.0, 127.1, 122.6, 121.6, 119.4 (q, J = 274.7 Hz), 103.1, 35.2 (q, J = 4.6 Hz), 66.4 (2C), 45.3 (2C). 19F NMR (282 MHz, CDCl3): δ −68.60. HRMS (ESI) m/z [M + H]+ calcd for C15H14F3N4OS: 355.0835; found: 355.0832.
5-(N-Benzylamino)-3-phenyl-7-(trifluoromethyl)pyrazolo[1,5-a]pyrimidine (6d). The purification of the crude product by chromatography on silica gel is carried out using (PE/EtOAc: 9.5/0.5) to afford 6d as a yellow solid in 80% yield. m.p. 173–175 °C; 1H NMR (300 MHz, CDCl3): δ 8.34 (s, 1H), 7.98 (d, J = 7.4 Hz, 2H), 7.46–7.25 (m, 7 H), 7.24 (t, J = 7.4 Hz, 1H), 6.49 (s, 1H), 5.43 (s, 1H), 4.78 (d, J = 5.4 Hz, 2 H); 13C NMR (75 MHz, CDCl3): δ 153.7, 145.4, 143.6, 137.7, 135.0 (q, J = 37.1 Hz), 132.2, 128.8 (2C), 128.6 (2C), 127.9 (2C), 127.9 (2C), 125.8 (2C), 119.3 (q, J = 274.4 Hz), 107.2, 98.5, 45.9; 19F NMR (282 MHz, CDCl3): δ −68.69; HRMS (ESI) m/z [M + H]+ calcd for C20H16F3N4: 369.1322; found: 369.1319.
5-(N-Benzylamino)-3-(thien-2-yl)-7-(trifluoromethyl)pyrazolo[1,5-a]pyrimidine (6e). The purification of the crude product by chromatography on silica gel is carried out using (PE/EtOAc: 9.7/0.3) to afford 6e as a yellow solid in 76% yield. m.p. 156–158 °C; 1H NMR (300 MHz, CDCl3): δ 8.24 (s, 1H), 7.47–7.34 (m, 6H), 7.23 (d, J = 4.6 Hz, 1H), 7.1 (dd, J = 5.3, 4.0 Hz, 1H), 6.45 (s, 1H), 5.43 (br s, 1H), 4.8 (d, J = 5.4 Hz, 2H); 13C NMR (75 MHz, CDCl3): δ 153.8, 144.6, 142.9, 137.6, 135.0 (q, J = 37.0 Hz), 133.9, 128.9 (2C), 128.1 (2C), 127.9, 127.2, 122.7, 122.0, 119.2 (q, J = 274.5 Hz), 103.2, 98.7, 45.8; 19F NMR (282 MHz, CDCl3): δ −68.62; HRMS (ESI) m/z [M + H]+ calcd for C18H14F3N4S: 375.0886; found: 375.0883.
3-([1,1’-Biphenyl]-3-yl)-5-[N-(4-methoxybenzylamino)]-7-(trifluoromethyl)pyrazolo[1,5-a]pyrimidine (6f). The purification of the crude product by chromatography on silica gel is carried out using (PE/EtOAc: 9/1) to afford 6f as a yellow solid in 90% yield. m.p. 124–126 °C; 1H NMR (300 MHz, CDCl3): δ 8.39 (s, 1 H), 8.36 (s, 1H), 7.99–7.96 (m, 1H), 7.69–7.67 (m, 2H), 7.50 (d, J = 7.6 Hz, 2H), 7.47–7.45 (m, 1H), 7.42 (d, J = 7.6 Hz, 2H), 7.35 (d, J = 8.6 Hz, 2H), 6.90 (d, J = 8.6 Hz, 2H), 6.48 (s, 1H), 5.40 (br s, 1H), 4.74 (d, J = 5.2 Hz, 2H), 3.83 (s, 3H); 13C NMR (75 MHz, CDCl3): δ 159.3, 153.7, 145.9, 143.6, 141.5, 141.5, 135.0 (q, J = 37.1 Hz), 132.8, 129.7, 129.3 (2C), 129.0, 128.7 (2C), 127.2, 127.2 (2C), 124.7, 124.5 (2C), 119.3 (q, J = 274.4 Hz), 114.3 (2C), 107.0, 98.6, 55.3, 45.3; 19F NMR (282 MHz, CDCl3): δ −68.64; HRMS (ESI) m/z [M + H]+ calcd for C27H22F3N4O: 475.1740; found: 475.1739
3-[(4-Hydroxyméthyl)phenyl]-5-((4-methoxybenzyl)amino)-7-(trifluoromethyl)pyrazolo[1,5-a]pyrimidine (6g). The purification of the crude product by chromatography on silica gel is carried out using (PE/EtOAc: 7.5/2.5) to afford 6g as a yellow solid in 82% yield. m.p. 184–186 °C; 1H NMR (300 MHz, Acetone-d6): δ 8.40 (s, 1H), 8.07 (d, J = 8.3 Hz, 2H), 7.61 (br s, 1H), 7.44 (d, J = 8.7 Hz, 2H), 7.40 (d, J = 8.3 Hz, 2H), 6.93 (s, 1H), 6.93 (d, J = 8.7 Hz, 2H), 4.73 (d, J = 5.6 Hz, 2H), 4.65 (d, J = 5.5 Hz, 2H), 4.17 (t, J = 5.7 Hz, 1H), 3.78 (s, 3H); 13C NMR (75 MHz, Acetone-d6): δ 159.1, 154.3, 145.8, 142.6, 139.4, 133.8 (q, J = 36.8 Hz), 131.5, 130.8, 129.1 (2C), 126.9 (2C), 125.2 (2C), 119.7 (q, J = 273.3 Hz), 113.8 (2C), 106.1, 99.8, 63.8, 54.6, 44.5; 19F NMR (282 MHz, Acetone-d6): δ −69.15; HRMS (ESI) m/z [M + H]+ calcd for C22H20F3N4O2: 429.1533; found: 429.1531.
5-[N-(4-Methoxybenzyl)amino]-3-(1H-pyrazol-4-yl)-7-(trifluoromethyl)pyrazolo[1,5-a]pyrimidine (6h). The purification of the crude product by chromatography on silica gel is carried out using (PE/EtOAc: 8/2) to afford 6h as a yellow solid in 50% yield. m.p. 197–199 °C; 1H NMR (300 MHz, Acetone-d6): δ 12.08 (s, 1H), 8.24 (s, 1 H), 8.14 (s, 2H), 7.43 (d, J = 8.7 Hz, 2H), 6.92 (d, J = 8.7 Hz, 2H), 6.88 (s, 1H), 4.74 (d, J = 5.5 Hz, 2H), 3.78 (s, 3H). 13C NMR (75 MHz, Acetone-d6): δ 159.1, 153.95, 145.0, 142.05, 135.55, 133.7 (q, J = 36.8 Hz), 130.8, 129.1 (2C), 122.0, 119.7 (q, J = 273.2 Hz), 113.8 (2C), 112.5, 110.1, 99.5, 54.6, 44.4. 19F NMR (282 MHz, Acetone-d6): δ −69.22. HRMS (ESI) m/z [M + H]+ calcd for C18H16F3N6O: 389.1332; found: 389.1331.
5-[N-(4-Methoxybenzyl)amino]-3-(pyridin-3-yl)-7-(trifluoromethyl)pyrazolo[1,5-a]pyrimidine (6i). The purification of the crude product by chromatography on silica gel is carried out using (PE/EtOAc: 8/2) to afford 6i as a yellow solid in 91% yield. m.p. 192–194 °C; 1H NMR (300 MHz, CDCl3): δ 9.29 (s, 1H), 8.45 (s, 1H), 8.33 (s, 1H), 8.29 (d, J = 8.0 Hz, 1H), 7.39–7.37 (m, 1H), 7.33 (d, J = 8.5 Hz, 2H), 6.90 (d, J = 8.5 Hz, 2H), 6.54 (s, 1H), 5.84 (br s, 1H), 4.68 (d, J = 5.2 Hz, 2H), 3.80 (s, 3H). 13C NMR (75 MHz, CDCl3): δ 158.9, 153.7, 146.4, 145.9, 142.5, 134.5 (q, J = 37.1 Hz), 132.2, 129.1, 128.9 (2C), 128.4, 123.2, 118.8 (q, J = 274.3 Hz), 113.8 (3C), 103.3, 98.8, 54.9, 45.0. 19F NMR (282 MHz, CDCl3): δ −68.58. HRMS (ESI) m/z [M + H]+ calcd for C20H17F3N5O: 400.1380; found: 400.1376.
3-(Dibenzo[b,d]thiophen-4-yl)-5-[N-(4-methoxybenzyl)amino]-7-(trifluoromethyl)pyrazolo[1,5-a]pyrimidine (6j). The purification of the crude product by chromatography on silica gel is carried out using (PE/EtOAc: 9/1) to afford 6j as a yellow solid in 92% yield. m.p. 198–200 °C; 1H NMR (300 MHz, Acetone-d6): δ 8.59 (s, 1H), 8.38–8.35 (m, 1H), 8.25 (dd, J = 7.7, 1.1 Hz, 1H), 8.23 (dd, J = 7.7, 1.1 Hz, 1H), 8.03–8.00 (m, 1H), 7.67 (t, J = 4.8 Hz, 1H), 7.63 (t, J = 7.7 Hz, 1H), 7.54 (dd, J = 5.7, 3.4 Hz, 2H), 7.41 (d, J = 8.7 Hz, 2H), 7.03 (s, 1H), 6.92 (d, J = 8.7 Hz, 2H), 4.69 (d, J = 5.7 Hz, 2H), 3.77 (s, 3H). 13C NMR (75 MHz, Acetone-d6): δ 159.1, 154.6, 146.5, 143.3, 139.0, 136.9, 136.15, 135.85, 133.9 (q, J = 36.8 Hz), 130.6, 129.2 (2C), 127.9, 126.9, 126.7, 124.9, 124.6, 122.6, 121.75, 119.7 (q, J = 273.2 Hz), 119.25, 113.75 (2C), 105.3, 100.5, 54.6, 44.4. 19F NMR (282 MHz, Acetone-d6): δ −69.04. HRMS (ESI) m/z [M + H]+ calcd for C27H20F3N4OS: 505.1304; found: 505.1303.
5-N-[4-(Hydroxycyclohexyl)amino]-3-(naphthalen-2-yl)-7-(trifluoromethyl)pyrazolo[1,5-a]pyrimidine (6k). The purification of the crude product by chromatography on silica gel is carried out using (PE/EtOAc: 9/1) to afford 6k as a yellow solid in 91% yield. m.p. 251–253 °C; 1H NMR (300 MHz, Acetone-d6): δ 8.78 (s, 1H), 8.56 (s, 1H), 8.28 (dd, J = 8.6, 1.7 Hz, 1H), 7.94 (d, J = 8.6 Hz, 1H), 7.89 (d, J = 8.5 Hz, 2H), 7.53–7.41 (m, 2H), 7.22 (d, J = 6.7, 1H), 6.86 (s, 1H), 4.16–4.05 (m, 1H), 3.86 (d, J = 4.4 Hz, 1H), 3.73–3.65 (m, 1H), 2.38–2.34 (m, 2H), 2.08–2.06 (m, 2H), 1.66–1.42 (m, 4H); 13C NMR (75 MHz, DMSO-d6): δ 154.1, 146.3, 142.8, 134.1, 133.8 (q, J = 36.6 Hz), 131.8, 130.7, 127.8, 127.6, 127.60, 126.05, 124.9, 124.2, 123.0, 119.7 (q, J = 273.3 Hz), 105.6, 100.0, 69.0, 50.3, 34.1 (2C), 29.7 (2C); 19F NMR (282 MHz, Acetone-d6): δ −62.10; HRMS (ESI) m/z [M + H]+ calcd for C23H22F3N4O: 427.1740; found: 427.1739.
N-[4-(Hydroxycyclohexyl)amino]-3-(3-(trifluoromethyl)phenyl)-7-(trifluoromethyl)pyrazolo[1,5-a]pyrimidine (6l). The purification of the crude product by chromatography on silica gel is carried out using (PE/EtOAc: 9.2/0.8) to afford 6l as a yellow solid in 77% yield. m.p. 257–259 °C; 1H NMR (300 MHz, Acetone-d6): δ 8.77 (s, 1H), 8.54 (s, 1H), 8.26 (d, J = 7.8 Hz, 1H), 7.63 (t, J = 7.8 Hz, 1H), 7.51 (d, J = 7.8 Hz, 1H), 7.27 (d, J = 6.9 Hz, 1H), 6.86 (s, 1H), 4.09–4.05 (m, 1H), 3.82 (d, J = 6.9 Hz, 1H), 3.67–3.60 (m, 1H), 2.29–2.26 (m, 2H), 2.08–2.05 (m, 2H), 1.54–1.42 (m, 4H). 13C NMR (75 MHz, Acetone-d6): δ 154. 4, 146.4, 142.5, 134.2, 133.8 (q, J = 36.5 Hz), 130.3 (q, J = 31.5 Hz), 129.2, 128.0, 124.8 (q, J = 272.0 Hz), 121.6 (q, J = 3.9 Hz), 121.2 (q, J = 3.7 Hz), 119.6 (q, J = 273.2 Hz), 104.1, 100.3, 68.9, 50.0, 33.9 (2C), 29.8 (2C). 19F NMR (282 MHz, Acetone-d6): δ −63.01, −69.09. HRMS (ESI) m/z [M + H]+ calcd for C20H19F6N4O: 445.1458; found: 445.1456.
3-(Naphthalen-2-yl)-5-[2-(tert-butoxycarboxylamino)-N-ethylamino]-7-(trifluoromethyl)pyrazolo[1,5-a]pyrimidine (6m). The purification of the crude product by chromatography on silica gel is carried out using (PE/EtOAc: 8.5/1.5) to afford 6m as a yellow solid in 74% yield. m.p. 183–185 °C; 1H NMR (300 MHz, Acetone-d6): δ 8.73 (s, 1 H), 8.57 (s, 1H), 8.33 (dd, J = 8.6, 1.6 Hz, 1H), 7.96–7.85 (m, 3H), 7.46–7.43 (m, 3H), 6.91 (s, 1H), 6.32 (br s, 1H), 3.82 (q, J = 5.9 Hz, 2H), 3.55 (q, J = 6.0 Hz, 2H), 1.40 (s, 9H); 13C NMR (75 MHz, Acetone-d6): δ 156.2, 155.0, 146.1, 142.9, 134.1, 131.8, 130.5, 127.9 (2C), 127.5, 125.9, 124.9, 124.4, 123.1, 119.7, 119.7 (q, J = 273.3 Hz), 105.9, 100.0, 78.0, 41.6, 39.4, 27.7 (3C); 19F NMR (282 MHz, Acetone-d6): δ −69.06; HRMS (ESI) m/z [M + H]+ calcd for C24H25F3N5O2: 472.1955; found: 472.1953.

3.5. Deprotection of N-Boc Protected Amine by TFA: Synthesis of 5-[2-Amino-N-ethylamino]-3-(naphthalen-2-yl)-7-(trifluoromethyl)pyrazolo[1,5-a]pyrimidine (9)

A solution of the N-Boc protected amine 6m in 5 mL of trifluoroacetic acid was stirred under argon atmosphere at room temperature for 6 h. After the trifluoacetic acid was removed under vacuum, the residue was mixed with water (1 mL), and the mixture was treated with an ammonia solution (10%) to pH = 8. The formed solid was filtered and washed with water and Et2O to give product 9 as a yellow solid in 95% yield. m.p. 171–173 °C; 1H NMR (300 MHz, Acetone-d6): δ 8.57 (s, 1H), 8.45 (s, 1H), 8.19 (dd, J = 8.6, 1.7 Hz, 1H), 7.83 (d, J = 8.5 Hz, 1H), 7.82 (d, J = 8.0 Hz, 1H), 7.77 (d, J = 8.0 Hz, 1H), 7.38 (td, J = 7.5, 1.3 Hz, 1H), 7.33 (td, J = 7.5, 1.3 Hz, 1H), 6.83 (s, 1H), 4.18–4.20 (m, 2H), 4.05–4.03 (m, 2H). HRMS (ESI) m/z [M + H]+ calcd for C24H25F3N5O2: 372.1455; found: 372.1453.

4. Conclusions

In conclusion, an efficient strategy to synthesize 3,5-disubstituted pyrazolo[1,5-a]pyrimidines bearing CF3 group at C-7 was developed, starting from the building block, 3-bromo-7-(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-5-one. A sequential SNAr with various amines and Suzuki cross-coupling were achieved at C-5 and C-3, respectively. The activation of the C-O bond of the amide function followed by the addition of several amines or p-methoxythiophenol allowed the functionalization of pyrazolo[1,5-a]pyrimidine core at C-5. The arylation at C-3 was carried out in good to excellent yields under Suzuki cross-coupling, requiring XPhosPdG2/XPhos as a catalytic system and potassium carbonate as a base in a mixture of EtOH/H2O (4/1) at 135 °C under microwave irradiation. Further studies are underway in our laboratory following this strategy in order to develop highly biologically active pyrazolo[1,5-a]pyrimidines.

Supplementary Materials

The following are available online. 1H-NMR and 13C-NMR of compounds 3, 4, 5ag, 6am.

Author Contributions

B.J. performed the experiments; A.T., M.A., G.G., and M.A. designed and supervised the study. B.J., A.T. and M.A. wrote the paper. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Acknowledgments

We thank the ‘‘Département d’analyses Chimiques et Médicales’’ (Tours, France) for chemical analyses.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Şener, N.; Erişkin, S.; Yavuz, S.; Şener, İ. Synthesis, characterization, solvatochromic properties, and antimicrobial-radical scavenging activities of new diazo dyes derived from pyrazolo[1,5-a]pyrimidine. J. Heterocyclic. Chem. 2017, 54, 3538–3548. [Google Scholar] [CrossRef]
  2. Bondock, S.; Fadaly, W.; Metwally, M.A. Synthesis and antimicrobial activity of some new thiazole, thiophene and pyrazole derivatives containing benzothiazole moiety. Eur. J. Med. Chem. 2010, 45, 3692–3701. [Google Scholar] [CrossRef]
  3. Hassan, A.S.; Mady, M.F.; Awad, H.M.; Hafez, T.S. Synthesis and antitumor activity of some new pyrazolo[1,5-a]pyrimidines. Chin. Chem. Lett. 2017, 28, 388–393. [Google Scholar] [CrossRef]
  4. Metwally, N.H.; Abdelrazek, F.M.; Eldaly, S.M. Synthesis and anticancer activity of some new heterocyclic compounds based on 1-cyanoacetyl-3,5-dimethylpyrazole. Res. Chem. Intermed. 2016, 42, 1071–1089. [Google Scholar] [CrossRef]
  5. Kamal, A.; Tamboli, J.R.; Nayak, V.L.; Adil, S.F.; Vishnuvardhan, M.V.P.S.; Ramakrishna, S. Synthesis of pyrazolo[1,5-a]pyrimidine linked aminobenzothiazole conjugates as potential anticancer agents. Bioorg. Med. Chem. Lett. 2013, 23, 3208–3215. [Google Scholar] [CrossRef]
  6. Williamson, D.S.; Parratt, M.J.; Bower, J.F.; Moore, J.D.; Richardson, C.M.; Dokurno, P.; Jackson, P.S. Structure-guided design of pyrazolo[1,5-a]pyrimidines as inhibitors of human cyclin-dependent kinase 2. Bioorg. Med. Chem. Lett. 2005, 15, 863–867. [Google Scholar] [CrossRef]
  7. Paruch, K.; Dwyer, M.P.; Alvarez, C.; Brown, C.; Chan, T.Y.; Doll, R.J.; Keertikar, K.; Knutson, C.; McKittrick, B.; Rivera, J.; et al. Pyrazolo[1,5-a]pyrimidines as orally available inhibitors of cyclin-dependent kinase 2. Bioorg. Med. Chem. Lett. 2007, 17, 6220–6223. [Google Scholar] [CrossRef]
  8. Ali, S.; Heathcote, D.A.; Kroll, S.H.; Jogalekar, A.S.; Scheiper, B.; Patel, H.; Brackow, J.; Siwicka, A.; Fuchter, M.J.; Periyasamy, M.; et al. The development of a selective cyclin-dependent kinase inhibitor that shows antitumor activity. Cancer Res. 2009, 69, 6208–6215. [Google Scholar] [CrossRef] [Green Version]
  9. Selleri, S.; Bruni, F.; Costagli, C.; Costanzo, A.; Guerrini, G.; Ciciani, G.; Gratteri, P.; Besnard, F.; Costa, B.; Montali, M.; et al. A novel selective GABAA α1 receptor agonist displaying sedative and anxiolytic-like properties in rodents. J. Med. Chem. 2005, 48, 6756–6760. [Google Scholar] [CrossRef]
  10. Chen, Y.L. Substituted 6,5-Hetero-Bicyclic Derivatives. WO 98/08847, 5 March 1998. [Google Scholar]
  11. Boes, M.; Stadler, H.; Riemer, C. Pyrazolopyrimidine and Pyrazolines and Process for Preparation Thereof. U.S. Patent 6194410, 27 February 2001. [Google Scholar]
  12. Ramsey, S.J.; Attkins, N.J.; Fish, R.; van der Graaf, P.H. Quantitative pharmacological analysis of antagonist binding kinetics at CRF1 receptors in vitro and in vivo. Br. J. Pharmacol. 2011, 164, 992–1007. [Google Scholar] [CrossRef] [Green Version]
  13. Tellew, J.E.; Lanier, M.; Moorjani, M.; Lin, E.; Luo, Z.; Slee, D.H.; Zhang, X.; Hoare, S.R.; Grigoriadis, D.E.; St Denis, Y.; et al. Discovery of NBI-77860/GSK561679, a potent corticotropin-releasing factor (CRF1) receptor antagonist with improved pharmacokinetic properties. Bioorg. Med. Chem. Lett. 2010, 20, 7259–7264. [Google Scholar] [CrossRef] [PubMed]
  14. Dusza, J.P.; Tomcufcik, A.S.; Albright, J.D. Aryl and Heteroaryl[[7-(3-disubstituted amino)phenyl]pyrazolo[1,5-a]pyrimidin-3-yl]methanones. U.S. Patent 4654347, 31 March 1987. [Google Scholar]
  15. Ivashchenko, A.V.; Golovina, E.S.; Kadieva, M.G.; Kysil, V.M.; Mitkin, O.D.; Okun, I.M. Antagonists of serotonin 5-HT 6 receptors. III. Pyridine-substituted 3-(phenylsulfonyl)pyrazolo[1,5-a]pyrimidines: Synthesis and structure–activity relationship. Pharm. Chem. J. 2012, 46, 406–410. [Google Scholar] [CrossRef]
  16. Fang, Z.; Wang, T.Q.; Li, H.; Zhang, G.; Wu, X.A.; Yang, L.; Peng, Y.L.; Zou, J.; Li, L.L.; Xiang, R.; et al. Discovery of pyrazolo[1,5-a]pyrimidine-3-carbonitrile derivatives as a new class of histone lysine demethylase 4D (KDM4D) inhibitors. Bioorg. Med. Chem. Lett. 2017, 27, 3201–3204. [Google Scholar] [CrossRef] [PubMed]
  17. Xu, Y.; Brenning, B.G.; Kultgen, S.G.; Foulks, J.M.; Clifford, A.; Lai, S.; Chan, A.; Merx, S.; McCullar, M.V.; Kanner, S.B.; et al. Synthesis and biological evaluation of pyrazolo[1,5-a]pyrimidine compounds as potent and selective Pim-1 inhibitors. ACS Med. Chem. Lett. 2015, 6, 63–67. [Google Scholar] [CrossRef] [Green Version]
  18. Wang, X.; Magnuson, S.; Pastor, R.; Fan, E.; Hu, H.; Tsui, V.; Deng, W.; Murray, J.; Steffek, M.; Wallweber, H.; et al. Discovery of novel pyrazolo[1,5-a]pyrimidines as potent pan-Pim inhibitors by structure-and property-based drug design. Bioorg. Med. Chem. Lett. 2013, 23, 3149–3153. [Google Scholar] [CrossRef]
  19. Liu, Y.; Laufer, R.; Patel, N.K.; Ng, G.; Sampson, P.B.; Li, S.W.; Lang, Y.; Feher, M.; Brokx, R.; Beletskaya, I.; et al. Discovery of pyrazolo[1, 5-a]pyrimidine TTK inhibitors: CFI-402257 is a potent, selective, bioavailable anticancer agent. ACS Med. Chem. Lett. 2016, 7, 671–675. [Google Scholar] [CrossRef] [Green Version]
  20. Paruch, K.; Dwyer, M.P.; Alvarez, C.; Brown, C.; Chan, T.Y.; Doll, R.J.; Keertikar, K.; Knutson, C.; McKittrick, B.; Rivera, J.; et al. Discovery of dinaciclib (SCH 727965): A potent and selective inhibitor of cyclin-dependent kinases. ACS Med. Chem. Lett. 2010, 1, 204–208. [Google Scholar] [CrossRef] [Green Version]
  21. Mackman, R.L.; Sangi, M.; Sperandio, D.; Parrish, J.P.; Eisenberg, E.; Perron, M.; Hui, H.; Zhang, L.; Siegel, D.; Yang, H.; et al. Discovery of an oral respiratory syncytial virus (RSV) fusion inhibitor (GS-5806) and clinical proof of concept in a human RSV challenge study. J. Med. Chem. 2015, 58, 1630–1643. [Google Scholar] [CrossRef]
  22. Hwang, J.Y.; Windisch, M.P.; Jo, S.; Kim, K.; Kong, S.; Kim, H.C.; Kim, S.; Kim, H.; Lee, M.E.; Kim, Y.; et al. Discovery and characterization of a novel 7-aminopyrazolo[1,5-a]pyrimidine analog as a potent hepatitis C virus inhibitor. Bioorg. Med. Chem. Lett. 2012, 22, 7297–7301. [Google Scholar] [CrossRef]
  23. Mikami, S.; Kawasaki, M.; Ikeda, S.; Negoro, N.; Nakamura, S.; Nomura, I.; Ashizawa, T.; Kokubo, H.; Hoffman, I.D.; Zou, H.; et al. Discovery of a novel series of pyrazolo[1,5-a]pyrimidine-based phosphodiesterase 2A inhibitors structurally different from N-((1S)-1-(3-fluoro-4-(trifluoromethoxy)phenyl)-2-methoxyethyl)-7-methoxy-2-oxo-2,3-dihydropyrido[2,3-b]pyrazine-4(1H)-carboxamide (TAK-915), for the treatment of cognitive disorders. Chem. Pharm. Bull. 2017, 65, 1058–1077. [Google Scholar]
  24. Koizumi, Y.; Tanaka, Y.; Matsumura, T.; Kadoh, Y.; Miyoshi, H.; Hongu, M.; Takedomi, K.; Kotera, J.; Sasaki, T.; Taniguchi, H.; et al. Discovery of a pyrazolo[1,5-a]pyrimidine derivative (MT-3014) as a highly selective PDE10A inhibitor via core structure transformation from the stilbene moiety. Bioorg. Med. Chem. 2019, 27, 3440–3450. [Google Scholar] [CrossRef] [PubMed]
  25. Yamagami, T.; Kobayashi, R.; Moriyama, N.; Horiuchi, H.; Toyofuku, E.; Kadoh, Y.; Kawanishi, E.; Izumoto, S.; Hiramatsu, H.; Nanjo, T.; et al. Scalable process design for a PDE10A inhibitor consisting of pyrazolopyrimidine and quinoxaline as key units. Org. Process Res. Dev. 2019, 23, 578–587. [Google Scholar] [CrossRef]
  26. Raheem, I.T.; Schreier, J.D.; Fuerst, J.; Gantert, L.; Hostetler, E.D.; Huszar, S.; Joshi, A.; Kandebo, M.; Kim, S.H.; Li, J.; et al. Discovery of pyrazolopyrimidine phosphodiesterase 10A inhibitors for the treatment of schizophrenia. Bioorg. Med. Chem. Lett. 2016, 26, 126–132. [Google Scholar] [CrossRef] [PubMed]
  27. Zhou, Y.; Wang, J.; Gu, Z.; Wang, S.; Zhu, W.; Aceña, J.L.; Soloshonok, V.A.; Izawa, K.; Liu, H. Next generation of fluorine-containing pharmaceuticals, compounds currently in phase II–III clinical trials of major pharmaceutical companies: New structural trends and therapeutic areas. Chem. Rev. 2016, 116, 422–518. [Google Scholar] [CrossRef]
  28. Jeffries, B.; Wang, Z.; Graton, J.; Holland, S.D.; Brind, T.; Greenwood, R.D.R.; Le Questel, J.Y.; Scott, J.S.; Chiarparin, E.; Linclau, B. Reducing the Lipophilicity of Perfluoroalkyl Groups by CF2–F/CF2–Me or CF3/CH3 Exchange. J. Med. Chem. 2018, 61, 10602–10618. [Google Scholar] [CrossRef] [Green Version]
  29. Aggarwal, R.; Masan, E.; Kaushik, P.; Kaushik, D.; Sharma, C.; Aneja, K.R. Synthesis and biological evaluation of 7-trifluoromethylpyrazolo[1,5-a]pyrimidines as anti-inflammatory and antimicrobial agents. J. Fluor. Chem. 2014, 168, 16–24. [Google Scholar] [CrossRef]
  30. Yoshida, M.; Mori, A.; Inaba, A.; Oka, M.; Makino, H.; Yamaguchi, M.; Fujita, H.; Kawamoto, T.; Goto, M.; Kimura, H.; et al. Synthesis and structure–activity relationship of tetrahydropyrazolopyrimidine derivatives—A novel structural class of potent calcium-sensing receptor antagonists. Bioorg. Med. Chem. 2010, 18, 8501–8511. [Google Scholar] [CrossRef]
  31. Emelina, E.E.; Petrov, A.A.; Selivanov, S.I.; Nelyubina, Y.V.; Antipin, M.Y. Highly regioselective synthesis of trifluoromethyl derivatives of pyrazolo[1,5-a]pyrimidines bearing fused cycloalkane rings using (2-ethoxycycloalkenyl)-2,2,2-trifluoroethanones. J. Fluor. Chem. 2009, 130, 861–869. [Google Scholar] [CrossRef]
  32. Shaaban, M.R. Microwave-assisted synthesis of fused heterocycles incorporating trifluoromethyl moiety. J. Fluor. Chem. 2008, 129, 1156–1161. [Google Scholar] [CrossRef]
  33. Petrov, A.A.; Emelina, E.E.; Selivanov, S.I. α-aminoazoles in synthesis of heterocycles: IV. Regiodirection of 3(5)-amino-5(3)-methylpyrazole reaction with hexafluoroacetylacetone. Russ. J. Org. Chem. 2008, 44, 263–269. [Google Scholar] [CrossRef]
  34. Martins, M.A.P.; Cunico, W.; Scapin, E.; Emmerich, D.J.; Fiss, G.F.; Rosa, F.A.; Bonacorso, H.G.; Zanatta, N.; Flores, A.F.C. Microwave-assisted regiospecific synthesis of 2-trifluoromethyl-7-trihalomethylated pyrazolo[1,5-a]pyrimidines. Lett. Org. Chem. 2006, 3, 358–362. [Google Scholar] [CrossRef]
  35. Krasovsky, A.L.; Hartulyari, A.S.; Nenajdenko, V.G.; Balenkova, E.S. Efficient syntheses of new CF3-containing diazolopyrimidines. Synthesis 2002, 2002, 133–137. [Google Scholar] [CrossRef]
  36. Emelina, E.E.; Petrov, A.A.; Firsov, A.V. Aminoazoles in Heterocycles Synthesis: II. Trifluoromethyl-containing Diketones in the Synthesis of Pyrazolo[1,5-a]pyrimidines. Russ. J. Org. Chem. 2001, 37, 852–858. [Google Scholar] [CrossRef]
  37. Jismy, B.; Guillaumet, G.; Allouchi, H.; Akssira, M.; Abarbri, A. Concise and efficient eccess to 5,7-disubstituted pyrazolo[1, 5-a]pyrimidines by Pd-Catalyzed sequential arylation, alkynylation and SNAr Reaction. Eur. J. Org. Chem. 2017, 6168–6178. [Google Scholar] [CrossRef]
  38. Petrignet, J.; Thiery, E.; Silpa, L.; Abarbri, M. Mild and Direct Access to 7-Substituted-4-trifluoromethylpyrimido[1,2-b]pyridazin-2-one Systems. Synthesis 2014, 46, 947–954. [Google Scholar] [CrossRef]
  39. Silpa, L.; Petrignet, J.; Abarbri, M. Direct access to fluorinated thiadiazolo[3,2-a]pyrimidin-7-one systems. Synlett 2014, 25, 1827–1830. [Google Scholar]
  40. Silpa, L.; Niepceron, A.; Laurent, F.; Brossier, F.; Pénichon, M.; Enguehart-Gueiffier, C.; Abarbri, M.; Silvestre, A.; Petrignet, J. Synthesis and evaluation of the anticoccidial activity of trifluoropyrido[1,2-a]pyrimidin-2-one derivatives. Bioorg. Med. Chem. Lett. 2016, 26, 114–120. [Google Scholar] [CrossRef]
  41. Jismy, B.; Allouchi, H.; Guillaumet, G.; Akssira, M.; Abarbri, A. An efficient synthesis of new 7-trifluoromethyl-2,5-disubstituted pyrazolo[1,5-a]pyrimidines. Synthesis 2018, 50, 1675–1686. [Google Scholar]
  42. Dwyer, M.P.; Keertikar, K.; Paruch, K.; Alvarez, C.; Labroli, M.; Poker, C.; Fischmann, T.O.; Mayer-Ezell, R.; Bond, R.; Wang, Y.; et al. Discovery of pyrazolo[1,5-a]pyrimidine-based Pim inhibitors: A template-based approach. Bioorg. Med. Chem. Lett. 2013, 23, 6178–6182. [Google Scholar] [CrossRef]
  43. Fox, C.J.; Hammerman, P.S.; Thompson, C.B. The Pim kinases control rapamycin-resistant T cell survival and activation. J. Exp. Med. 2005, 201, 259–266. [Google Scholar] [CrossRef] [Green Version]
Sample Availability: Not available.
Scheme 1. Synthesis of 3-bromo-7-(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-5-one 4.
Scheme 1. Synthesis of 3-bromo-7-(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-5-one 4.
Molecules 25 02062 sch001
Scheme 2. Various nucleophilic aromatic substitutions at C-5 position of 4.
Scheme 2. Various nucleophilic aromatic substitutions at C-5 position of 4.
Molecules 25 02062 sch002
Scheme 3. Path A and B to design 3,5-disubstituted 7-(trifluoromethyl)pyrazolo[1,5-a]pyrimidine 6a.
Scheme 3. Path A and B to design 3,5-disubstituted 7-(trifluoromethyl)pyrazolo[1,5-a]pyrimidine 6a.
Molecules 25 02062 sch003
Scheme 4. Synthesis of various 3,5-disubstitued 7-(trifluoromethyl)pyrazolo[1,5-a]pyrimidines 6a–m.
Scheme 4. Synthesis of various 3,5-disubstitued 7-(trifluoromethyl)pyrazolo[1,5-a]pyrimidines 6a–m.
Molecules 25 02062 sch004
Scheme 5. Synthesis of trifluoromethylated analogue 9. The IC50 value is against Pim1 kinase [17,42].
Scheme 5. Synthesis of trifluoromethylated analogue 9. The IC50 value is against Pim1 kinase [17,42].
Molecules 25 02062 sch005
Table 1. Optimization of Suzuki–Miyaura cross-coupling between 5f and p-methoxyphenylboronic acid.
Table 1. Optimization of Suzuki–Miyaura cross-coupling between 5f and p-methoxyphenylboronic acid.
Molecules 25 02062 i001
EntryOrganic SolventBase[Pd]LigandT (°C)TimeRatio a
6a7
1DioxaneNa2CO3PdCl2(PPh3)2-11012 h1090
2DioxaneNa2CO3PdCl2dppf-11012 h2080
3DioxaneK2CO3PdCl2dppf-11012 h2278
4DioxaneK2CO3XPhosPdG2XPhos13512 h3070
5 bDioxaneK2CO3XPhosPdG2XPhos135 b40 min4555
6 bEtOHK2CO3XPhosPdG2XPhos135 b40 min100 (93) c0
7 bEtOHNa2CO3XPhosPdG2XPhos135 b40 min8416
8 bEtOHK2CO3XPhosPdG2-135 b40 min5644
9 b,dEtOHK2CO3XPhosPdG2-13540 min100 (85) c0
a The ratio of mixture (6a/7) was calculated from the crude 1H NMR spectrum. b Reaction was performed under microwave irradiation. c Yield of isolated product 6a. d 5 mol% of catalyst and ligand were used in this case. PdCl2(PPh3)2: Bis(triphenylphosphine)palladium(II) dichloride. PdCl2dppf: [1,1′-Bis(diphenylphosphino)ferrocene]dichloropalladium(II).XPhosPdG2: Chloro(2-dicyclohexylphosphino-2′,4′,6′-triisopropyl-1,1′-biphenyl)[2-(2′-amino-1,1′-biphenyl)]-palladium. XPhos: 2-Dicyclohexylphosphino-2′,4′,6′-triisopropylbiphenyl.

Share and Cite

MDPI and ACS Style

Jismy, B.; Tikad, A.; Akssira, M.; Guillaumet, G.; Abarbri, M. Efficient Access to 3,5-Disubstituted 7-(Trifluoromethyl)pyrazolo[1,5-a]pyrimidines Involving SNAr and Suzuki Cross-Coupling Reactions. Molecules 2020, 25, 2062. https://doi.org/10.3390/molecules25092062

AMA Style

Jismy B, Tikad A, Akssira M, Guillaumet G, Abarbri M. Efficient Access to 3,5-Disubstituted 7-(Trifluoromethyl)pyrazolo[1,5-a]pyrimidines Involving SNAr and Suzuki Cross-Coupling Reactions. Molecules. 2020; 25(9):2062. https://doi.org/10.3390/molecules25092062

Chicago/Turabian Style

Jismy, Badr, Abdellatif Tikad, Mohamed Akssira, Gérald Guillaumet, and Mohamed Abarbri. 2020. "Efficient Access to 3,5-Disubstituted 7-(Trifluoromethyl)pyrazolo[1,5-a]pyrimidines Involving SNAr and Suzuki Cross-Coupling Reactions" Molecules 25, no. 9: 2062. https://doi.org/10.3390/molecules25092062

Article Metrics

Back to TopTop