Next Article in Journal
Minimal Residual Disease Detection: Bridging Molecular and Clinical Strategies for Recurrence Prevention in Gynecologic Cancers
Previous Article in Journal
Integrative Neuroimmune Role of the Parasympathetic Nervous System, Vagus Nerve and Gut Microbiota in Stress Modulation: A Narrative Review
Previous Article in Special Issue
In Silico and In Vivo Evaluation of Novel 2-Aminobenzothiazole Derivative Compounds as Antidiabetic Agents
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

1,2,3-Triazolo-Bridged Click Coupling of Pinane-Based Azidodiol Enantiomers with Pyrimidine- and Purine-Based Building Blocks: Synthesis, Antiproliferative, and Antimicrobial Evaluation

1
Institute of Pharmaceutical Chemistry, University of Szeged, Eötvös utca 6, H-6720 Szeged, Hungary
2
Department of Biotechnology and Microbiology, University of Szeged, Közép fasor 52, H-6726 Szeged, Hungary
3
Institute of Pharmacodynamics and Biopharmacy, University of Szeged, Eötvös utca 6, H-6720 Szeged, Hungary
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2025, 26(23), 11705; https://doi.org/10.3390/ijms262311705
Submission received: 5 November 2025 / Revised: 29 November 2025 / Accepted: 2 December 2025 / Published: 3 December 2025
(This article belongs to the Special Issue Drug Discovery: Design, Synthesis and Activity Evaluation)

Abstract

Key intermediate azidodiols were synthesized according to literature from commercially available (+)- and (−)-α-pinene in a four-step sequence, including epoxidation with mCPBA, allylic rearrangement, a second epoxidation and, finally, a regioselective azidolysis of the resulting epoxide by sodium azide, yielding the enantiomerically pure azidodiols. The pyrimidine-based alkyne building blocks were prepared from dichloropyrimidines following our method reported previously, while the purine-containing alkyne analogues were synthesized in a procedure of two or three steps. Click reactions were carried out in the presence of Cu(OAc)2 and sodium ascorbate. The obtained pinane-coupled 2,4-diaminopyrimidines were screened for antiproliferative activity by MTT assay on HeLa, MD231, SiHa, MCF-7, and A2780 human cancer cell lines compared with fibroblast cells (NIH/3T3), on Gram-positive and Gram-negative pathogenic bacteria, and two yeasts, and the SAR was explained in detail. The prepared compounds showed moderate antiproliferative activity. While the starting azidodiols (+)-2 and (−)-2 exhibited excellent and selective antibacterial activities against S. aureus with a moderate antimycotic effect on C. krusei, only the (−)-enantiomer was active against P. aeruginosa. In a similar manner, most pyrimidine and purine derivatives also expressed moderate antimycotic effect against C. krusei. One of the purine-based derivatives (−)-30 possessed remarkable and selective antibacterial effect against P. aeruginosa.

Graphical Abstract

1. Introduction

Antimicrobial resistance (AMR) and cancer are among the greatest global health concerns nowadays. These threats highlight the urgent need for new therapeutic strategies and pharmacological agents. According to the World Health Organization (WHO) report, AMR directly caused an estimated 1.27 million deaths and contributed to 4.95 million others in 2019. These numbers validate how it is making infections increasingly difficult to treat, thereby undermining the safety of essential medical procedures such as surgery. Similarly, cancer remains a leading cause of death worldwide. Over 20 million new cases and nearly 10 million deaths have been reported in 2022. The major challenges in therapy include treatment resistance and tumour heterogeneity, which further complicate effective management of the disease. These alarming figures underscore the critical need to discover and develop new compounds that could serve as potential candidates for future drug development [1,2,3].
Natural compounds have not only implied an inspiration in modern pharmaceutical research but also served as readily available raw materials in large quantities, enabling cost-effective and environmentally friendly syntheses. Terpenes are also of outstanding importance among such natural compounds. Of these, the monoterpene skeleton α-pinene is particularly attractive, because both enantiomers are cheaply available, making them useful starting materials for enantioselective syntheses. In recent years, we reported the stereoselective syntheses and applications of pinane-based bi- and trifunctional building blocks such as β-amino acids, 1,3-aminoalcohols, aminodiols, and diaminoalcohols with remarkable pharmacological and catalytic activities [4,5,6,7,8].
On the other hand, the role of nitrogen heterocycles in medicinal chemistry is unique and prominent [9,10]. Within this class, 2,4-diaminopyrimidines possess outstanding importance, as they have well-established roles in cancer chemotherapy [11,12,13], such as methotrexate or in the design of antimicrobial agents, for example, trimethoprim [14,15]. These examples underscore their central position in therapeutic development and drug discovery [16]. In parallel, purine heterocycles are also noteworthy as they form the structural foundation of nucleic acids and they are found in many clinically important drugs. These include antiviral agents (e.g., acyclovir) [17] and anticancer therapies (e.g., mercaptopurine) [18]. In addition, some purine derivatives are actively being explored for their antimicrobial potential [19].
As a continuation of our systematic research work combining the structural diversity of terpenes with the pharmacological potential of nitrogen heterocycles, our aim was to explore new chemical species and create compounds with enhanced biological activities, combining the monoterpenic α-pinane skeleton with 2,4-diaminopyrimidine and purine moieties [4,20].

2. Results and Discussion

2.1. Synthesis of Pinan-Based Azides

Azidodiol intermediates (+)- and (−)-2 were obtained from commercially available (+)- and (−)-α-pinene, following a four-step sequence described in the literature [21,22]. The transformation involved the initial epoxidation of the pinene system with mCPBA, followed by allylic rearrangement, a subsequent epoxidation and, finally, a regioselective ring opening of the resulting epoxide with NaN3 in the presence of a catalytic amount of NH4Cl, furnishing the enantiomerically pure azidodiols in good yields (Scheme 1). All compounds were prepared in both enantiomeric forms, but for easier understanding, only the products obtained by starting from (+)-α-pinene are shown in Section 2.

2.2. Synthesis of Key-Intermediate Alkynes

The first group of 2,4-diaminopyrimidine-based alkynes was synthesized according to the procedure published in our recent article [20]. The regioselective reaction of propargylamine with halogen-substituted pyrimidines 3 and 4, followed by the reaction of the obtained 4-aminopyrimidines 5 and 6 with aromatic and heteroaromatic amines, resulted in 2,4-diaminopyrimidines 712 (Scheme 2).
The purine-based alkynes 1519 were prepared according to reported procedures [23,24], starting from 5-amino to 4,6-dichloropyrimidine and propargylamine, followed by ring closure of diamine 14 with triethyl orthoformate (TEOF) or benzoyl chloride. Compound 15 was finally substituted at position 6 via Suzuki coupling or in its reaction with aniline or benzylamine. The obtained new derivatives bearing 6-phenyl, 6-benzylamino, or N-(4-(trifluoromethyl)phenyl)-6-amino substituents on the 9H-purine scaffold are outlined in Scheme 3 [23,24,25].

2.3. Coupling of the Pinane Moiety with Alkyne-Functionalized Heterocycles via Click Reaction

Click chemistry enables the straightforward assembly of diverse molecular fragments in a modular fashion, similar to fitting puzzle pieces together, offering an efficient strategy for constructing a wide range of functional molecules and materials [26,27]. In particular, it is a very efficient method in the copper-catalyzed azide–alkyne cycloaddition (CuAAC) reaction, since it is a rapid, biocompatible, high-yielding reaction that allows the quick construction of complex molecules, accelerating drug discovery and optimization processes [28,29].
In the synthesis of the new hybrid structures, both azides (+)-2 and (−)-2 underwent CuAAC reactions with the previously prepared alkynes 712 and 1419, using Cu(OAc)2·H2O and sodium ascorbate as catalysts, as illustrated in Scheme 4. Reactions were carried out under mild conditions, typically at 45 °C in a 2:1 mixture of tert-BuOH/H2O or THF/H2O, depending on the solubility of the alkyne. For the pyrimidine-based alkynes, either pure tert-BuOH or THF was suitable, as observed also in our previous work, resulting in 2228 [20]. However, in contrast to the diterpene series we reported recently, no solubility problem or precipitation of the products was observed. For the purine-based alkynes, THF proved to be the optimal solvent choice for obtaining compounds 2933. All reactions proceeded in good to excellent yields, and the resulting compounds proved to be stable (Scheme 4).

2.4. In Vitro Antiproliferative Studies of Pinane-Based Diaminopyrimidines and Structure–Activity Relationship

The antiproliferative potential of the synthesized monoterpene-based compounds was assessed in vitro against a panel of human adherent cancer cell lines, including breast (MDA-MB-231, MCF-7), cervical (HeLa, SiHa), and ovarian (A2780) cells, using the MTT assay. Selected results are presented in Figure 1 and Table 1, while data for all examined products are presented in Table S1 in the Supplementary Materials. Analysis of the observed activities allowed preliminary insights into structure–activity relationships (SAR). Compounds bearing a monoamino-substituted pyrimidine unit (2022) displayed minimal antiproliferative effects, which is in accordance with our previous results with other pinane-based amino alcohols and diols bearing similar monoaminopyrimidine substituents, but without the triazole ring linker. [4].
In the case of 2,4-diaminopyrimidine-derived compounds 2328, the activities ranged from mild to moderate. The main factors influencing activity included the type of halogen substituent on the pyrimidine ring, the nature of the ring attached to the pyrimidine core, and the type of substituent group present on the phenyl ring connected to the pyrimidine unit. Compared with our previous results with the directly diol-type pinane-condensed diaminopyrimidines, the introduction of the methylene-triazole linker increased the antiproliferative activity [4].
The most active derivatives were those bearing a trifluoromethyl group at the para position of the aniline substituent. Notably, the type of halogen substituent on the pyrimidine ring also played an important role: 5-fluoro-substituted (+)-23 and (−)-23 showed good antiproliferative activity, in particular, against HeLa adenocarcinoma cells. Meanwhile, 5-chloro analogues (+)-24 and (−)-24 had a more inhibitory effect toward both estrogen receptor-positive (ER+) breast cancer cells (MCF-7). The cancer selectivity of the most potent derivatives, however, is limited, as indicated by calculated selectivity indices (SIs, the ratios of the IC50 values obtained on fibroblasts and cancer cells) that are less than or close to 1. Furthermore, both the 5-chloro and 5-fluoro derivatives exhibited only mild effects against A2780 ovarian cancer cells (Figure 1, Table 1 and Table S1).
These observations are consistent with earlier findings by our group [4]. In another study, wherein we employed the diterpene allo-gibberic acid scaffold instead of the pinane system to prepare similar 2,4-diaminopyrimidine hybrids [25], we observed a comparable trend. In that work, compounds bearing 4-trifluoromethylphenyl moieties either linked through a triazole bridge or a flexible –CH2–CH2– spacer proved to be highly effective, albeit with little selectivity. Comparing the enantiomers, the stereochemistry also appeared to play a weak role, as the (−)-enantiomers ((−)-23 and (−)-24) were generally more effective against MDA-MB-231 and MCF7 cells than their (+)-counterparts.
The results indicate that the type of substituent at the para position of the phenyl ring also influenced cell growth inhibition, which decreased notably when the trifluoromethyl group was replaced with a methyl ester function ((+)-27 and (−)-27). This reduced activity may be explained by the lower lipophilicity of the methyl ester moiety compared to that of the CF3 group [30]. In addition, the ester functionality might be hydrolyzed by intracellular esterase enzymes, further diminishing its contribution to activity [31]. Similarly, the introduction of the 4-morpholine group ((+)-28 and (−)-28), which is even more hydrophilic than the methyl ester, led to an even greater reduction in activity across nearly all tested cell lines.
These results also indicate that compounds bearing a 1-methyl-1H-pyrazol-4-yl ring (25 and 26) exhibited very low activity, consistent with our previous observation [25].
Considering the purine-hybrid compounds, they exhibited very weak antiproliferative activity in both cancerous and non-cancerous cells, except for two compounds that showed only moderate effects. Nevertheless, subtle differences in their inhibition patterns provide valuable clues for developing further structure–activity relationship (SAR) insights. For example, comparing (+)-22 and (−)-22 with their cyclic analogues ((+)-29 and (−)-29), all four compounds showed similar weak activity against HeLa cells; however, the ring closure clearly enhanced activity against MCF-7 and MDA-MB-231 cells. In addition, introducing a phenyl substituent at position 8 of the purine ring ((+)-30 and (−)-30), formed derivatives selectively improved the activity of the (+)-enantiomer, which inhibited MCF-7, MDA-MB-231, and A2780 cells at a 30 μM concentration. In contrast, substitution at C6 with a phenyl group ((+)-31 and (−)-31) diminished the activity of the (+)-enantiomer at the same concentration, while both (−)-enantiomers remained inactive. In a similar manner, incorporating a benzylamine moiety at C6 did not improve the activity either. The most promising members of this series were (+)-33 and (−)-33, bearing an amino-4-(trifluoromethyl)phenyl substituent at C6. These compounds showed weak to moderate activity against both MCF-7 and NIH/3T3 cells. These findings are consistent with previous reports indicating that 1,4-disubstituted 1,2,3-triazolylpurines generally display low cytotoxicity [32,33]. Consequently, further investigation of these molecules is needed, particularly regarding their structural optimization and pharmacological evaluation.

2.5. In Vitro Studies of Antibacterial Effects of 4-Amino- and 2,4-Diaminopyrimidine and Purine Derivatives

Pyrimidine and purine-based compounds, including benzothiazole derivatives, substituted adenines and aminopurines, exerted remarkable antimicrobial activities against several bacteria and yeasts [19,34]; therefore, antimicrobial activities of the analogues were tested against two Gram-positive and two Gram-negative bacteria, as well as two yeasts (Figure 2, Table 2 and Table S2). Generally, almost all examined compounds show moderate to high inhibitory activities against Bacillus subtilis and Escherichia coli. The obtained results clearly show that 4-monoamino-substituted derivatives ((+)- and (−)-20, (+)- and (−)-21) have only weak antibacterial activity, but these compounds possess remarkable antifungal activity against Candida krusei. In turn, the introduction of a primary amino function at position 5 (22) caused moderate antibacterial activity against B. subtilis. Introduction of an amino function at position 2 of the pyrimidine ring practically did not increase the antibacterial effect, but these compounds showed moderate antifungal activity against C. krusei. Similar results were obtained in the case of purine-based compounds (2933) except compound (−)-30, which possessed excellent antibacterial activity against P. aeruginosa in 10 μg/mL concentration.
Since there are several examples of the antibacterial, antifungal or even antiviral activities of alicyclic or heterocyclic compounds bearing azido function [20,35,36,37,38], our azidodiol intermediates (+)- and (−)-2 were also tested. Interestingly, they showed remarkable and selective antimicrobial activity against both Gram-positive and Gram-negative bacteria and a similar moderate activity against yeasts. Remarkable differences were observed between the two enantiomers. Namely, compound (+)-2 showed selective inhibition against S. aureus, while (−)-2 was effective against both S. aureus and P. aeruginosa in 10 μg/mL. It must also be mentioned that these compounds did not show cytotoxicity against human fibroblast cells (NIH/3T3, see Table S1 in Supplementary Materials). Compering with our previous results with the directly diol-type pinane-condensed diaminopyrimidines, the introduction of methylene-triazole linker increased the antifungal activity [4].

3. Materials and Methods

3.1. General Methods

Commercially available reagents were used as obtained from suppliers (Novochem Co., Ltd., 1089 Budapest, Hungary, Orczy út 6.; Merck Ltd., Budapest, Hungary; and VWR International Ltd., Debrecen, Hungary), while solvents were dried according to standard procedures. Chromatographic separations and monitoring of reactions were carried out on Merck Kieselgel 60 (Merck Ltd., Budapest, Hungary). Optical rotations were measured in MeOH at 20 °C with a PerkinElmer 341 polarimeter (PerkinElmer Inc., Shelton, CT, USA). Melting points were determined with a Kofler apparatus (Nagema, Dresden, Germany). HRMS flow injection analysis was performed with a Thermo Scientific Q Exactive Plus hybrid quadrupole-Orbitrap (Thermo Fisher Scientific, Waltham, MA, USA) mass spectrometer coupled to a Waters Acquity I-Class UPLC™ (Waters, Manchester, UK). 1H-, 13C J-MOD-, and 19F-NMR spectra were recorded on a Bruker Avance DRX 500 spectrometer (Bruker Biospin, Karlsruhe, Baden-Württemberg, Germany) [500 MHz (1H), 125 MHz (13C J-MOD), and 470 MHz (19F) δ = 0 (TMS)]. Chemical shifts are expressed in ppm (δ) relative to TMS as an internal reference. J values are given in Hz. All 1H-, 13C, J-MOD-, 19F-NMR, COSY, NOESY, 2D-HMBC, and 2D-HMQC and HRMS spectra are available in the Supporting Information file, as Figures S1–S218; Schemes S1 and S2; Tables S1 and S2.

3.2. Starting Materials

(+)- and (−)-α-Pinene were obtained from Merck KGaA, Darmstadt, Germany. The preparation of azidodiol intermediates (+)- and (−)-2 was accomplished according to literature methods, with spectroscopic data identical to those reported therein [22]. Similarly, intermediates 712, 14, 15 and 17 were prepared via methods reported in the literature and in our earlier publications [20,23,25].

3.3. Synthesis of New Compounds

3.3.1. 6-Chloro-8-phenyl-9-(prop-2-yn-1-yl)-9H-purine 16

To a solution of 14 (0.15 g, 0.82 mmol) in dry toluene (7 mL) NH4Cl was added (6 eq., 0.26 g), followed by the addition of benzoyl chloride (1 eq., 96 μL). The reaction mixture was heated at 100 °C for 2 h, then it was cooled, POCl3 (5.6 mL) was added and the mixture was heated again to 100 °C for 24 h [23]. After stopping the reaction and cooling to room temperature, the reaction mixture was slowly added to ice water dropwise. Afterwards, pH was adjusted to 7–8 by the careful addition of ammonia water (25%), and the mixture was extracted with EtOAc (3 × 25 mL). The combined organic phase was dried over Na2SO4, filtered, and concentrated under reduced pressure. The product was purified by column chromatography using n-hexane/EtOAc (2:1). Yield: 0.12 g, 54%; white crystals; m.p.: 189–191 °C; 1H NMR (500 MHz, DMSO-d6) δ (ppm): 3.54 (1H, t, J = 2.4 Hz), 5.22 (2H, d, J = 2.4 Hz), 7.71–7.65 (3H, m), 8.01 (2H, dd, J = 2.1, 7.4 Hz), 8.87 (1H, s); 13C NMR (125 MHz, DMSO-d6) δ (ppm): 34.4, 77.2, 78.1, 127.4, 128.5, 129.6 (2C), 130.9, 131.9 (2C), 149.0, 152.2, 153.6, 155.8; HRMS (ESI): m/z calcd. for C14H10ClN4 [M + H]+: 269.05885. Found: 269.05847.

3.3.2. N-Benzyl-9-(prop-2-yn-1-yl)-9H-purin-6-amine 18

To a solution of 15 (0.10 g, 0.5 mmol) in acetonitrile (10 mL), (2 eq., 0.11 mL) of benzylamine and (1.5 eq., 0.1 mL) of TEA were added. The reaction mixture was stirred for 24 h at 40 °C [25]. After the evaporation of the solvent, the crude residue was dissolved in 30 mL EtOAc and washed with NaHCO3. Then, the organic layer was dried over Na2SO4, followed by filtration and evaporation. The product was purified by column chromatography using DCM/MeOH (39:1). Yield: 0.12 g, 91%; white crystals; m.p.: 146.5–148.5 °C; 1H NMR (500 MHz, CDCl3) δ(ppm): 2.51 (1H, t, J = 2.3 Hz), 4.88 (2H, s), 4.97 (2H, d, J = 2.3 Hz), 6.19 (1H, s), 7.40–7.27 (5H, m), 7.92 (1H, s), 8.44 (1H, s); 13C NMR (125 MHz, CDCl3) δ (ppm): 33.0 (2C), 74.9, 76.0, 127.5 (2C), 127.8, 128.7 (2C), 138.4 (2C), 138.9, 153.5, 154.8 (2C); HRMS (ESI): m/z calcd. for C15H14N5 [M + H]+: 264.12437. Found: 264.12377.

3.3.3. 9-(Prop-2-yn-1-yl)-N-(4-(trifluoromethyl)phenyl)-9H-purin-6-amine 19

To a solution of 15 (0.10 g, 0.5 mmol) in isopropyl alcohol (5 mL), (1.5 eq., 0.1 mL) of 4-(trifluoromethyl)aniline and two drops of conc. HCl was added. The reaction mixture was stirred for 4 h at 85 °C [23]. After cooling to room temperature, the saturated solution of NaHCO3 (10 mL) was added, and extraction was made with DCM (3 × 30 mL). The organic layer was dried over Na2SO4, filtered, and concentrated. The product was purified by column chromatography using DCM/MeOH (39:1). Yield: 0.16 g, 97%; white crystals; m.p.: 161–163 °C; 1H NMR (500 MHz, CDCl3) δ (ppm): 2.56 (1H, dd, J = 2.5, 2.5 Hz), 5.03 (2H, d, J = 2.5 Hz), 7.64 (2H, d, J = 8.5 Hz), 7.92 (1H, s), 7.98 (2H, d, J = 8.5 Hz), 8.10 (1H, s), 8.61 (1H, s)); 13C NMR (125 MHz, CDCl3) δ (ppm): 33.2, 75.3, 75.7, 119.5 (2C), 121.0, 123.2, 125.0 (q, J = 29.4 Hz), 126.3 (q, J = 3.7 Hz), 140.2, 141.8, 149.5, 151.8, 152.9; 19F NMR (470 MHz, CDCl3) δ (ppm): −61.9; HRMS (ESI): m/z calcd. for C15H11F3N5 [M + H]+: 318.09611. Found: 318.09533.

3.4. General Procedure for Preparation of 1,2,3-Triazols by Click Reaction (2033)

To a solution of azide (+)-2 or (−)-2 (0.05 g, 0.237 mmol) in a mixture of tert-BuOH/H2O (2:1, 12 mL), or THF/H2O (2:1, 12 mL), Cu(OAc)2·H2O (0.05 eq.), sodium ascorbate (0.1 eq.), and the appropriate acetylene derivatives (512, 1419) (1.1 eq., 0.26 mmol) were added. The mixture was stirred for 48 h at 45 °C. Then, the organic solvent was evaporated, and the residue was dissolved in water (10 mL) and extracted with DCM (3 × 30 mL). The organic phase was dried over Na2SO4 and evaporated at low pressure, and the crude product was purified by column chromatography on silica gel.

3.4.1. (1S,2R,3R,5S)-2-((4-(((2-Chloro-5-fluoropyrimidin-4-yl)amino)methyl)-1H-1,2,3-triazol-1-yl)methyl)-6,6-dimethylbicyclo [3.1.1]heptane-2,3-diol (+)-20

The reaction was accomplished starting from (+)-2 and alkyne 5 in tert-BuOH/H2O (2:1) according to the general procedure. The product was purified by column chromatography on silica gel with DCM/MeOH (19:1). Yield: 0.050 g, 53%; white crystals; m.p.: 189–191 °C; α D 20 = +4.0 (c 0.1, MeOH); 1H NMR (500 MHz, DMSO-d6) δ (ppm): 1.04 (3H, s), 1.16 (3H, s), 1.29 (1H, d, J = 10.1 Hz), 1.52 (1H, t, J = 5.8 Hz), 1.59 (1H, dd, J = 3.0, 13.7 Hz), 1.83 (1H, s), 2.06–1.98 (1H, m), 2.43–2.34 (1H, m), 4.00–3.93 (1H, m), 4.27 (1H, d, J = 14.0 Hz), 4.33 (1H, d, J = 14.0 Hz), 4.57 (1H, s), 4.59 (2H, d, J = 5.8 Hz), 5.36 (1H, d, J = 6.3 Hz), 7.88 (1H, s), 8.11 (1H, d, J = 3.3 Hz), 8.69 (1H, t, J = 5.8 Hz); 13C NMR (125 MHz, DMSO-d6) δ (ppm): 24.2, 27.8, 28.0, 36.0, 38.0, 38.5, 40.4, 48.2, 58.5, 64.8, 74.5, 125.1, 140.4 (d, J = 20.4 Hz), 143.6, 145.8 (d, J = 256.5 Hz), 153.7 (d, J = 13.4 Hz), 153.9 (d, J = 2.8 Hz); 19F NMR (470 MHz, DMSO-d6) δ (ppm): −157.14; HRMS (ESI): m/z calcd. for C17H23ClFN6O2 [M + H]+: 397.15550. Found: 397.15438.
The (1R,2S,3S,5R)-enantiomer (−)-20 was synthesized under conditions used for (+)-20; α D 20 = −4.0 (c 0.1, MeOH); all the spectroscopic data and the m.p. were similar to those for the (1R,2S,3S,5R)-enantiomer. HRMS (ESI): m/z calcd. for C17H23ClFN6O2 [M + H]+: 397.15550. Found: 397.15446.

3.4.2. (1S,2R,3R,5S)-2-((4-(((2,5-Dichloropyrimidin-4-yl)amino)methyl)-1H-1,2,3-triazol-1-yl)methyl)-6,6-dimethylbicyclo[3.1.1]heptane-2,3-diol (+)-21

The reaction was accomplished starting from (+)-2 and alkyne 6 in tert-BuOH/H2O (2:1) according to the general procedure. The product was purified by column chromatography on silica gel with DCM/MeOH (39:1). Yield: 0.047 g, 48%; yellowish white crystals; m.p.: 70–73 °C; α D 20 = +3.0 (c 0.1, MeOH); 1H NMR (500 MHz, CDCl3) δ (ppm): 1.11 (3H, s), 1.31 (3H, s), 1.43 (1H, d, J = 10.6 Hz), 1.67 (1H, dd, J = 5.2, 13.9 Hz), 1.98 (2H, d, J = 6.0 Hz), 2.30–2.23 (1H, m), 2.54 (1H, td, J = 5.9, 13.9 Hz), 2.62 (1H, d, J = 5.3 Hz), 3.95 (1H, s), 4.19 (1H, d, J = 13.8 Hz), 4.47 (1H, td, J = 5.0, 10.0 Hz), 4.53 (1H, d, J = 13.8 Hz), 4.77 (2H, ddd, J = 5.6, 15.3, 21.0 Hz), 6.25 (1H, s), 7.78 (1H, s), 8.04 (1H, s); 13C NMR (125 MHz, CDCl3) δ (ppm): 24.3, 27.6, 27.7, 36.6, 37.5, 38.6, 40.5, 50.4, 60.0, 65.2, 74.5, 113.5, 125.2, 142.9, 153.8, 158.3, 158.5; HRMS (ESI): m/z calcd. for C17H23Cl2N6O2 [M + H]+: 413.12595. Found: 413.12510.
The (1R,2S,3S,5R)-enantiomer (−)-21 was synthesized analogously to (+)-21; α D 20 = −3.5 (c 0.1, MeOH); all the spectroscopic data and the m.p. were similar to those for the (1R,2S,3S,5R)-enantiomer. HRMS (ESI): m/z calcd. for C17H23Cl2N6O2 [M + H]+: 413.12595. Found: 413.12515.

3.4.3. (1S,2R,3R,5S)-2-((4-(((5-Amino-6-chloropyrimidin-4-yl)amino)methyl)-1H-1,2,3-triazol-1-yl)methyl)-6,6-dimethylbicyclo[3.1.1]heptane-2,3-diol (+)-22

The reaction was accomplished starting from (+)-2 and alkyne 14 in THF/H2O (2:1) according to the general procedure. The product was purified by column chromatography on silica gel with CHCl3/MeOH (19:1) then increased to (9:1). Yield: 0.074 g, 79%; yellow crystals; m.p.: 94–97 °C; α D 20 = +9.0 (c 0.1, MeOH); 1H NMR (500 MHz, CDCl3) δ (ppm): 1.09 (3H, s), 1.30 (3H, s), 1.42 (1H, d, J = 10.5 Hz), 1.66 (1H, dd, J = 4.7, 13.8 Hz), 2.01–1.94 (2H, m), 2.29–2.21 (1H, m), 2.55–2.47 (1H, m), 4.14 (1H, d, J = 13.9 Hz), 4.43 (1H, dd, J = 5.0, 9.4 Hz), 4.52 (1H, d, J = 13.9 Hz), 4.71 (2H, d, J = 5.0 Hz), 5.97 (1H, s), 7.76 (1H, s), 8.03 (1H, s); 13C NMR (125 MHz, CDCl3) δ (ppm): 24.3, 27.6, 27.7, 36.6, 37.6, 38.6, 40.4, 50.4, 60.1, 64.8, 74.5, 122.5, 125.1, 142.3, 144.2, 148.9, 153.9; HRMS (ESI): m/z calcd. for C17H25ClN7O2 [M + H]+: 394.17582. Found: 394.17501.
The (1R,2S,3S,5R)-enantiomer (−)-22 was synthesized analogously to (+)-22; α D 20 = −6.0 (c 0.1, MeOH); all the spectroscopic data and the m.p. were similar to those for the (1R,2S,3S,5R)-enantiomer. HRMS (ESI): m/z calcd. for C17H25ClN7O2 [M + H]+: 394.17582. Found: 394.17480.

3.4.4. (1S,2R,3R,5S)-2-((4-(((5-Fluoro-2-((4-(trifluoromethyl)phenyl)amino)pyrimidin-4-yl)amino)methyl)-1H-1,2,3-triazol-1-yl)methyl)-6,6-dimethylbicyclo[3.1.1]heptane-2,3-diol (+)-23

The reaction was accomplished starting from (+)-2 and alkyne 7 in tert-BuOH/H2O (2:1) according to the general procedure. The product was purified by column chromatography on silica gel with DCM/MeOH (19:1). Yield: 0.090 g, 73%; white crystals; m.p.: 105–108 °C; α D 20 = +7.0 (c 0.1, MeOH); 1H NMR (500 MHz, CDCl3) δ (ppm): 1.10 (3H, s), 1.29 (3H, s), 1.40 (1H, d, J = 10.6 Hz), 1.67–1.63 (1H, m), 1.99–1.92 (2H, m), 2.27–2.20 (1H, m), 2.55–2.48 (1H, m), 3.92 (1H, broad s), 4.15 (1H, d, J = 13.8 Hz), 4.45 (1H, dt, J = 4.9, 4.8 Hz), 4.51 (1H, d, J = 13.8 Hz), 4.77 (2H, dt, J = 5.7, 15.8 Hz), 5.67 (1H, t, J = 5.6 Hz), 7.16 (1H, s), 7.53 (1H, d, J = 8.6 Hz), 7.68 (3H, d, J = 7.1 Hz), 7.83 (1H, d, J = 3.0 Hz); 13C NMR (125 MHz, CDCl3) δ (ppm): 24.2, 27.5, 27.7, 36.3, 37.5, 38.6, 40.5, 50.4, 60.0, 65.2, 74.5, 117.8 (2C), 124.5, 126.1 (q, J = 3.8 Hz) (2C), 138.9, 139.1, 140.8, 142.8, 143.2, 143.9, 152.3 (d, J = 12.2 Hz), 155.0 (d, J = 2.9 Hz); 19F NMR (470 MHz, CDCl3) δ (ppm): −167.11, −61.69; HRMS (ESI): m/z calcd. for C24H28F4N7O2 [M + H]+: 522.22351. Found: 522.22254.
The (1R,2S,3S,5R)-enantiomer (−)-23 was synthesized analogously to (+)-23; α D 20 = −6.5 (c 0.1, MeOH); all the spectroscopic data and the m.p. were similar to those for the (1R,2S,3S,5R)-enantiomer. HRMS (ESI): m/z calcd. for C24H28F4N7O2 [M + H]+: 522.22351. Found: 522.22137.

3.4.5. (1S,2R,3R,5S)-2-((4-(((5-Chloro-2-((4-(trifluoromethyl)phenyl)amino)pyrimidin-4-yl)amino)methyl)-1H-1,2,3-triazol-1-yl)methyl)-6,6-dimethylbicyclo[3.1.1]heptane-2,3-diol (+)-24

The reaction was accomplished starting from (+)-2 and alkyne 8 in tert-BuOH/H2O (2:1) according to the general procedure. The product was purified by column chromatography on silica gel with DCM/MeOH (19:1). Yield: 0.074 g, 58%; white crystals; m.p.: 140–142 °C; α D 20 = +5.0 (c 0.1, MeOH); 1H NMR (500 MHz, DMSO) δ (ppm): 1.02 (3H, s), 1.11 (3H, s), 1.25 (1H, d, J = 10.4 Hz), 1.51 (1H, t, J = 5.8 Hz), 1.55 (1H, dd, J = 5.1, 13.8 Hz), 1.83–1.77 (1H, m), 1.99–1.92 (1H, m), 2.41–2.33 (1H, m), 3.98–3.92 (1H, m), 4.24 (1H, d, J = 14.0 Hz), 4.32 (1H, d, J = 14.0 Hz), 4.49 (1H, s), 4.68 (2H, d, J = 5.9 Hz), 5.32 (1H, d, J = 6.2 Hz), 7.52 (2H, d, J = 8.6 Hz), 7.78 (1H, t, J = 5.8 Hz), 7.85 (3H, d, J = 7.4 Hz), 8.03 (1H, s); 13C NMR (125 MHz, DMSO) δ (ppm): 24.2, 27.7, 27.9, 36.7, 38.0, 38.5, 40.4, 48.2, 58.5, 64.8, 74.5, 105.2, 118.5 (2C), 121.0, 121.2, 124.5, 126.0 (2C, q, J = 3.6 Hz), 144.8, 144.9, 153.6, 157.8, 158.0; 19F NMR (470 MHz, DMSO) δ (ppm): −59.82; HRMS (ESI): m/z calcd. for C24H28ClF3N7O2 [M + H]+: 538.19396. Found: 538.19358.
The (1R,2S,3S,5R)-enantiomer (−)-24 was synthesized analogously to (+)-24; α D 20 = −6.0 (c 0.1, MeOH); all the spectroscopic data and the m.p. were similar to those for the (1R,2S,3S,5R)-enantiomer. HRMS (ESI): m/z calcd. for C24H28ClF3N7O2 [M + H]+: 538.19396. Found: 538.19237.

3.4.6. (1S,2R,3R,5S)-2-((4-(((5-Fluoro-2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)amino)methyl)-1H-1,2,3-triazol-1-yl)methyl)-6,6-dimethylbicyclo[3.1.1]heptane-2,3-diol (+)-25

The reaction was accomplished starting from (+)-2 and alkyne 9 in tert-BuOH/H2O (2:1) according to the general procedure. The product was purified by column chromatography on silica gel with DCM/MeOH (9:1). Yield: 0.096 g, 88%; pinkish white crystals; m.p.: 110–113 °C; α D 20 = +1.0 (c 0.2, MeOH); 1H NMR (500 MHz, CDCl3) δ (ppm): 1.10 (3H, s), 1.29 (3H, s), 1.41 (1H, d, J = 10.5 Hz), 1.67 (1H, dd, J = 4.6, 14.0 Hz), 1.98–1.91 (2H, m), 2.26–2.18 (1H, m), 2.55–2.47 (1H, m), 3.85 (3H, s), 4.17 (1H, d, J = 13.7 Hz), 4.37 (1H, dd, J = 5.0, 9.5 Hz), 4.50 (1H, d, J = 13.7 Hz), 4.72 (2H, ddd, J = 5.1, 14.9, 19.3 Hz), 5.74 (1H, s), 6.91 (1H, s), 7.43 (1H, s), 7.63 (1H, s), 7.66 (1H, s), 7.74 (1H, d, J = 3.1 Hz); 13C NMR (125 MHz, CDCl3) δ (ppm): 24.2, 27.5, 27.6, 36.3, 37.6, 38.6, 39.2, 40.5, 50.2, 59.8, 65.0, 74.6, 121.2, 123.3, 124.5, 131.1, 138.6 (d, J = 20.2 Hz), 141.1 (d, J = 244.2 Hz), 144.2, 152.6 (d, J = 12.3 Hz), 155.5; 19F NMR (470 MHz, CDCl3) δ (ppm): −169.82; HRMS (ESI): m/z calcd. for C21H29FN9O2 [M + H]+: 458.24228. Found: 458.24172.
The (1R,2S,3S,5R)-enantiomer (−)-25 was synthesized analogously to (+)-25; α D 20 = −3.0 (c 0.2, MeOH); all the spectroscopic data and the m.p. were similar to those for the (1R,2S,3S,5R)-enantiomer. HRMS (ESI): m/z calcd. for C21H29FN9O2 [M + H]+: 458.24228. Found: 458.24147.

3.4.7. (1S,2R,3R,5S)-2-((4-(((5-Chloro-2-((1-methyl-1H-pyrazol-4-yl)amino)pyrimidin-4-yl)amino)methyl)-1H-1,2,3-triazol-1-yl)methyl)-6,6-dimethylbicyclo[3.1.1]heptane-2,3-diol (+)-26

The reaction was accomplished starting from (+)-2 and alkyne 10 in tert-BuOH/H2O (2:1) according to the general procedure. The product was purified by column chromatography on silica gel with DCM/MeOH (9:1). Yield: 0.107 g, 95%; pale pink crystals; m.p.: 116–119 °C; α D 20 = +2.0 (c 0.1, MeOH); 1H NMR (500 MHz CDCl3,) δ (ppm): 1.10 (3H, s), 1.28 (3H, s), 1.40 (1H, d, J = 10.5 Hz), 1.67 (2H, dd, J = 5.2, 14.2 Hz), 1.95 (2H, d, J = 5.7 Hz), 2.26–2.19 (1H, m), 2.55–2.47 (1H, m), 3.85 (3H, s), 4.17 (1H, d, J = 13.8 Hz), 4.38 (1H, dd, J = 5.0, 9.4 Hz), 4.50 (1H, d, J = 13.9 Hz), 4.73 (2H, tdd, J = 6.7, 13.4, 13.4 Hz), 5.91 (1H, s), 6.97 (1H, s), 7.44 (1H, s), 7.61 (1H, s), 7.65 (1H, s), 7.86 (1H, s); 13C NMR (125 MHz, CDCl3) δ (ppm): 24.3, 27.5, 27.6, 36.8, 37.6, 38.6, 39.2, 40.5, 50.1, 59.8, 64.9, 74.5, 121.4, 122.8, 124.5, 131.2, 144.3 (2C), 152.8, 157.7, 157.9; HRMS (ESI): m/z calcd. for C21H29ClN9O2 [M + H]+: 474.21273. Found: 474.21236.
The (1R,2S,3S,5R)-enantiomer (−)-26 was synthesized analogously to (+)-26; α D 20 = −2.5 (c 0.1, MeOH); all the spectroscopic data and the m.p. were similar to those for the (1R,2S,3S,5R)-enantiomer. HRMS (ESI): m/z calcd. for C21H29ClN9O2 [M + H]+: 474.21273. Found: 474.21125.

3.4.8. Methyl 4-((5-chloro-4-(((1-(((1S,2R,3R,5S)-2,3-dihydroxy-6,6-dimethylbicyclo[3.1.1]heptan-2-yl)methyl)-1H-1,2,3-triazol-4-yl)methyl)amino)pyrimidin-2-yl)amino)benzoate (+)-27

The reaction was accomplished starting from (+)-2 and alkyne 12 in THF/H2O (2:1) according to the general procedure. The product was purified by column chromatography on silica gel with DCM/MeOH (19:1). Yield: 0.110 g, 88%; white crystals; m.p.: 131–134 °C; α D 20 = +5.5 (c 0.1 MeOH); 1H NMR (500 MHz, CDCl3) δ (ppm): 1.09 (3H, s), 1.28 (3H, s), 1.39 (1H, d, J = 10.7 Hz), 1.65 (1H, dd, J = 5.0, 14.0 Hz), 1.98–1.93 (2H, m), 2.23 (1H, ddd, J = 5.7, 5.7, 10.8 Hz), 2.55–2.49 (1H, m), 2.81 (1H, broad s), 3.88 (3H, s), 3.93 (1H, s), 4.14 (1H, d, J = 13.8 Hz), 4.43 (1H, dd, J = 5.2, 9.4 Hz), 4.49 (1H, d, J = 13.8 Hz), 4.77 (2H, dt, J = 5.7, 16.2 Hz), 5.92 (1H, t, J = 5.4 Hz), 7.27 (1H, s), 7.64 (2H, d, J = 8.4 Hz), 7.67 (1H, s), 7.94 (1H, s), 7.97 (2H, d, J = 8.4 Hz); 13C NMR (125 MHz, CDCl3) δ (ppm): 24.2, 27.5, 27.7, 36.8, 37.4, 38.6, 40.5, 50.3, 51.8, 59.9, 65.2, 74.5, 106.1, 117.8 (2C), 123.3, 124.6, 130.8 (2C), 144.0, 144.1, 153.2, 157.4, 157.7, 166.9; HRMS (ESI): m/z calcd. for C25H31ClN7O4 [M + H]+: 528.21206. Found: 528.21088.
The (1R,2S,3S,5R)-enantiomer (−)-27 was synthesized analogously to those used for (+)-27; α D 20 = −4.5 (c 0.1, MeOH); all the spectroscopic data and the m.p. were similar to those for the (1R,2S,3S,5R)-enantiomer. HRMS (ESI): m/z calcd. for C25H31ClN7O4 [M + H]+: 528.21206. Found: 528.21124.

3.4.9. (1S,2R,3R,5S)-2-((4-(((5-Fluoro-2-((4-morpholinophenyl)amino)pyrimidin-4-yl)amino)methyl)-1H-1,2,3-triazol-1-yl)methyl)-6,6-dimethylbicyclo[3.1.1]heptane-2,3-diol (+)-28

The reaction was accomplished starting from (+)-2 and alkyne 11 in tert-BuOH/H2O (2:1) according to the general procedure. The product was purified by column chromatography on silica gel with DCM/MeOH (19:1). Yield: 0.093, 73%; yellowish-white crystals; m.p.: 125–128 °C; α D 20 = +6.0 (c 0.1 MeOH); 1H NMR (500 MHz, CDCl3) δ (ppm): 1.09 (3H, s), 1.29 (3H, s), 1.40 (1H, d, J = 10.6 Hz), 1.64 (1H, dd, J = 5.0, 13.9 Hz), 1.98–1.91 (2H, m), 2.27–2.20 (1H, m), 2.54–2.46 (1H, m), 3.10 (4H, dd, J = 4.7, 4.7 Hz), 3.85 (4H, dd, J = 4.6, 4.6 Hz), 4.11 (1H, d, J = 13.8 Hz), 4.41 (1H, dd, J = 5.2, 9.6 Hz), 4.47 (1H, d, J = 13.8 Hz), 4.71 (2H, ddt, J = 5.9, 13.2, 12.2 Hz), 5.54 (1H, t, J = 5.7 Hz), 6.82 (1H, s), 6.88 (2H, d, J = 8.7 Hz), 7.43 (2H, d, J = 8.7 Hz), 7.59 (1H, s), 7.76 (1H, d, J = 2.4 Hz); 13C NMR (125 MHz, CDCl3) δ (ppm): 24.3, 27.6, 27.7, 36.1, 37.5, 38.6, 40.5, 50.2 (2C), 50.4, 59.9, 65.1, 67.0 (2C), 74.5, 116.6 (2C), 121.1 (2C), 124.8, 133.1, 139.3, 144.3, 147.0, 152.2, 152.3, 156.2; 19F NMR (470 MHz, CDCl3) δ (ppm): −169.35; HRMS (ESI): m/z calcd. for C27H36FN8O3 [M + H]+: 539.28944. Found: 539.28715.
The (1R,2S,3S,5R)-enantiomer (−)-28 was synthesized analogously to (+)-28; α D 20 = −8.0 (c 0.1, MeOH); all the spectroscopic data and the m.p. were like those for the (1R,2S,3S,5R)-enantiomer. HRMS (ESI): m/z calcd. for C27H36FN8O3 [M + H]+: 539.28944. Found: 539.28820.

3.4.10. (1S,2R,3R,5S)-2-((4-((6-Chloro-9H-purin-9-yl)methyl)-1H-1,2,3-triazol-1-yl)methyl)-6,6-dimethylbicyclo[3.1.1]heptane-2,3-diol (+)-29

The reaction was accomplished starting from (+)-2 and alkyne 15 in THF/H2O (2:1) according to the general procedure. The product was purified by column chromatography on silica gel with CHCl3/MeOH (19:1) increased to (9:1). Yield: 0.090 g, 94%; white crystals; m.p.: 120–123 °C; α D 20 = +3 (c 0.1, MeOH); 1H NMR (500 MHz, CDCl3) δ (ppm): 1.09 (3H, s), 1.30 (3H, s), 1.39 (1H, d, J = 10.6 Hz), 1.68–1.62 (1H, m), 1.99–1.92 (2H, m), 2.29–2.22 (1H, m), 2.55–2.48 (1H, m), 2.63 (1H, broad s), 3.98 (1H, s), 4.18 (1H, d, J = 13.8 Hz), 4.39–4.34 (1H, m), 4.53 (1H, d, J = 13.8 Hz), 5.56 (2H, q, J = 12.4 Hz), 7.91 (1H, s), 8.30 (1H, s), 8.75 (1H, s); 13C NMR (125 MHz, CDCl3) δ (ppm): 24.2, 27.5, 27.6, 37.6, 38.6, 39.2, 40.4, 50.1, 60.0, 64.9, 74.5, 125.6, 131.5, 140.6, 145.2, 151.2, 151.5, 152.1; HRMS (ESI): m/z calcd. for C18H23ClN7O2 [M + H]+: 404.16018. Found: 404.15907.
The (1R,2S,3S,5R)-enantiomer (−)-29 was synthesized analogously to (+)-29; α D 20 = −6.0 (c 0.1, MeOH); all the spectroscopic data and the m.p. were similar to those for the (1R,2S,3S,5R)-enantiomer. HRMS (ESI): m/z calcd. for C18H23ClN7O2 [M + H]+: 404.16018. Found: 404.15909.

3.4.11. (1S,2R,3R,5S)-2-((4-((6-Chloro-8-phenyl-9H-purin-9-yl)methyl)-1H-1,2,3-triazol-1-yl)methyl)-6,6-dimethylbicyclo[3.1.1]heptane-2,3-diol (+)-30

The reaction was accomplished starting from (+)-2 and alkyne 16 in THF/H2O (2:1) according to the general procedure. The product was purified by column chromatography on silica gel with toluene/EtOH (9:1).Yield: 0.112 g, 98%; white crystals; m.p.: 159–162 °C; α D 20 = +5.5 (c 0.1, MeOH); 1H NMR (500 MHz, CDCl3) δ (ppm): 1.08 (3H, s), 1.29 (3H, s), 1.39 (1H, d, J = 10.6 Hz), 1.63 (1H, dd, J = 4.9, 14.2 Hz), 1.99–1.91 (2H, m), 2.27–2.21 (1H, m), 2.44 (1H, d, J = 5.7 Hz), 2.53–2.46 (1H, m), 3.90 (1H, s), 4.15 (1H, d, J = 13.9 Hz), 4.36 (1H, ddt, J = 5.0, 5.0, 4.9 Hz), 4.50 (1H, d, J = 13.9 Hz), 5.60 (2H, q, J = 14.2 Hz), 7.61–7.57 (3H, m), 7.89 (1H, s), 8.06–8.02 (2H, m), 8.73 (1H, s); 13C NMR (125 MHz, CDCl3) δ (ppm): 24.2, 27.5, 27.6, 37.4, 38.6, 39.9, 40.4, 50.2, 59.9, 65.0, 74.5, 126.2, 128.2, 129.1 (2C), 130.1 (2C), 131.3, 131.6, 141.7, 150.2, 151.5, 153.5, 156.2; HRMS (ESI): m/z calcd. for C24H27ClN7O2 [M + H]+: 480.19147. Found: 480.19051.
The (1R,2S,3S,5R)-enantiomer (−)-30 was synthesized analogously to (+)-30; α D 20 = −5.0 (c 0.1, MeOH); all the spectroscopic data and the m.p. were similar to those for the (1R,2S,3S,5R)-enantiomer. HRMS (ESI): m/z calcd. for C24H27ClN7O2 [M + H]+: 480.19147. Found: 480.19019.

3.4.12. (1S,2R,3R,5S)-6,6-Dimethyl-2-((4-((6-phenyl-9H-purin-9-yl)methyl)-1H-1,2,3-triazol-1-yl)methyl)bicyclo[3.1.1]heptane-2,3-diol (+)-31

The reaction was accomplished starting from (+)-2 and alkyne 17 in THF/H2O (2:1) according to the general procedure. The product was purified by column chromatography on silica gel with DCM/MeOH (19:1). Yield: 0.080 g, 76%; white crystals; m.p.: 120–122 °C; α D 20 = +6.5 (c 0.1, MeOH); 1H NMR (500 MHz, CDCl3) δ (ppm): 1.09 (3H, s), 1.29 (3H, s), 1.39 (1H, d, J = 10.6 Hz), 1.65 (1H, d, J = 5.3 Hz), 1.95 (2H, d, J = 6.0 Hz), 2.28–2.21 (1H, m), 2.54–2.47 (1H, m), 2.72 (1H, d, J = 5.7 Hz), 4.01 (1H, s), 4.17 (1H, d, J = 13.9 Hz), 4.36 (1H, ddt, J = 5.0, 5.0, 4.9 Hz), 4.52 (1H, d, J = 13.9 Hz), 5.58 (2H, q, J = 13.9 Hz), 7.58–7.50 (3H, m), 7.91 (1H, s), 8.29 (1H, s), 8.74 (2H, dd, J = 1.5, 8.2 Hz), 9.00 (1H, s); 13C NMR (125 MHz, CDCl3) δ (ppm): 24.2, 27.5, 27.6, 37.5, 38.6, 38.9, 40.4, 50.2, 59.9, 64.9, 74.5, 125.6, 128.7 (2C), 129.8 (2C), 131.1, 135.5, 141.3, 144.3, 152.1 (2C), 152.4, 155.1; HRMS (ESI): m/z calcd. for C24H28N7O2 [M + H]+: 446.23044. Found: 446.22916.
The (1R,2S,3S,5R)-enantiomer (−)-31 was synthesized analogously to (+)-31; α D 20 = −6.5 (c 0.1, MeOH); all the spectroscopic data and the m.p. were similar to those for the (1R,2S,3S,5R)-enantiomer. HRMS (ESI): m/z calcd. for C24H28N7O2 [M + H]+: 446.23044. Found: 446.22935.

3.4.13. (1S,2R,3R,5S)-2-((4-((6-(Benzylamino)-9H-purin-9-yl)methyl)-1H-1,2,3-triazol-1-yl)methyl)-6,6-dimethylbicyclo[3.1.1]heptane-2,3-diol (+)-32

The reaction was accomplished starting from (+)-2 and alkyne 18 in THF/H2O (2:1) according to the general procedure. The product was purified by column chromatography on silica gel with DCM/MeOH (19:1). Yield: 0.106 g, 94%; white crystals; m.p.: 122–125 °C; α D 20 = +9.0 (c 0.1, MeOH); 1H NMR (500 MHz, CDCl3) δ (ppm): 1.01 (3H, s), 1.10–1.04 (1H, m), 1.19 (1H, d, J = 10.5 Hz), 1.26 (3H, s), 1.75–1.70 (1H, m), 1.99 (1H, t, J = 5.7 Hz), 2.18–2.09 (2H, m), 4.05 (1H, d, J = 13.6 Hz), 4.15–4.08 (1H, m), 4.41 (1H, s), 4.57 (1H, d, J = 13.6 Hz), 4.85 (1H, d, J = 12.6 Hz), 5.42 (2H, s), 6.27 (1H, s), 7.37–7.26 (5H, m), 7.95 (1H, s), 8.24 (1H, s), 8.37 (1H, s); 13C NMR (125 MHz, CDCl3) δ (ppm): 24.2, 27.5, 27.6, 37.4, 38.4, 39.2, 40.4, 44.9, 50.3, 60.2, 63.0, 74.3, 118.9, 126.8, 127.6, 128.2, 128.6 (3C), 137.9, 139.6, 141.1, 148.4, 153.0, 153.9; HRMS (ESI): m/z calcd. for C25H31ClN8O2 [M + H]+: 475.25699. Found: 475.25567.
The (1R,2S,3S,5R)-enantiomer (−)-32 was synthesized analogously to (+)-32; α D 20 = −9.0 (c 0.1, MeOH); all the spectroscopic data and the m.p. were similar to those for the (1R,2S,3S,5R)-enantiomer. HRMS (ESI): m/z calcd. for C25H31ClN8O2 [M + H]+: 475.25699. Found: 475.25576.

3.4.14. (1S,2R,3R,5S)-6,6-Dimethyl-2-((4-((6-((4-(trifluoromethyl)phenyl)amino)-9H-purin-9-yl)methyl)-1H-1,2,3-triazol-1-yl)methyl)bicyclo[3.1.1]heptane-2,3-diol (+)-33

The reaction was accomplished starting from (+)-2 and alkyne 19 in THF/H2O (2:1) according to the general procedure. The product was purified by column chromatography on silica gel with DCM/MeOH (19:1). Yield: 0.125 g, 99%; white crystals; m.p.: 139–141 °C; α D 20 = +6.5 (c 0.1, MeOH); 1H NMR (500 MHz, CDCl3) δ (ppm): 1.04 (3H, s), 1.30 (3H, s), 1.36–1.31 (1H, m), 1.48 (1H, d, J = 10.4 Hz), 1.83–1.78 (1H, m), 2.23–2.13 (2H, m), 2.33–2.26 (1H, m), 4.13 (1H, d, J = 13.5 Hz), 4.23–4.17 (1H, m), 4.58 (1H, s), 4.69 (1H, d, J = 13.5 Hz), 5.53 (2H, s), 6.22 (1H, d, J = 6.5 Hz), 7.32 (2H, d, J = 8.6 Hz), 7.52 (2H, d, J = 8.6 Hz), 8.06 (1H, s), 8.23 (1H, s), 8.48 (1H, s), 8.49 (1H, s); 13C NMR (125 MHz, CDCl3) δ (ppm): 24.3, 27.6, 28.0, 37.9, 38.4, 39.6, 40.5, 50.3, 60.5, 62.9, 74.5, 118.6 (2C), 119.2, 124.0 (q, J = 32.6 Hz), 125.8 (2C, q, J = 3.8 Hz), 127.5, 127.7, 140.6, 140.7, 141.8, 149.0, 150.8, 152.1; 19F NMR (470 MHz, CDCl3) δ (ppm): −61.98; HRMS (ESI): m/z calcd. for C25H28F3N8O2 [M + H]+: 529.22873. Found: 529.22747.
The (1R,2S,3S,5R)-enantiomer (−)-33 was synthesized analogously to (+)-33; α D 20 = −4.0 (c 0.1, MeOH); all the spectroscopic data and the m.p. were similar to those for the (1R,2S,3S,5R)-enantiomer. HRMS (ESI): m/z calcd. for C25H28F3N8O2 [M + H]+: 529.22873. Found: 529.22742.

3.5. Determination of Antiproliferative Effect

The growth-inhibitory effects of the isosteviol-based 1,3-aminoalcohols were determined by a standard MTT (3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide) assay on a panel containing five cell lines, including Hela and SiHa (cervical cancer), MDA-MB-231 and MCF-7 (breast cancers), and A2780 ovarian cancer cells. NIH/3T3 fibroblasts were additionally used to obtain preliminary information on the cancer selectivity. All cell lines were purchased from the European Collection of Cell Cultures (Salisbury, UK), except for the SiHa (American Tissue Culture Collection (ATCC, Manassas, VA, USA). The cells were maintained in a minimal essential medium supplemented with 10% fetal bovine serum, 1% non-essential amino acids, and 1% penicillin–streptomycin at 37 °C in a humidified atmosphere containing 5% CO2. All media and supplements were obtained from Lonza Group Ltd. (Basel, Switzerland). Cancer cells were seeded into 96-well plates (5000 cells/well); after overnight incubation, the test compound was added in two different concentrations (10 µM and 30 µM) and incubated for another 72 h under cell-culturing conditions. In the next step, 20 μL of 5 mg/mL MTT solution was added to each well and incubated for a further 4 h. The medium was removed, and the precipitated formazan crystals were dissolved in DMSO after 60 min of shaking at 37 °C. As a final step, the absorbance was measured at 545 nm by using a microplate reader. Untreated cells were included as controls. In the case of the most effective compounds, the assays were repeated with a set of dilutions (1.0–30 μM) to determine the IC50 values. Two independent experiments were performed with five wells for each condition. Calculations were performed using the GraphPad Prism 10.0 software (GraphPad Software Inc., San Diego, CA, USA).

3.6. Antimicrobial Analyses

The synthesized compounds were dissolved in MeOH and diluted with H2O to reach concentration levels of up to 100 µg/mL and 10 µg/mL with a final MeOH content of 10%. The completeness of the dissolution was observed visually in each case. Then, the resulting solutions were investigated in a microdilution assay against bacterial and yeast strains, including Bacillus subtilis SZMC 0209 (Gram-positive), Staphylococcus aureus SZMC 14611 (Gram-positive), Escherichia coli SZMC 6271 (Gram-negative), and Pseudomonas aeruginosa SZMC 0177 (Gram-negative) as well as Candida albicans SZMC 1533 and Candida krusei SZMC 1352, respectively, according to the M07-A10 CLSI guideline and our previous work [4]. The microbe suspensions were prepared from fresh cultures cultivated overnight in different fermentation broths for both bacteria (10 g/L peptone, 5 g/L NaCl, 5 g/L yeast extract) and yeast (20 g/L peptone, 10 g/L yeast extract, 20 g/L glucose) at 37 °C, and their concentrations were set to 4 × 105 cells/mL with sterile media. After that, 100 μL of suspension containing the bacterial or yeast cells and 50 μL of sterile broth as well as 50 μL of the test solutions were placed into the wells of 96-well plates and incubated for 24 h at 37 °C and 32 °C for bacteria and yeasts, respectively. The blank sample was a mixture of 150 μL broth and 50 μL of a test solution used for background correction, while 100 μL of microbial suspension supplemented with 50 μL of sterile broth and 50 μL of 10% MeOH was applied as a negative control. For the positive control, ampicillin or nystatin was applied for bacteria or fungi, respectively, at two final concentration levels (100 µg/mL and 10 µg/mL). The inhibitory effects were calculated as the percentage of the negative control after blank correction using the following formula: inhibitory effect (%) = 100 − ((ODtest − ODblank)/ODnegative) × 100)). Here, ODtest represents the OD values of the test solution, ODnegative represents the OD values of the negative control, and ODblank represents the OD values of blank samples.

4. Conclusions

Key intermediate azidodiols, synthesized according to literature from commercially available (+)- and (−)-α-pinene in a four-step sequence, were reacted with pyrimidine- and purine-based propargyl-type alkynes in a click reaction carried out in the presence of Cu(OAc)2 and Na-ascorbate. The resulting methylene-1,2,3-triazolo-linked nucleoside analogues were screened for antiproliferative activity by MTT assay on HeLa, MD231, SiHa, MCF-7 and A2780 human cancer cell lines compared with fibroblast cells (NIH/3T3). From weak to moderate antiproliferative activities were observed and the SAR determination showed that, meanwhile, the stereochemistry had a minimal but consequent effect on activity. The diaminopyrimidine ring with 5-fluoro, 5-chloro, and 2-(4-trifluoromethyl)phenyl substituents proved essential for moderate activity with selectivity on HeLa and MCF-7 cell lines.
All prepared compounds were tested on Gram-positive and Gram-negative pathogenic bacteria and two yeasts, and the SAR was explained in detail. The starting azidodiols had excellent and selective antibacterial activities against S. aureus and P. aeruginosa with moderate antifungal effect on C. krusei. Pyrimidines and purine derivatives also expressed a moderate antifungal effect against C. krusei. One of the purine-based derivatives possessed a remarkable and selective antibacterial effect against P. aeruginosa.
By comparing the two series of enantiomers, it can be clearly stated that compounds derived from (+)-α-pinene have a more pronounced growth-inhibitory effect on HeLa and A2780 cell lines, whereas compounds derived from (−)-α-pinene inhibit the growth of the MCF7 cell line more effectively. In case of antibacterial and antifungal activities, there are no significant differences between the enantiomeric series.
The purine-coupled monoterpene derivatives seem to be an interesting group to extend the study of the substituent effect and the modified purine structure to obtain more effective antifungal and antibacterial compounds. The antibacterial effect experienced in the case of azidodiol compounds makes it interesting and necessary to investigate additional terpene-based azidodiols in order to discover potential antibiotics.

Supplementary Materials

The following supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/ijms262311705/s1.

Author Contributions

Z.S., A.S. and I.Z. conceived and designed the experiments; K.T. (microbiological assay), A.K. (MTT assay) and D.D. (chemistry) performed the experiments, analyzed the data and wrote the experimental part; Z.S., A.S. and I.Z. discussed the results and contributed to write the paper. The authors have reviewed and edited the output and take full responsibility for the content of this publication. All authors have read and agreed to the published version of the manuscript.

Funding

We are grateful for financial support from the Hungarian Research Foundation (NKFI K138871). Project no. TKP2021-EGA-32 has been implemented with the support provided by the Ministry of Innovation and Technology of Hungary from the National Research, Development and Innovation Fund, financed under the TKP2021-EGA funding scheme.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

The original contributions presented in this study are included in the article/Supplementary Materials. Further inquiries can be directed to the corresponding author.

Acknowledgments

We are grateful for financial support from the Hungarian Research Foundation, and the Ministry of Innovation and Technology of Hungary. The high-resolution mass spectrometric analysis was performed by Robert Berkecz.

Conflicts of Interest

The authors declare no conflicts of interest. The funders had no role in the design of the study; in the collection, analyses, or interpretation of data; in the writing of the manuscript; or in the decision to publish the results.

Abbreviations

The following abbreviations are used in this manuscript:
THFTetrahydrofurane
DCMDichlormethane
mCPBAmeta-Chloroperbenzoic acid
MWMicrowave
TEOFTriethyl orthoformate
TEATriethylamine
CuAACCopper-catalyzed azide–alkyne cycloaddition

References

  1. Antimicrobial Resistance. Available online: https://www.who.int/news-room/fact-sheets/detail/antimicrobial-resistance (accessed on 6 October 2025).
  2. Global Cancer Burden Growing, Amidst Mounting Need for Services. Available online: https://www.who.int/news/item/01-02-2024-global-cancer-burden-growing--amidst-mounting-need-for-services (accessed on 6 October 2025).
  3. Bray, F.; Laversanne, M.; Sung, H.; Ferlay, J.; Siegel, R.L.; Soerjomataram, I.; Jemal, A. Global cancer statistics 2022: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J. Clin. 2024, 74, 229–263. [Google Scholar] [CrossRef]
  4. Raji, M.; Le, T.M.; Huynh, T.; Szekeres, A.; Nagy, V.; Zupkó, I.; Szakonyi, Z. Divergent Synthesis, Antiproliferative and Antimicrobial Studies of 1,3-Aminoalcohol and 3-Amino-1,2-Diol Based Diaminopyrimidines. Chem. Biodivers. 2022, 19, e202200077. [Google Scholar] [CrossRef]
  5. Nyamwihura, R.J.; Ogungbe, I.V. The pinene scaffold: Its occurrence, chemistry, synthetic utility, and pharmacological importance. RSC Adv. 2022, 12, 11346–11375. [Google Scholar] [CrossRef] [PubMed]
  6. Gonda, T.; Balázs, A.; Tóth, G.; Fülöp, F.; Szakonyi, Z. Stereoselective synthesis and transformations of pinane-based 1,3-diaminoalcohols. Tetrahedron 2017, 73, 2638–2648. [Google Scholar] [CrossRef]
  7. Csillag, K.; Németh, L.; Martinek, T.A.; Szakonyi, Z.; Fülöp, F. Stereoselective synthesis of pinane-type tridentate aminodiols and their application in the enantioselective addition of diethylzinc to benzaldehyde. Tetrahedron Asymmetry 2012, 23, 144–150. [Google Scholar] [CrossRef]
  8. Dhar, P.; Chan, P.; Cohen, D.T.; Khawam, F.; Gibbons, S.; Snyder-Leiby, T.; Dickstein, E.; Rai, P.K.; Watal, G. Synthesis, Antimicrobial Evaluation, and Structure–Activity Relationship of α-Pinene Derivatives. J. Agric. Food Chem. 2014, 62, 3548–3552. [Google Scholar] [CrossRef] [PubMed]
  9. Sanglikar, G.; Kumar Tengli, A. A critical review on purine and pyrimidine heterocyclic derivatives and their designed molecules in Cancer. Results Chem. 2025, 15, 102210. [Google Scholar] [CrossRef]
  10. Kerru, N.; Gummidi, L.; Maddila, S.; Gangu, K.K.; Jonnalagadda, S.B. A Review on Recent Advances in Nitrogen-Containing Molecules and Their Biological Applications. Molecules 2020, 25, 1909. [Google Scholar] [CrossRef] [PubMed]
  11. Lawrence, H.R.; Martin, M.P.; Luo, Y.; Pireddu, R.; Yang, H.; Gevariya, H.; Ozcan, S.; Zhu, J.-Y.; Kendig, R.; Rodriguez, M.; et al. Development of o-Chlorophenyl Substituted Pyrimidines as Exceptionally Potent Aurora Kinase Inhibitors. J. Med. Chem. 2012, 55, 7392–7416. [Google Scholar] [CrossRef]
  12. Tomović Pavlović, K.; Kocić, G.; Šmelcerović, A. Myt1 kinase inhibitors—Insight into structural features, offering potential frameworks. Chem. Biol. Interact. 2024, 391, 110901. [Google Scholar] [CrossRef]
  13. Jadhav, M.; Sankhe, K.; Bhandare, R.R.; Edis, Z.; Bloukh, S.H.; Khan, T.A. Synthetic Strategies of Pyrimidine-Based Scaffolds as Aurora Kinase and Polo-like Kinase Inhibitors. Molecules 2021, 26, 5170. [Google Scholar] [CrossRef] [PubMed]
  14. Bibi, M.; Qureshi, N.A.; Sadiq, A.; Farooq, U.; Hassan, A.; Shaheen, N.; Asghar, I.; Umer, D.; Ullah, A.; Khan, F.A.; et al. Exploring the ability of dihydropyrimidine-5-carboxamide and 5-benzyl-2,4-diaminopyrimidine-based analogues for the selective inhibition of L. major dihydrofolate reductase. Eur. J. Med. Chem. 2021, 210, 112986. [Google Scholar] [CrossRef] [PubMed]
  15. Denny, W.A. Inhibitors of F1F0-ATP synthase enzymes for the treatment of tuberculosis and cancer. Future Med. Chem. 2021, 13, 911–926. [Google Scholar] [CrossRef]
  16. Nammalwar, B.; Bunce, R.A. Recent Advances in Pyrimidine-Based Drugs. Pharmaceuticals 2024, 17, 104. [Google Scholar] [CrossRef]
  17. Das, S.K.; Behera, B.; Purohit, C.S. Scaffolds of purine privilege for biological cytotoxic targets: A review. Pharm. Chem. J. 2023, 57, 777–792. [Google Scholar] [CrossRef]
  18. Wimmer, Z. Purine Scaffold in Agents for Cancer Treatment. ACS Omega 2025, 10, 18170–18183. [Google Scholar] [CrossRef]
  19. Tunçbilek, M.; Ateş-Alagöz, Z.; Altanlar, N.; Karayel, A.; Özbey, S. Synthesis and antimicrobial evaluation of some new substituted purine derivatives. Bioorganic Med. Chem. 2009, 17, 1693–1700. [Google Scholar] [CrossRef] [PubMed]
  20. Depp, D.; Sebők, N.R.; Szekeres, A.; Szakonyi, Z. Stereoselective Synthesis and Antimicrobial Studies of Allo-Gibberic Acid-Based 2,4-Diaminopyrimidine Chimeras. Pharmaceuticals 2025, 18, 168. [Google Scholar] [CrossRef] [PubMed]
  21. Szakonyi, Z.; Fülöp, F. Carbocyclic nucleosides from enantiomeric, α-pinane-based aminodiols. Tetrahedron Asymmetry 2010, 21, 831–836. [Google Scholar] [CrossRef]
  22. Szakonyi, Z.; Hetényi, A.; Fülöp, F. Synthesis and application of monoterpene-based chiral aminodiols. Tetrahedron 2008, 64, 1034–1039. [Google Scholar] [CrossRef]
  23. Wang, W.; Li, Z.; Gao, W.; Liu, X.; Lv, Y.; Hao, Z.; Tang, L.; Li, K.; Zhao, B.; Fan, Z. Design, Synthesis, and Evaluation of Novel Isothiazole-Purines as a Pyruvate Kinase-Based Fungicidal Lead Compound. J. Agric. Food Chem. 2021, 69, 9461–9471. [Google Scholar] [CrossRef]
  24. Huang, K.-X.; Xie, M.-S.; Zhao, G.-F.; Qu, G.-R.; Guo, H.-M. Synthesis of Chiral Cyclopropyl Carbocyclic Purine Nucleosides via Asymmetric Intramolecular Cyclopropanations Catalyzed by a Chiral Ruthenium(II) Complex. Adv. Synth. Catal. 2016, 358, 3627–3632. [Google Scholar] [CrossRef]
  25. Khdar, Z.A.; Le, T.M.; Schelz, Z.; Zupkó, I.; Szakonyi, Z. Aminodiols, aminotetraols and 1,2,3-triazoles based on allo-gibberic acid: Stereoselective syntheses and antiproliferative activities. RSC Adv. 2024, 14, 36698–36712. [Google Scholar] [CrossRef] [PubMed]
  26. Bauer, D.; Cornejo, M.A.; Hoang, T.T.; Lewis, J.S.; Zeglis, B.M. Click Chemistry and Radiochemistry: An Update. Bioconjugate Chem. 2023, 34, 1925–1950. [Google Scholar] [CrossRef] [PubMed]
  27. Xiao, Z.; Gu, Y.; Dong, H.; Liu, B.; Jin, W.; Li, J.; Ma, P.; Xu, H.; Hou, W. Strategic application of CuAAC click chemistry in the modification of natural products for anticancer activity. Eur. J. Med. Chem. Rep. 2023, 9, 100113. [Google Scholar] [CrossRef]
  28. Schütz, C.; Ho, D.-K.; Hamed, M.M.; Abdelsamie, A.S.; Röhrig, T.; Herr, C.; Kany, A.M.; Rox, K.; Schmelz, S.; Siebenbürger, L.; et al. New PqsR Inverse Agonist Potentiates Tobramycin Efficacy to Eradicate Pseudomonas aeruginosa Biofilms. Adv. Sci. 2021, 8, 2004369. [Google Scholar] [CrossRef]
  29. Khandelwal, R.; Vasava, M.; Abhirami, R.B.; Karsharma, M. Recent advances in triazole synthesis via click chemistry and their pharmacological applications: A review. Bioorganic Med. Chem. Lett. 2024, 112, 129927. [Google Scholar] [CrossRef] [PubMed]
  30. Zhang, R.; Qin, X.; Kong, F.; Chen, P.; Pan, G. Improving cellular uptake of therapeutic entities through interaction with components of cell membrane. Drug Deliv. 2019, 26, 328–342. [Google Scholar] [CrossRef] [PubMed]
  31. Madikonda, A.K.; Ajayakumar, A.; Nadendla, S.; Banothu, J.; Muripiti, V. Esterase-responsive nanoparticles (ERN): A targeted approach for drug/gene delivery exploits. Bioorganic Med. Chem. 2024, 116, 118001. [Google Scholar] [CrossRef]
  32. De O. Torres, N.M.P.; Cardoso, G.D.A.; Silva, H.; de Freitas, R.P.; Alves, R.B. New purine-triazole hybrids as potential anti-breast cancer agents: Synthesis, antiproliferative activity, and ADMET in silico study. Med. Chem. Res. 2023, 32, 1816–1831. [Google Scholar]
  33. Burcevs, A.; Sebris, A.; Traskovskis, K.; Chu, H.-W.; Chang, H.-T.; Jovaišaitė, J.; Juršėnas, S.; Turks, M.; Novosjolova, I. Synthesis of Fluorescent C–C Bonded Triazole-Purine Conjugates. J. Fluoresc. 2024, 34, 1091–1097. [Google Scholar] [CrossRef] [PubMed]
  34. Gupta, Y.K.; Gupta, V.; Singh, S. Synthesis, characterization and antimicrobial activity of pyrimidine based derivatives. J. Pharm. Res. 2013, 7, 491–495. [Google Scholar] [CrossRef]
  35. Shaheen, F.; Safa, Y.; Sultan, M.; Bibi, I.; Rehman, F.; Shah, S.I. Synthesis, characterization and biological activity of azides and its derivatives. Desalination Water Treat. 2022, 280, 330–337. [Google Scholar] [CrossRef]
  36. Singh, G.; Radha, A.; Kaur, H.; Malik, S.; Stanzin, J.; Komal; Singh, B.; Baliyan, D.; Singh, K.N. Versatile Benzophenone Derivatives: Broad Spectrum Antibiotics, Antioxidants, Anticancer Agents and Fe(III) Chemosensor. Appl. Organomet. Chem. 2024, 39, e7964. [Google Scholar] [CrossRef]
  37. Wilde, M.I.; Langtry, H.D. Zidovudine: An update of its pharmacodynamic and pharmacokinetic properties, and therapeutic efficacy. Drugs 1993, 46, 515–578, Erratum in Drugs 1993, 46, 1080. [Google Scholar] [CrossRef] [PubMed]
  38. Yıldırım, M.; Aksakal, E.; Özgeriş, F.; Ozgeris, B.; Gormez, A. Biological Evaluation of Azide Derivative as Antibacterial and Anticancer Agents. Nat. Prod. Biotechnol. 2022, 2, 105–113. [Google Scholar] [CrossRef]
Scheme 1. Synthesis of pinane-based azidodiols. (i) mCPBA, NaHCO3, DCM, 10 °C, 1.5 h, 82%; (ii) Al(O-iPr)3, dry toluene, reflux, 3 h, 70%; (iii) mCPBA, 5% NaH2PO4, 0 °C, 2 h, 60%; (iv) NaN3, 20% NH4Cl, EtOH/H2O, reflux, 36 h, 80%.
Scheme 1. Synthesis of pinane-based azidodiols. (i) mCPBA, NaHCO3, DCM, 10 °C, 1.5 h, 82%; (ii) Al(O-iPr)3, dry toluene, reflux, 3 h, 70%; (iii) mCPBA, 5% NaH2PO4, 0 °C, 2 h, 60%; (iv) NaN3, 20% NH4Cl, EtOH/H2O, reflux, 36 h, 80%.
Ijms 26 11705 sch001
Scheme 2. Synthesis of 2,4-diaminopyrimidine-derived alkynes 712. (i) propargylamine (2 eq.), TEA (1 eq.), MeCN, 25 °C, 24 h for 5 and 6; (ii) (a) 4-trifluoromethylaniline (1.1 eq.), EtOH, MW, 150 °C, 200 W, 19 bar, 2 h for 7 and 8; (b) 1-methyl-1H-pyrazol-4-amine (1.1 eq.), EtOH, MW, 150 °C, 200 W, 19 bar, 2 h for 9 and 10; (c) 4-morpholinoaniline (1 eq.), EtOH, MW, 100 °C, 200 W, 17 bar, 2 h for 11; (d) methyl 4-aminobenzoate (1 eq.), EtOH, MW, 100 °C, 200 W, 17 bar, 2 h for 12.
Scheme 2. Synthesis of 2,4-diaminopyrimidine-derived alkynes 712. (i) propargylamine (2 eq.), TEA (1 eq.), MeCN, 25 °C, 24 h for 5 and 6; (ii) (a) 4-trifluoromethylaniline (1.1 eq.), EtOH, MW, 150 °C, 200 W, 19 bar, 2 h for 7 and 8; (b) 1-methyl-1H-pyrazol-4-amine (1.1 eq.), EtOH, MW, 150 °C, 200 W, 19 bar, 2 h for 9 and 10; (c) 4-morpholinoaniline (1 eq.), EtOH, MW, 100 °C, 200 W, 17 bar, 2 h for 11; (d) methyl 4-aminobenzoate (1 eq.), EtOH, MW, 100 °C, 200 W, 17 bar, 2 h for 12.
Ijms 26 11705 sch002
Scheme 3. Synthesis of purine-derived alkynes 1519. (i) propargylamine (2 eq.), TEA (1 eq.), reflux, 24 h for 14; (ii) TEOF (39 eq.), HCl (conc.) (3.5 eq.), RT, 24 h for 15; (iii) benzoyl chloride (1 eq.), NH4Cl (6 eq.), POCl3 (75 eq.), toluene, 100 °C, 24 h for 16; (iv) PhB(OH)2 (1.2 eq.), Pd(PPh3)4 (0.05 eq.), K2CO3 (2.2 eq.), dry toluene, argon atm., 100 °C, 12 h for 17; (v) benzylamine (2 eq.), TEA (1.5 eq), MeCN, 40 °C, 24 h for 18; (vi) 4-trifluoromethylaniline (1.5 eq.), 2 drops HCl (conc.), iPrOH, 85 °C, 4 h for 19.
Scheme 3. Synthesis of purine-derived alkynes 1519. (i) propargylamine (2 eq.), TEA (1 eq.), reflux, 24 h for 14; (ii) TEOF (39 eq.), HCl (conc.) (3.5 eq.), RT, 24 h for 15; (iii) benzoyl chloride (1 eq.), NH4Cl (6 eq.), POCl3 (75 eq.), toluene, 100 °C, 24 h for 16; (iv) PhB(OH)2 (1.2 eq.), Pd(PPh3)4 (0.05 eq.), K2CO3 (2.2 eq.), dry toluene, argon atm., 100 °C, 12 h for 17; (v) benzylamine (2 eq.), TEA (1.5 eq), MeCN, 40 °C, 24 h for 18; (vi) 4-trifluoromethylaniline (1.5 eq.), 2 drops HCl (conc.), iPrOH, 85 °C, 4 h for 19.
Ijms 26 11705 sch003
Scheme 4. Synthesis of pinane-based hybrid compounds 20–33. (i) Cu(OAc)2·H2O (0.05 eq.), Na ascorbate (0.1 eq.), alkyne (1.1 eq.), 45 °C, 48 h; tert-BuOH/H2O (2:1) for (+)-20, (+)-21, (+)-2326 and (+)-28 or THF/H2O (2:1) for (+)-22, (+)-27 and (+)-2933.
Scheme 4. Synthesis of pinane-based hybrid compounds 20–33. (i) Cu(OAc)2·H2O (0.05 eq.), Na ascorbate (0.1 eq.), alkyne (1.1 eq.), 45 °C, 48 h; tert-BuOH/H2O (2:1) for (+)-20, (+)-21, (+)-2326 and (+)-28 or THF/H2O (2:1) for (+)-22, (+)-27 and (+)-2933.
Ijms 26 11705 sch004
Figure 1. Selected antiproliferative properties of the prepared pyrimidine and purine derivatives against cancer cells and NIH/3T3 fibroblasts. Numeric values are given in Table S1 in the Supplementary Materials. *: data from reference [24].
Figure 1. Selected antiproliferative properties of the prepared pyrimidine and purine derivatives against cancer cells and NIH/3T3 fibroblasts. Numeric values are given in Table S1 in the Supplementary Materials. *: data from reference [24].
Ijms 26 11705 g001
Figure 2. Selected antimicrobial properties of the prepared pyrimidine and purine derivatives against both Gram-negative and Gram-positive bacteria as well as yeast strains. Numeric values are given in Table 2 and Table S2 in the Supplementary Materials.
Figure 2. Selected antimicrobial properties of the prepared pyrimidine and purine derivatives against both Gram-negative and Gram-positive bacteria as well as yeast strains. Numeric values are given in Table 2 and Table S2 in the Supplementary Materials.
Ijms 26 11705 g002
Table 1. Selected data and IC50 values of the most active derivatives (+)-23, (−)-23, (+)-24 and (−)-24.
Table 1. Selected data and IC50 values of the most active derivatives (+)-23, (−)-23, (+)-24 and (−)-24.
Comp.Conc.
(μM)
Growth Inhibition (%) ± SEM 1,2
Calculated IC50 (μM)
HeLaSiHaMCF-7MDA-MB-231A2780NIH/3T3
(+)-231066.04 ± 1.2721.67 ± 2.4530.45 ± 1.86
3089.81 ± 1.3442.81 ± 2.0384.53 ± 1.2520.30 ± 1.8980.44 ± 1.5748.65 ± 2.058
IC507.54>3018.59>3014.79>30
(−)-231023.39 ± 2.3928.48 ± 2.4210.18 ± 1.9425.34 ± 1.7740.61 ± 2.06
3096.09 ± 1.8294.73 ± 2.0193.28 ± 1.0296.44 ± 2.9893.19 ± 1.5376.30 ± 3.295
IC5013.6819.8511.7715.7813.8011.71
(+)-241026.01 ± 2.8010.89 ± 1.8436.81 ± 3.5945.43 ± 1.7623.49 ± 1.60
30100.94 ± 0.4795.32 ± 1.8691.45 ± 0.9090.81 ± 1.4795.25 ± 1.3090.34 ± 1.79
IC5011.0616.2116.1520.9311.8614.64
(−)-241026.61 ± 1.3662.86 ± 0.9734.81 ± 0.7032.98 ± 2.6442.45 ± 0.77
3097.56 ± 0.7697.59 ± 1.2898.24 ± 0.4795.68 ± 2.10101.38 ± 0.7296.77 ± 1.22
IC5012.7618.908.7911.6911.0610.51
1: Mean ± SEM values from two determinations with five parallel wells in each. 2: Inhibition values less than 10% are regarded as negligible and are not given numerically.
Table 2. Antimicrobial activities of selected compounds according to Figure 2.
Table 2. Antimicrobial activities of selected compounds according to Figure 2.
Comp. 1,2Conc. (µg/mL)Growth Inhibition (%) 2 ± SEM 3
B. subtilisS. aureusP. aeruginosaE. coliC. albicansC. krusei
Amp10084.7 ± 7.0100.0 ± 3.5100.0 ± 3.2100.4 ± 8.6
1040.1 ± 6.368.48 ± 2.299.7 ± 3.860.2 ± 8.3
Nyst10099.0 ± 2.299.0 ± 2.1
1095.9 ± 16.084.3 ± 20.0
(+)-210016.6 ±4.1101.3 ± 7.0024.9 ± 2.636.8 ± 12.458.0 ± 13.4
1014.6 ± 6.4101.2 ± 8.517.7 ± 6.524.4 ± 2.4054.4 ± 13.9
(−)-21003.3 ± 6.8101.1 ± 8.30032.8 ±1.573.9 ± 10.7
100.6 ± 6.0101.6 ± 5.195.1± 29.613.7 ± 3.17.5 ± 21.157.8 ± 10.5
(+)-201005.2 ± 8.50017.1 ± 7.14.2 ± 8.773.1 ± 3.4
106.2 ± 4.70018.5 ± 2.62.0 ± 16.361.4 ± 1.8
(−)-2010010.5 ± 7.112.1 ± 4.9012.7 ± 7.2064.1 ± 20.1
1012.8 ± 9.210.8 ± 5.8020.2 ± 2.87.1 ± 9.767.5 ± 18.0
(−)-2110022.2 ± 2.17.6 ± 2.5026.3 ± 2.2064.5 ± 10.1
1014.3 ± 7.413.6 ± 5.2026.6 ± 8.02.7 ± 4.566.2 ± 11.8
(+)-2510020.7 ± 3.50022.0 ± 3.7024.3 ± 5.8
1016.5 ± 7.96.3 ± 1.3026.3 ± 2.5068.3 ± 8.4
(+)-2610016.2 ± 8.60017.8 ± 4.639.7 ± 3.80
1015.6 ± 9.74.5 ± 3.5021.8 ± 5.320.1 ± 9.166.8 ± 4.3
(−)-2610016.2 ± 7.72.6 ± 6.6020.5 ± 1.77.2 ± 3.460.7 ± 5
1014.9 ± 8.110.4 ± 3.2027.8 ± 9.5067.5 ± 15.2
(+)-2810023.6 ± 5.30018.5 ± 5.7066.2 ± 10.4
1016.5 ± 4.69.1 ± 3.2026.6 ± 6.57.9 ± 13.765.3 ± 6.2
(−)-3010023.1 ± 5.70015.52 ± 4.80066.4 ± 2.4
1014.7 ± 5.5094.0 ± 28.711.21 ± 6.534.2 ± 0.962.5 ± 12.6
(+)-3210015.9 ± 5.200101.7 ± 16.1
1018.2 ± 4.25.3 ± 4.6018.5 ± 5.41.4 ± 3.666.5 ± 9.9
1: Amp: ampicillin, Nys: nystatin; numeric values for all prepared monoterpenic compounds are given in Table S2 in the Supplementary Materials. 2: Solutions, which were not tested due to solubility problems, are marked with a en dash (–). 3: Mean ± SEM values from two determinations with 3 parallel wells in each.
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

Depp, D.; Tari, K.; Szekeres, A.; Kovács, A.; Zupkó, I.; Szakonyi, Z. 1,2,3-Triazolo-Bridged Click Coupling of Pinane-Based Azidodiol Enantiomers with Pyrimidine- and Purine-Based Building Blocks: Synthesis, Antiproliferative, and Antimicrobial Evaluation. Int. J. Mol. Sci. 2025, 26, 11705. https://doi.org/10.3390/ijms262311705

AMA Style

Depp D, Tari K, Szekeres A, Kovács A, Zupkó I, Szakonyi Z. 1,2,3-Triazolo-Bridged Click Coupling of Pinane-Based Azidodiol Enantiomers with Pyrimidine- and Purine-Based Building Blocks: Synthesis, Antiproliferative, and Antimicrobial Evaluation. International Journal of Molecular Sciences. 2025; 26(23):11705. https://doi.org/10.3390/ijms262311705

Chicago/Turabian Style

Depp, Dima, Kitti Tari, András Szekeres, Adriána Kovács, István Zupkó, and Zsolt Szakonyi. 2025. "1,2,3-Triazolo-Bridged Click Coupling of Pinane-Based Azidodiol Enantiomers with Pyrimidine- and Purine-Based Building Blocks: Synthesis, Antiproliferative, and Antimicrobial Evaluation" International Journal of Molecular Sciences 26, no. 23: 11705. https://doi.org/10.3390/ijms262311705

APA Style

Depp, D., Tari, K., Szekeres, A., Kovács, A., Zupkó, I., & Szakonyi, Z. (2025). 1,2,3-Triazolo-Bridged Click Coupling of Pinane-Based Azidodiol Enantiomers with Pyrimidine- and Purine-Based Building Blocks: Synthesis, Antiproliferative, and Antimicrobial Evaluation. International Journal of Molecular Sciences, 26(23), 11705. https://doi.org/10.3390/ijms262311705

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Article metric data becomes available approximately 24 hours after publication online.
Back to TopTop