Sapna
Saini‡
be,
G. Lakshma
Reddy‡
ae,
Anjali
Gangwar
be,
Harpreet
Kour
ae,
Gajanan G.
Nadre
ce,
Ramajayan
Pandian
de,
Sunny
Pal
b,
Utpal
Nandi
def,
Rashmi
Sharma
*be and
Sanghapal D.
Sawant
*ace
aNatural Products and Medicinal Chemistry Division, CSIR-Indian Institute of Integrative Medicine, Canal Road, Jammu-180001, India. E-mail: sd.sawant@ncl.res.in
bInfectious Diseases Division, CSIR – Indian Institute of Integrative Medicine, Canal Road, Jammu – 180001, India. E-mail: rashmi.sharma.09@iiim.res.in
cOrganic Chemistry Division, CSIR-National Chemical Laboratory, Dr. Homi Bhabha Road, Pune-411008, India
dPharmacology Division, CSIR – Indian Institute of Integrative Medicine, Canal Road, Jammu – 180001, India
eAcademy of Scientific and Innovative Research, Ghaziabad – 201002, India
fDepartment of Chemical Sciences, Bose Institute, Unified Academic Campus, Kolkata – 700 091, India
First published on 26th December 2024
Nitrofuran and pyrazolopyrimidine-based compounds possess a broad antimicrobial spectrum including Gram-positive and Gram-negative bacteria. In the present work, a series of conjugates of these scaffolds was synthesized and evaluated for antimicrobial activity against Staphylococcus aureus and methicillin-resistant S. aureus (MRSA). Many compounds showed MIC values of ≤2 μg ml−1, with compound 35 demonstrating excellent activity (MICs: 0.7 and 0.15 μg ml−1 against S. aureus and MRSA, respectively) and safety up to 50 μg ml−1 in HepG2 cells. Compound 35 also exhibited no hemolytic activity, biofilm eradication, and effectiveness against efflux-pump-overexpressing strains (NorA, TetK, MsrA) without resistance development. It showed synergistic effects with vancomycin (S. aureus) and rifampicin (MRSA). Mechanistic studies revealed that compound 35 exhibits good membrane-targeting abilities, as evidenced by DAPI/PI staining and scanning electron microscopy (SEM). In an intracellular model, it reduced bacterial load efficiently in both S. aureus and MRSA strains. With a strong in vitro profile, compound 35 demonstrated favorable oral pharmacokinetics at 30 mg kg−1 and potent in vivo anti-MRSA activity, highlighting its potential against antibiotic-resistant infections.
There is an urgent need to find newer molecules and scaffolds to overcome the limitation of these resistances. In this study, we present a series of molecules bearing conjugates of nitrofuran and pyrazolopyrimidine; two distinct and biologically important classes of molecules with significant MICs against S. aureus and MRSA as potent antibacterial agents targeting S. aureus and MRSA.
The nitrofurans are widely studied and typically used as antibiotics or antimicrobials.17 They appear to inhibit a number of microbial enzyme systems but their mechanism is still unclear. Their primary action is bacteriostatic, but at high doses, they are also bactericidal. They appear to inhibit a number of microbial enzyme systems but their mechanism is still unclear. There are many nitrofuran-based molecules that are used as a drug or being clinically investigated. Nitrofurantoin (1) is used to treat urinary tract infection; nifurtimox (2) is used for the treatment of Chagas disease.18 There are several other molecules reported in the nitrofuran class as antibacterial agents like nifuroxazide (3), furazolidone (4), nifuratel, ranbezolid, furylfuramide, nitrofurazone, nifurquinazole, nifurtoinol, nifurzide, etc.19–23 Some of the representative molecules that possess nitrofuran cores are shown in Fig. 1. On the other hand, the pyrazolopyrimidine scaffold has also been very well exploited and many analogs based on this scaffold are reported as anti-bacterial agents such as compounds (5) and (6), as shown in Fig. 1.24–27 The pyrazolopyrimidine scaffold is considered as a bioisostere of the biogenic purine class and this nucleus has a high impact in the field of pharmaceutical sciences with a vast spectrum of biological activities that includes adenosine receptor antagonists,28,29 anti-viral,30,31 anti-cancer and some others32,33 Further, there are a good number of literature reports on their PDE inhibitory potential34,35 including some approved drugs that are known for anti-inflammatory36,37 activities also.
![]() | ||
Fig. 1 Structures of some biologically important nitrofuran and pyrazolopyrimidine class of molecules. |
Both the nitrofuran and pyrazolopyrimidine scaffolds represent promising candidates as antibacterial agents and it is expected that these molecules could further give some potential candidates in synergy. Keeping in mind the importance of these scaffolds, we started a program based on the literature report of H. A. Burch in 1968, in which 4-amino-6-(5-nitro-2-furyl)-lH-pyrazolo[3,4-d]pyrimidines were presented to show significant activity against S. aureus with a low MIC of 0.1 μg ml−1.25 The concept of bioisosterism was used while designing our present series, where the idea was that the molecules that exhibit similar volume, shape, and/or physicochemical properties can produce broadly similar biological effects.38 Considering this report, we have designed a bioisostere series based on nitrofuranyl–pyrazolopyrimidine scaffold (Fig. 2).
We synthesized 38 novel compounds exemplified as 7-amino-1-methyl-5-(5-nitrofuran-2-yl)-3-propyl-1H-pyrazolo[4,3-d]pyrimidine analogs and screened against S. aureus and MRSA. This synthesis was based on known methods and one of our recent reports, where a microwave-assisted strategy was established for the preparation of 5-substituted-1H-pyrazolo[4,3-d]pyrimidin-7(6H)-one as mTOR inhibitors; similar procedures were adopted for the preparation of this series for the synthesis of compound 8.39 This was utilized further for the preparation of final compounds using literature-reported methods.40
After evaluating the in vitro antibacterial activity of various derivatives, we identified a potent compound, compound 35, which demonstrated remarkable activity against both S. aureus ATCC 29213 and MRSA ATCC 15187 isolates. This compound was selected for further investigation. We evaluated its safety index by assessing its cytotoxicity and hemolytic activity. Additionally, we investigated time-kill kinetics, the potential for resistance development, combinational effects with standard drugs, and its mechanism of action through mechanistic studies, including scanning electron microscopy and DAPI/PI staining. Ex vivo studies were conducted to evaluate its efficiency in reducing bacterial burden within macrophages infected with bacterial cells. Pharmacokinetic (PK) studies, along with evaluations of in vivo safety and antibacterial efficacy, were performed using a mouse model of systemic MRSA infection. The present study offers valuable insights into developing new antimicrobials based on nitrofuranyl–pyrazolopyrimidine hybrid conjugates.
Further in our observation, we screened more analogs with different substitutions on arylamines, including the – OMe (compound 36) or –OCF3 (compound 37) substitutions at the para-position of arylamines, to see the effect of protection on active compound 35. However, these compounds turned out to be inactive with MICs >32 μg ml−1. Some compounds with heteroatom bearing cyclic aliphatic rings along with the presence of the hydroxyl group at varying positions were also designed, synthesized and screened (Table 2, entries 29–32), all these molecules were found to be inactive.
Apart from this, various halo substitutions were tried on arylamine rings (Table 3), in which the compounds with 3- and 4-fluoro substitution on the arylamine ring were found active, i.e., compound 38 and 39, respectively. Other compounds from the halo series were not having significant activity. Next, as shown in Table 3, different hetero aryl amines were placed to see the effect on the activity; some of the compounds have shown good activity, and further work has been undertaken to see the structure–activity relationship of these substitutions on this scaffold.
These antibacterial activity results prompted us to further examine the cytotoxicity effect on the safety of the nitrofuranyl–pyrazolopyrimidine compounds on HepG2 cell lines using MTT assay. As a result, no compound was found to have a cytotoxic effect up to 50 μg ml−1 concentrations (Tables 1–3). Thus, the overall observation of the results indicates that compound 35 was the most active.
Consequently, further studies, including in vivo efficacy, physicochemical parameter analysis, and pharmacokinetic profiling of this compound, were planned. Fig. 3 illustrates the dose–response curve for compound 35 against S. aureus and MRSA. The curve shows a clear growth inhibition with increasing concentrations of the compound, with MIC values of 0.7 and 0.15 μg ml−1, respectively. Additionally, the minimum bactericidal concentration (MBC) of compound 35 was found to be equivalent to the MIC.
Based on the excellent antibacterial activity of compound 35, we further evaluated the ability of compound 35 to inhibit S. aureus and MRSA biofilms. Compound 35 was tested at 1/2×, 1×, 2×, 4×, and 6× MIC concentrations, with vancomycin being used as a drug control at the respective concentrations. The compound effectively inhibited biofilm formation, with a notable decrease in biofilm formation as its concentration increased, as shown in Fig. 4. Compound 35 inhibited biofilm formation at all tested concentrations, whereas vancomycin was ineffective at 1/2× MIC concentration. The compound was further assessed for its ability to disrupt preformed biofilms in S. aureus and MRSA where compound 35 was able to show maximum 30% disruption in preformed biofilm in both bacterial strains at 4× and 6× MIC concentration, while the drug control vancomycin exhibited 40–50% disruption at 4× and 6× MIC concentrations (ESI,† Fig. S1). Since the compound effectively prevents biofilm formation but shows limited ability to disrupt existing biofilms, it is better suited for preventing biofilm development than for treating established infections.
Antimicrobial resistance, particularly due to overexpression of efflux pumps, poses a significant hurdle in the discovery of antibacterial drugs. As discussed by Poole (2007), targeting efflux pumps can be an effective approach to combat antibiotic resistance.41 In this context, compound 35 was further tested against several strains of S. aureus that overexpress different efflux pumps: NorA (SA-1199B), MsrA (SA-K2191), and TetK (SA-K2192) as well as a ciprofloxacin-sensitive strain (SA-1199) used as a control. MIC values for these strains are provided in Table 4.
Strain | Phenotype | MIC (μg ml−1) | ||
---|---|---|---|---|
Ciprofloxacin | Vancomycin | Compound 35 | ||
SA-1199 | Ciprofloxacin-sensitive | 0.25 | 0.25 | 0.3 |
SA-1199B | NorA overexpressing | 4.14 | 0.25 | 0.6 |
SA-K2191 | MsrA overexpressing | 4.14 | 0.25 | 0.15 |
SA-K2192 | TetK overexpressing | 0.25 | 0.56 | 0.3 |
Compound 35 showed similar MIC values for both S. aureus 29213 and NorA overexpressing strain, S. aureus 1199B, while strains (SA-1199 and SA-K2192) showed a onefold decrease in MIC value and SA-K2191 showed a twofold decrease in MIC value clearly shown in Fig. 5 representing a dose–response curve of compound 35 against these strains. These results indicate that compound 35 remains effective against strains with overexpressed efflux pumps, highlighting its potential for targeting resistant strains.
Further, the hemolytic activity (HC50) of compound 35 against rabbit erythrocytes was determined to confirm their toxicity and membrane selectivity. This assay helps evaluate the safety profile of compound 35 with respect to erythrocytes. We assessed the effect of compound 35 on erythrocytes at various concentrations, with vancomycin serving as a drug control. Triton-X, a lysing agent, was used as a positive control at 1% concentration, which demonstrated 100% hemolytic activity, while PBS with 0% lysis served as the negative control. The growth control, which contained undisturbed RBCs, exhibited minimal hemolysis. Compound 35 showed no hemolytic activity comparable to the standard drug vancomycin, whereas 1% Triton-X induced substantial hemolysis. This assay further predicted compound 35 as safe against erythrocytes.42Fig. 6 presents the cell viability and percentage hemolysis of the test compounds at different concentrations. Compound 35 shows a safe profile against macrophage cell lines used and erythrocytes up to a concentration of 200 μg ml−1.
![]() | ||
Fig. 7 The table displays the calculated FIC indices and their outcome against S. aureus and MRSA with a graphical image showing outcomes in one place for S. aureus and MRSA. |
For MRSA, compound 35 showed both concentration- and time-dependent killing. At 1× MIC, it was bacteriostatic. At 2× MIC, it exhibited initial killing but exhibited some regrowth after 8–10 h, possibly due to the compound's labile nature. At 4× MIC, compound 35 was bactericidal and showed killing at the initial duration (within 2 h), earlier than vancomycin.
In addition, the synergistic effect of compound 35 with vancomycin in S. aureus and with rifampicin in MRSA has been verified using time-kill curves.47 The combination of compound 35 with vancomycin at synergistic concentrations demonstrated bacterial growth inhibition but was less effective, achieving a >10log reduction in CFU for up to 8 h. However, regrowth was observed after 24 h, likely due to the bacteriostatic nature of the combination. The combination of compound 35 and rifampicin at synergistic concentrations exhibits a moderately improved effect compared to rifampicin alone at the FIC concentration, which is already effective, as shown in Fig. S3.†
Assessing the tendency of bacteria to develop resistance is a crucial factor in determining the effectiveness of newly developed antimicrobials. Consequently, we investigated the resistance propensity of compound 35 against both S. aureus and MRSA by exposing the bacteria to sublethal concentrations of the compound over 20 days. Vancomycin and norfloxacin were employed as control agents in this assay. Fig. 9(C and D) depict that compound 35, similar to vancomycin, has a minimal tendency to induce resistance in both S. aureus and MRSA strains over 16 days. In contrast, fluoroquinolone norfloxacin (positive control) significantly increased the original MIC by 200-fold for S. aureus and 3200-fold for MRSA by day 20.48,49 The lack of resistance development observed with compound 35 may be attributed to its rapid bactericidal action and membrane-targeting mechanism.50
Subsequently, we assessed the integrity of bacterial membranes using fluorescence microscopy and percentage permeability analysis. The fluorescence LIVE/DEAD bacterial viability assay employs two dyes: propidium iodide (PI) and 4′,6-diamidino-2-phenylindole (DAPI). DAPI, a blue fluorescent nucleic acid stain, can penetrate cell membranes regardless of their integrity, staining both live and damaged cells. In contrast, PI, a red fluorescent nucleic acid stain, passes only through damaged cell membranes. To further explore the antimicrobial mechanism of compound 35, S. aureus and MRSA were treated with or without the compound and stained with DAPI and PI dyes. As shown in Fig. 12, blue fluorescence was visible in both the growth control and compound 35-treated groups, while strong red fluorescence appeared only in the compound-treated group. The growth control exhibited blue fluorescence with minimal traces of red, indicating an intact bacterial membrane. In contrast, both the drug control and compound-treated groups showed red and blue fluorescence in their respective channels, signifying their effectiveness in disrupting the bacterial membrane. MRSA also shows similar results as shown in Fig. S6 of the ESI.† These findings demonstrate that compound 35 compromises the bacterial cell membrane integrity in both S. aureus ATCC 29213 and MRSA ATCC 15187.
The efficiency of compound 35 to permeate the bacterial membrane was further evaluated using a propidium iodide (PI) permeabilization assay. Various concentrations of the compound (1×, 2×, 4×, 8× MIC) were tested, and permeability in both S. aureus and MRSA was analyzed over time. In both strains, permeability steadily increased, reaching a maximum of 60% in S. aureus and 45% in MRSA at 80 min compared to drug control vancomycin (4× MIC and 8× MIC) which is giving maximum permeability of 22% in S. aureus and 18% in MRSA (Fig. 13). The effect of increasing compound concentration on bacterial permeability was minimal, as shown in Fig. 13.
Overall, our findings demonstrate that compound 35 effectively kills bacteria by disrupting the cell membrane. However, we cannot exclude the possibility that other mechanisms may also contribute to bacterial killing.
Parameter | Unit | IV @ 1 mg kg−1 | PO @ 30 mg kg−1 |
---|---|---|---|
t 1/2,β | h | 0.50 | 0.35 |
T max | h | — | 0.50 |
C max | ng ml−1 | — | 2010 |
C 0 | ng ml−1 | 552 | — |
AUC0−t | ng h ml−1 | 90.8 | 3146.1 |
AUC0−∞ | ng h ml−1 | 94.8 | 3149.8 |
CL | ml min−1 kg−1 | 176 | 159 |
V d | L kg−1 | 1.69 | 4.75 |
After a 24 h incubation period, all mice were sacrificed, and their lungs and kidneys were isolated to analyze the bacterial load reduction. The organs were thoroughly homogenized and plated on Mueller–Hinton agar (MHA) supplemented with 20 μg ml−1 ciprofloxacin to detect the bacterial load (CFU ml−1). The plates were incubated for 24 h, and bacterial loads in each group were compared. Fig. 15 illustrates the reduction in bacterial load (CFU ml−1) in kidneys and lungs across different groups: untreated, treated with compound 35, and vancomycin as the drug control. Statistical analysis was conducted using an unpaired t-test in GraphPad Prism, with a p-value <0.05 considered significant.
In the kidneys, the group treated with 50 mg kg−1 exhibited a significantly greater difference (P < 0.0001) compared to the untreated group than the group treated with 100 mg kg−1 (P < 0.0004) and the vancomycin control group (P < 0.0023). In contrast, in the lungs, the groups treated with 50 mg kg−1 and 100 mg kg−1 compound 35 showed lesser significant differences compared to the untreated group, with P-values of 0.0068 and 0.0085, respectively, compared to the vancomycin control group (P < 0.0009).
Step 1: 1-methyl-5-(5-nitrofuran-2-yl)-3-propyl-1H-pyrazolo[4,3-d]pyrimidin-7(6H)-one (8)
In a typical procedure, a solution of 4-amino-1-methyl-3-propyl-1H-pyrazole-5-carboxamide (500 mg, 2.74 mmol) and 5-nitrofuran-2-carbaldehyde (395 mg 2.8 mmol) in DMSO:
H2O (1
:
1) with K2S2O8 added (2.225 g 8.74 mmol) was taken in a sealed reaction tube and the reaction mixture was irradiated under microwave conditions for 3 min with a power of 350 W at 100 °C. After completion, the reaction mass was diluted with EtOAc (20 ml) and water (30 ml) was added. The organic layer was separated and extracted with EtOAc (2 × 10 ml). The combined organic layer was then washed with brine solution, concentrated under vacuum and the residue was purified by column chromatography, affording a yellow solid, 1-methyl-5-(5-nitrofuran-2-yl)-3-propyl-1H-pyrazolo[4,3-d]pyrimidin-7(6H)-on, i.e., compound 8 (707 mg) in 85% yield.
Yellow solid; 1H NMR (400 MHz, DMSO) δ 8.30 (s, 1H), 7.86 (s, 1H), 4.15 (s, 3H), 2.81 (t, J = 7.6 Hz, 2H), 1.84–1.68 (m, 2H), 0.96 (t, J = 7.6 Hz, 3H). 13C NMR (101 MHz, DMSO) δ 152.00, 147.45, 145.70, 139.85, 136.82, 124.70, 115.06, 114.06, 68.9, 37.92, 27.02, 21.50, 13.80; HRMS (ESI) calcd for C13H14N5O4 [M + H]+: 304.1045, found 304.1040.
Step 2: 7-chloro-1-methyl-5-(5-nitrofuran-2-yl)-3-propyl-1H-pyrazolo[4,3-d]pyrimidine (9)
Compound 8 1-methyl-5-(5-nitrofuran-2-yl)-3-propyl-1H-pyrazolo[4,3-d]pyrimidin-7(6H)-one (700 mg, 2.31 mmol) was dissolved in POCl3 (4 ml), DIPEA (402 μl 2.31 mmol) was added, and the mixture was refluxed for 5 h. After the reaction was completed, the POCl3 was removed under vacuum. Then, workup was carried out using CHCl3 (20 ml) and ice water (15 ml). The organic layer was separated and the water layer re-extracted with CHCl3 (2 × 15 ml). The combined organic layers were washed with brine solution, dried over anhydrous sodium sulfate, and concentrated. The residue was purified by column chromatography. Compound 9 was obtained as a light yellow solid (592 mg, yield 80%).
Yellow solid; 1H NMR (400 MHz, DMSO) δ 8.32 (s, 1H), 7.88 (s, 1H), 4.16 (s, 3H), 2.82 (t, J = 7.4 Hz, 2H), 1.84–1.68 (m, 2H), 0.96 (t, J = 7.3 Hz, 3H). 13C NMR (101 MHz, DMSO) δ 153.75, 152.07, 147.43, 145.67, 139.96, 136.88, 124.72, 115.02, 114.06, 37.92, 27.02, 21.52, 13.79; mass ESI [M + H]+: 321.9.
Step 3: (a) procedure for aliphatic amines
Synthesis of 4-(1-methyl-5-(5-nitrofuran-2-yl)-3-propyl-1H-pyrazolo[4,3-d]pyrimidin-7-yl)morpholine ( 10 )
In a typical procedure, morpholine (20 mg, 0.23 mmol, 1 eq.) was dissolved in dry DMF (2 ml) and to it was added K2CO3 (96 mg, 0.70 mmol 3q); then, 7-chloro-1-methyl-5-(5-nitrofuran-2-yl)-3-propyl-1H-pyrazolo[4,3-d]pyrimidine (75 mg. 0.23 mmol, 1 eq.) was added to the mixture. The reaction mixture was stirred for 3–4 h at 25 °C. After completion of the reaction, 20 ml EtOAc and 30 ml water were added and the organic layer was separated. The water layer was re-extracted with EtOAc (2 × 20 ml) and the combined organic layers were washed with brine solution, dried over anhydrous sodium sulfate, and concentrated. The residue was purified by column chromatography. Compound 10 was obtained as a yellow solid (75 mg, yield 86%).
Yellow solid; m.p.: 206–208 °C; 1H NMR (400 MHz, CDCl3) δ 7.44 (d, J = 3.7 Hz, 1H), 7.32 (d, J = 3.7 Hz, 1H), 4.14 (s, 3H), 3.94 (t, J = 4.4 Hz, 4H), 3.62 (t, J = 4.4 Hz, 4H), 3.02 (t, J = 7.6 Hz, 2H), 1.94–1.84 (m, 2H), 1.05 (t, J = 7.4 Hz, 3H); 13C NMR (101 MHz, DMSO) δ 154.14, 153.40, 151.70, 146.98, 145.93, 143.54, 123.63, 114.74, 114.09, 65.57, 49.39, 39.20, 27.17, 21.35, 13.85. HRMS (ESI) calcd for C17H21N6O4 [M + H]+: 373.1624, found 373.1614.
(b) Procedure for aromatic amines
4-((1-Methyl-5-(5-nitrofuran-2-yl)-3-propyl-1H-pyrazolo[4,3-d]pyrimidin-7-yl)amino)phenol ( 35 )
In a typical procedure, 7-chloro-1-methyl-5-(5-nitrofuran-2-yl)-3-propyl-1H-pyrazolo[4,3-d]pyrimidine (75 mg, 0.23 mmol) and 4-aminophenol (38 mg, 0.35 mmol) were dissolved in iPrOH (5 ml). To this solution was added 37% conc. HCl (1–2 drops). The mixture was heated at reflux for 3–6 h. Then the reaction was cooled and evaporated at reduced pressure. The residue was diluted with CHCl3 (20 ml), neutralized by Et3N and washed with water (20 ml), and the organic layer was separated. The water layer was re-extracted with CHCl3 (2 × 10 ml) and the combined organic layer was washed with brine solution, dried over anhydrous sodium sulfate, and concentrated. The residue was purified by column chromatography; compound 35 was obtained as a yellow solid (75 mg, yield 76%).
Yellow solid; m.p.: 251–253 °C; 1H NMR (400 MHz, acetone d6) δ 7.66 (d, J = 9.2 Hz, 2H), 7.6 (d, J = 4 Hz, 1H), 7.25 (d, J = 4 Hz, 1H), 6.93 (d, J = 9.2 Hz, 2H), 4.4 (s, 3H), 2.93 (t, J = 7.6 Hz, 2H), 1.91–1.82 (m, 2H), 1.01 (t, J = 7.2 Hz, 3H); 13C (126 MHz, acetone-d6), δ 156.06, 155.36, 152.94, 149.03, 148.73, 146.19, 143.76, 131.27, 125.41, 122.11, 115.91, 114.47, 114.06, 39.76, 29.36, 22.35, 14.26; HRMS (ESI) calcd for C19H19N6O4 [M + H]+: 395.1468, found 395.1463.
1-Methyl-5-(5-nitrofuran-2-yl)-3-propyl-7-(pyrrolidin-1-yl)-1H-pyrazolo[4,3-d]pyrimidine ( 11 )
Orange-colored solid; m.p.: 156–158 °C; 1H NMR (400 MHz, CDCl3) δ 7.42 (d, J = 4 Hz, 1H), 7.29 (d, J = 4 Hz, 1H), 4.20 (s, 3H), 3.87 (t, J = 6.4 Hz, 4H), 3.0 (t, J = 7.6 Hz, 2H), 2.05 (t, J = 6.4 Hz, 4 Hz), 1.93–1.83 (m, 2H), 1.05 (t, J = 7.6 Hz, 3H); 13C NMR (126 MHz, DMSO) δ 154.81, 151.46, 149.92, 146.72, 144.64, 142.80, 122.08, 114.76, 113.54, 50.15, 42.07, 27.10, 25.07, 21.42, 13.88; HRMS (ESI) calcd for C17H21N6O3 [M + H]+: 357.1675, found 357.1667.
1-Methyl-7-(4-methylpiperazin-1-yl)-5-(5-nitrofuran-2-yl)-3-propyl-1H-pyrazolo[4,3-d]pyrimidine ( 12 )
Yellow solid; m.p.: 176–178 °C; 1H NMR (400 MHz, CDCl3) δ 7.43 (d, J = 4 Hz 1H), 7.32 (d, J = 4 Hz, 1H), 4.12 (s, 3H), 3.65 (t, J = 4.2 Hz, 4H), 3.0 (t, J = 7.6 Hz, 2H), 2.65 (t, J = 4.2 Hz, 4H), 2.39 (s, 3H), 1.9 (m, 2H), 1.05 (t, J = 7.2 Hz 3H); 13C NMR (126 MHz, CDCl3) δ 154.78, 153.83, 152.12, 148.19, 147.84, 144.71, 124.51, 113.37, 113.12, 54.39, 49.33, 46.14, 38.89, 27.79, 22.05, 14.10; HRMS (ESI) calcd for C18H24N7O3 [M + H]+: 386.1941, found 386.1927.
1-Methyl-5-(5-nitrofuran-2-yl)-7-(4-phenylpiperidin-1-yl)-3-propyl-1H-pyrazolo[4,3-d]pyrimidine ( 13 )
Yellow solid; m.p.: 187–189 °C; 1H NMR (400 MHz, CDCl3) δ 7.43 (d, J = 3.7 Hz, 1H), 7.37–7.33 (m, 3H), 7.29–7.23 (m, 3H), 4.20–4.17 (m, 5H), 3.26–3.19 (m, 2H), 3.02 (t, J = 7.6 Hz 2H), 2.90–2.82 (m, 1H), 2.12–2.07 (m, 2H), 2.03–1.96 (m, 2H), 1.95–1.86 (m, 2H), 1.06 (t, J = 7.4 Hz, 3H). 13C NMR (126 MHz, CDCl3) δ 154.90, 154.40, 148.07, 147.90, 145.09, 145.01, 144.57, 128.67, 126.76, 126.67, 124.66, 113.43, 113.09, 50.35, 42.44, 38.86, 32.84, 27.82, 22.10, 14.13; HRMS (ESI) calcd for C24H27N6O3 [M + H]+: 447.2145, found 447.2140.
1-Methyl-5-(5-nitrofuran-2-yl)-7-(piperidin-1-yl)-3-propyl-1H-pyrazolo[4,3-d]pyrimidine ( 14 )
Yellow solid; m.p.: 160–162 °C; 1H NMR (400 MHz, CDCl3) δ 7.42 (d, J = 4 Hz, 1H), 7.30 (d, J = 4 Hz, 1H), 4.33 (s, 3H), 3.56 (t, J = 4.4 Hz, 4H), 3.0. (t, J = 7.6 Hz, 2H),1.94–1.75 (m, 8H), 1.05 (t, J = 7.2 Hz, 3H); 13C NMR (126 MHz, DMSO) δ 154.30, 153.81, 151.61, 147.03, 145.81, 143.24, 123.70, 114.77, 113.90, 49.90, 39.05, 27.18, 24.92, 23.75, 21.36, 13.87. HRMS (ESI) calcd for C18H23N6O3 [M + H]+: 371.1832, found 371.1831.
1-(1-Methyl-5-(5-nitrofuran-2-yl)-3-propyl-1H-pyrazolo[4,3-d]pyrimidin-7-yl)piperidin-4-one ( 15 )
Yellow solid; m.p.: 214–216 °C; 1H NMR (400 MHz, CDCl3) δ 7.4 (d, J = 3.6 Hz, 1H), 7.3 (d, J = 3.6 Hz, 1H), 4.16 (s, 3H), 3.93 (t, J = 6.1 Hz, 4H), 3.0 (t, J = 7.6 Hz, 2H), 2.69 (t, J = 6.1 Hz, 4H), 1.92–1.83 (m, 2H), 1.03 (t, J = 7.4 Hz, 3H); 13C NMR (101 MHz, DMSO) δ 207.05, 154.15, 153.03, 151.70, 146.94, 145.87, 143.64, 123.64, 114.77, 114.12, 47.82, 40.19, 39.34, 27.17, 21.37, 13.85; HRMS (ESI) calcd for C18H21N6O4 [M + H]+: 385.1624, found 385.1610.
1-(1-Methyl-5-(5-nitrofuran-2-yl)-3-propyl-1H-pyrazolo[4,3-d]pyrimidin-7-yl)piperidin-4-ol ( 16 )
Yellow solid; m.p.: 202–204 °C; 1H NMR (400 MHz, CDCl3) δ 7.43 (d, J = 3.7 Hz, 1H), 7.31 (d, J = 3.7 Hz, 1H), 4.13 (s, 3H), 4.06 (s, 1H), 4.08–4.01 (m, 1H), 3.95–3.89 (m, 2H), 3.39–3.33 (m, 2H), 3.01 (t, J = 7.6 Hz, 2H), 2.19–2.07 (m, 2H), 1.94–1.86 (m, 2H), 1.85–1.75 (m, 2H), 1.05 (t, J = 7.3 Hz, 3H); 13C NMR (126 MHz, CDCl3) δ 154.81, 154.14, 152.11, 148.05, 147.83, 144.57, 124.58, 113.42, 113.09, 67.13, 46.92, 38.74, 33.71, 27.79, 22.08, 14.11; HRMS (ESI) calcd for C18H23N6O4 [M + H]+: 387.1781, found 387.1769.
1-Methyl-5-(5-nitrofuran-2-yl)-N-(2-(piperidin-1-yl)ethyl)-3-propyl-1H-pyrazolo[4,3-d]pyrimidin-7-amine ( 17 )
Yellow solid; m.p.: 210–213 °C 1H NMR (400 MHz, CDCl3) δ 7.41 (d, J = 3.8 Hz, 1H), 7.30 (d, J = 3.8 Hz, 1H), 6.70 (s, 1H), 4.28 (s, 3H), 3.73 (m, 2H), 2.97 (t, J = 7.6 Hz, 2H), 2.69 (t, J = 5.9 Hz, 2H), 2.51 (br, 4H), 1.94–1.81 (m, 2H), 1.66–1.56 (m, 4H), 1.55–1.46 (m, 2H), 1.04 (t, J = 7.4 Hz, 3H); 13C NMR (126 MHz, CDCl3) δ 155.24, 151.98, 150.19, 148.51, 146.46, 142.32, 121.27, 113.45, 112.98, 47.23, 39.19, 37.77, 31.12, 27.68, 26.36, 25.85, 22.13, 14.08; HRMS (ESI) calcd for C20H28N7O3 [M + H]+: 414.2254, found 414.2250.
1-Methyl-5-(5-nitrofuran-2-yl)-3-propyl-N-(tetrahydro-2H-pyran-4-yl)-1H-pyrazolo[4,3-d]pyrimidin-7-amine ( 18 )
Orange color solid; m.p.: 238–240 °C; 1H NMR (400 MHz, CDCl3) δ 7.42 (d, J = 3.7 Hz, 1H), 7.26 (d, J = 3.7 Hz,1H), 5.06 (d, J = 7.0 Hz, 1H), 4.60–4.48 (m, 1H), 4.27 (s, 3H), 4.11–4.04 (m, 2H), 3.65 (m, 2H), 2.97 (t, J = 7.6 Hz, 2H), 2.22 (m, 2H), 1.92–1.81 (m, 2H), 1.64 (m, 2H), 1.03 (t, J = 7.4 Hz, 3H); 13C NMR (126 MHz, DMSO) δ 154.72, 151.54, 148.93, 147.54, 144.48, 141.33, 120.95, 114.77, 113.80, 66.26, 47.30, 39.17, 31.73, 27.11, 21.51, 13.82; HRMS (ESI) calcd for C18H23N6O4 [M + H]+: 387.1781, found 387.1772.
1-Methyl-N-(2-morpholinoethyl)-5-(5-nitrofuran-2-yl)-3-propyl-1H-pyrazolo[4,3-d]pyrimidin-7-amine ( 19 )
Yellow solid; m.p.: 185–186 °C; 1H NMR (400 MHz, CDCl3) δ 7.41 (d, J = 3.7 Hz, 1H), 7.30 (d, J = 3.7 Hz, 1H), 6.40 (s, 1H), 4.29 (s, 3H), 3.82–3.72 (m, 6H), 2.97 (t, J = 7.6 Hz, 2H), 2.77 (t, J = 5.9 Hz, 2H), 2.63–2.57 (m, 4H), 1.92–1.82 (m, 2H), 1.04 (t, J = 7.4 Hz, 3H); 13C NMR (101 MHz, DMSO) δ 154.85, 151.46, 149.62, 147.72, 144.36, 141.08, 120.94, 114.75, 113.39, 66.25, 56.55, 53.26, 39.07, 37.37, 27.12, 21.46, 13.81; HRMS (ESI) calcd for C19H26N7O4 [M + H]+: 416.2046, found 416.2041.
1-Methyl-N-(3-morpholinopropyl)-5-(5-nitrofuran-2-yl)-3-propyl-1H-pyrazolo[4,3-d]pyrimidin-7-amine ( 20 )
Yellow solid; m.p.: 202–204 °C; 1H NMR (400 MHz, CDCl3) δ 7.42 (d, J = 3.6 Hz, 1H), 7.29 (d, J = 3.6 Hz, 1H), 6.48 (br s 1H), 4.31 (s, 3H), 3.82 (m, 2H), 3.73 (t, J = 4.4 Hz, 4H), 2.97 (t, J = 7.6 Hz 2H), 2.61 (t, J = 6.0 Hz, 2H), 2.54 (t, J = 4.4 Hz, 4H), 1.99–1.93 (m, 2H), 1.92–1.82 (m, 2H), 1.04 (t, J = 7.4 Hz, 3H); 13C NMR (101 MHz, DMSO) δ 154.93, 151.49, 149.68, 147.73, 144.38, 141.03, 121.02, 114.75, 113.47, 66.10, 56.15, 53.33, 39.18, 39.15, 27.11, 25.16, 21.48, 13.80; HRMS (ESI) calcd for C20H28N7O4 [M + H]+: 430.2203, found 430.2200.
N-Benzyl-1-methyl-5-(5-nitrofuran-2-yl)-3-propyl-1H-pyrazolo[4,3-d]pyrimidin-7-amine ( 21 )
Yellow solid; m.p.: 190–192 °C; 1H NMR (400 MHz, CDCl3) δ 7.48 (d, J = 7.2 Hz, 2H), 7.42 (d, J = 4 Hz, 1H), 7.39–7.33 (m, 3H), 7.29 (d, J = 4 Hz, 1H), 5.53 (t, J = 5.2 Hz, 1H), 4.92 (d, J = 5.2 Hz, 2H), 4.24 (s, 3H), 2.97 (t, J = 7.6 Hz, 2H), 1.91–1.82 (m, 2H) 1.05 (t, J = 7.2 Hz, 3H); 13C NMR (101 MHz, DMSO) δ 154.73, 151.48, 149.31, 147.55, 144.45, 141.24, 139.41, 128.15, 127.70, 126.72, 120.92, 114.68, 113.52, 43.87, 39.24, 27.11, 21.47, 13.80; HRMS (ESI) calcd for C20H21N6O3 [M + H]+: 393.1675, found 393.1669.
N-(4-Methoxybenzyl)-1-methyl-5-(5-nitrofuran-2-yl)-3-propyl-1H-pyrazolo[4,3-d]pyrimidin-7-amine ( 22 )
Yellow solid; m.p.: 160–162 °C; 1H NMR (400 MHz, CDCl3) δ 7.41 (m, 3H), 7.31 (d, J = 3.7 Hz, 1H), 6.92 (d, J = 8.4 Hz, 2H), 5.47 (t, J = 4.8 Hz, 1H),4.84 (d, J = 4.8 Hz, 2H), 4.23 (s, 3H), 3.81 (s, 3H), 2.96 (t, J = 7.6 Hz, 2H), 1.91–1.81 (m, 2H), 1.03 (t, J = 7.3 Hz, 3H); 13C NMR (126 MHz, DMSO) δ 158.14, 154.78, 151.47, 149.19, 147.54, 144.38, 141.17, 131.31, 129.16, 120.88, 114.72, 113.50, 54.93, 43.26, 39.27, 27.11, 21.49, 13.81; HRMS (ESI) calcd for C21H23N6O4 [M + H]+: 423.1781, found 423.1771.
N-(3-Fluoro-4-methoxybenzyl)-1-methyl-5-(5-nitrofuran-2-yl)-3-propyl-1H-pyrazolo[4,3-d]pyrimidin-7-amine ( 23 )
Yellow solid; m.p.: 198–199 °C; 1H NMR (400 MHz, CDCl3) δ 7.42 (d, J = 3.7 Hz, 1H), 7.29 (d, J = 3.7 Hz, 1H), 7.24 (s, 1H), 7.19 (dd, J = 11.8, 2.0 Hz, 1H), 6.99–6.94 (m, 1H), 5.52 (t, J = 5.4 Hz, 1H), 4.83 (d, J = 5.4 Hz, 2H), 4.25 (s, 3H), 3.89 (s, 3H), 2.96 (t, J = 7.6 Hz, 2H), 1.91–1.82 (m, 2H), 1.02 (t, J = 7.4 Hz, 3H); 13C NMR (101 MHz, DMSO) δ 154.70, 152.41, 151.49, 149.99, 149.14, 147.47, 145.95, 144.39, 141.21, 132.41, 124.06, 120.89, 115.55, 114.67, 113.46, 55.84, 43.00, 39.23, 27.10, 21.48, 13.77; HRMS (ESI) calcd for C21H22FN6O4 [M + H]+: 441.1687, found 441.1678.
6-((1-Methyl-5-(5-nitrofuran-2-yl)-3-propyl-1H-pyrazolo[4,3-d]pyrimidin-7-yl)amino)hexan-1-ol ( 24 )
Yellow solid; m.p.: 142–144 °C; 1H NMR (400 MHz, CDCl3) δ 7.42 (d, J = 3.7 Hz, 1H), 7.30 (d, J = 3.7 Hz, 1H), 5.26 (t, J = 5.1 Hz, 1H), 4.27 (s, 3H), 3.74 (m, 2H), 3.68 (t, J = 6.4 Hz, 2H), 2.97 (t, J = 7.6 Hz 2H), 1.92–1.84 (m, 2H), 1.82–1.75 (m, 2H), 1.65–1.59 (m, 2H), 1.52–1.47 (m, 4H), 1.03 (t, J = 7.4 Hz, 3H); 13C NMR (101 MHz, DMSO) δ 154.95, 151.48, 149.61, 147.72, 144.38, 141.05, 120.97, 114.71, 113.45, 60.65, 40.41, 39.14, 32.45, 28.46, 27.12, 26.38, 25.26, 21.47, 13.81; HRMS (ESI) calcd for C19H27N6O4 [M + H]+: 403.2094, found 403.2084.
5-((1-Methyl-5-(5-nitrofuran-2-yl)-3-propyl-1H-pyrazolo[4,3-d]pyrimidin-7-yl)amino)pentan-1-ol ( 25 )
Yellow solid; m.p.: 169–171 °C; 1H NMR (400 MHz, CDCl3) δ 7.42 (d, J = 3.7 Hz, 1H), 7.30 (d, J = 3.7 Hz, 1H), 5.32 (t, J = 5.3 Hz, 1H), 4.27 (s, 3H), 3.80–3.69 (m, 4H), 2.97 (t, J = 7.6 Hz 2H), 1.92–1.78 (m, 4H), 1.74–1.67 (m, 2H), 1.61–1.53 (m, 2H), 1.03 (t, J = 7.4 Hz, 3H); 13C NMR (101 MHz, DMSO) δ 154.94, 151.47, 149.58, 147.70, 144.37, 141.02, 120.95, 114.72, 113.45, 60.63, 40.46, 39.13, 32.17, 28.28, 27.12, 23.01, 21.48, 13.82; HRMS (ESI) calcd for C18H25N6O4 [M + H]+: 389.1937, found 389.1925.
4-((1-Methyl-5-(5-nitrofuran-2-yl)-3-propyl-1H-pyrazolo[4,3-d]pyrimidin-7-yl)amino)butan-1-ol ( 26 )
Yellow solid; m.p.: 154.5–156.5 °C; 1H NMR (400 MHz, CDCl3) δ 7.42 (d, J = 3.7 Hz, 1H), 7.30 (d, J = 3.8 Hz, 1H), 5.89 (t, J = 5.0 Hz, 1H), 4.25 (s, 3H), 3.83–3.78 (m, 4H), 2.96 (t, J = 7.6 Hz, 2H) 1.94–1.75 (m, 7H), 1.03 (t, J = 7.4 Hz, 3H); 13C NMR (101 MHz, DMSO) δ 154.93, 151.49, 149.60, 147.72, 144.38, 141.03, 120.96, 114.75, 113.57, 60.52, 40.39, 39.13, 29.97, 27.12, 25.19, 21.49, 13.82; HRMS (ESI) calcd for C17H23N6O4 [M + H]+: 375.1781, found 375.1771.
3-((1-Methyl-5-(5-nitrofuran-2-yl)-3-propyl-1H-pyrazolo[4,3-d]pyrimidin-7-yl)amino)propan-1-ol ( 27 )
Yellow solid; m.p.: 181–183 °C; 1H NMR (400 MHz, CDCl3) δ 7.42 (s, 2H), 6.54 (s, 1H), 4.26 (s, 3H), 3.97–3.89 (m, 4H), 2.96 (t, J = 7.6 Hz, 2H), 2.01 (m, 2H), 1.90–1.80 (m, 2H), 1.02 (t, J = 7.4 Hz, 3H); 13C NMR (101 MHz, DMSO) δ 154.93, 151.48, 149.60, 147.71, 144.38, 140.98, 120.97, 114.72, 113.60, 59.10, 39.05, 38.40, 31.48, 27.11, 21.46, 13.80; HRMS (ESI) calcd for C16H21N6O4 [M + H]+: 361.1624, found 361.1620.
2-((1-Methyl-5-(5-nitrofuran-2-yl)-3-propyl-1H-pyrazolo[4,3-d]pyrimidin-7-yl)amino)ethanol ( 28 )
Orange-colored solid; m.p.: 204–206 °C; 1H NMR (400 MHz, DMSO) δ 7.81 (d, J = 3.8 Hz, 1H), 7.43 (s, 1H), 7.39 (d, J = 3.8 Hz, 1H), 4.86 (t, J = 5.6 Hz, 1H), 4.22 (s, 3H), 3.69 (s, 2H), 2.84 (t, J = 7.5 Hz, 2H), 1.83–1.72 (m, 2H), 0.95 (t, J = 7.4 Hz, 3H); 13C NMR (126 MHz, DMSO) δ 154.86, 151.52, 149.72, 147.62, 144.33, 141.03, 120.99, 114.78, 113.68, 58.96, 43.12, 39.18, 27.11, 21.52, 13.83; HRMS (ESI) calcd for C15H19N6O4 [M + H]+: 347.1468, found 347.1457.
2-(1-(1-Methyl-5-(5-nitrofuran-2-yl)-3-propyl-1H-pyrazolo[4,3-d]pyrimidin-7-yl)piperidin-4-yl)ethanol ( 29 )
Yellow solid; m.p.: 195–197 °C; 1H NMR (400 MHz, CDCl3) δ 7.43 (d, J = 3.7 Hz, 1H), 7.31 (d, J = 3.7 Hz, 1H), 4.12–4.06 (m, 5H), 3.78 (t, J = 6.3 Hz, 2H), 3.13–3.06 (m, 2H), 3.0 (t, J = 7.6 Hz, 2H), 1.99–1.79 (m, 5H), 1.65–1.59 (m, 2H), 1.52–1.45 (m, 2H), 1.05 (t, J = 7.4 Hz, 3H); 13C NMR (101 MHz, DMSO) δ 154.34, 153.68, 151.60, 147.04, 145.85, 143.29, 123.74, 114.71, 113.86, 58.19, 49.29, 39.05, 38.99, 31.96, 31.37, 27.18, 21.32, 13.84; HRMS (ESI) calcd for C20H27N6O4 [M + H]+: 415.2094, found 415.2083.
(1-(1-Methyl-5-(5-nitrofuran-2-yl)-3-propyl-1H-pyrazolo[4,3-d]pyrimidin-7-yl)piperidin-4-yl)methanol ( 30 )
Yellow solid; m.p.: 225–227 °C; 1H NMR (400 MHz, CDCl3) δ 7.43 (d, J = 3.6 Hz, 1H), 7.31 (d, J = 3.6 Hz, 1H), 4.11 (m, 5H), 3.62 (d, J = 5.9 Hz, 2H), 3.16–3.05 (m, 2H), 3.01 (t, J = 7.6 Hz, 2H), 2.02–1.80 (m, 5H), 1.58–1.47 (m, 3H), 1.05 (t, J = 7.3 Hz, 3H); 13C NMR (126 MHz, DMSO) δ 154.31, 153.72, 151.65, 147.06, 145.83, 143.27, 123.76, 114.80, 113.96, 65.55, 49.05, 39.10, 38.12, 28.11, 27.19, 21.39, 13.88; HRMS (ESI) calcd for C19H25N6O4 [M + H]+: 401.1937, found 401.1923.
3-(4-(1-Methyl-5-(5-nitrofuran-2-yl)-3-propyl-1H-pyrazolo[4,3-d]pyrimidin-7-yl)piperazin-1-yl)phenol ( 31 )
Orange color solid; m.p.: 225–227 °C; 1H NMR (400 MHz, CDCl3) δ 7.44 (d, J = 3.7 Hz, 1H), 7.33 (d, J = 3.7 Hz, 1H), 7.17–7.13 (m, 1H), 6.58 (dd, J = 8.2, 1.8 Hz, 1H), 6.48 (d, J = 1.8 Hz, 1H), 6.39 (dd, J = 8.2, 1.8 Hz, 1H), 4.17 (s, 3H), 3.76 (t, J = 4.4 Hz, 4H), 3.42 (t, J = 4.4 Hz, 4H), 3.03 (t, J = 7.6 Hz, 2H), 1.94–1.85 (m, 2H), 1.06 (t, J = 7.4 Hz, 3H); 13C NMR (126 MHz, DMSO) δ 158.12, 154.15, 153.40, 152.18, 151.67, 146.96, 145.91, 143.46, 129.62, 123.63, 114.76, 114.07, 106.85, 106.64, 102.78, 48.80, 47.85, 39.26, 27.19, 21.36, 13.87; HRMS (ESI) calcd for C23H26N7O4 [M + H]+: 464.2046, found 464.2033.
2-(4-(1-Methyl-5-(5-nitrofuran-2-yl)-3-propyl-1H-pyrazolo[4,3-d]pyrimidin-7-yl)piperazin-1-yl)phenol ( 32 )
Yellow solid; m.p.: 221–223 °C; 1H NMR (400 MHz, CDCl3) δ 7.44 (d, J = 3.7 Hz, 1H), 7.34 (d, J = 3.7 Hz, 1H), 7.21 (d, J = 7.8 Hz, 1H), 7.17–7.11 (m, 1H), 7.00 (d, J = 8.0 Hz, 1H), 6.96–6.87 (m, 2H), 4.19 (s, 3H), 3.80 (t, J = 4.4 Hz, 4H), 3.14 (t, J = 4.4 Hz, 4H) 3.03 (t, J = 7.6 Hz, 2H) 1.95–1.85 (m, 2H), 1.06 (t, J = 7.3 Hz, 3H); 13C NMR (126 MHz, DMSO) δ 154.21, 153.45, 151.67, 150.15, 147.00, 145.91, 143.43, 139.39, 123.65, 123.25, 119.40, 118.87, 115.64, 114.77, 114.10, 49.64, 49.17, 39.26, 27.20, 21.38, 13.88; HRMS (ESI) calcd for C23H26N7O4 [M + H]+: 464.2046, found 464.2036.
2-((1-Methyl-5-(5-nitrofuran-2-yl)-3-propyl-1H-pyrazolo[4,3-d]pyrimidin-7-yl)amino)phenol ( 33 )
Light orange color solid; m.p.: 212–214 °C; 1H NMR (500 MHz, DMSO) δ 8.33 (d, J = 7.9 Hz, 1H), 7.78 (d, J = 3.8 Hz, 1H), 7.22 (d, J = 3.8 Hz, 1H), 7.19–6.81 (m, 3H), 4.36 (s, 3H), 2.88 (t, J = 7.5 Hz, 2H), 1.85–1.79 (m, 2H), 0.99 (t, J = 7.3 Hz, 3H); 13C NMR (126 MHz, DMSO) δ 154.35, 151.56, 148.31, 147.29, 147.27, 144.44, 141.74, 126.39, 124.61, 122.41, 121.10, 119.15, 114.83, 114.72, 113.64, 39.88, 27.10, 21.49, 13.87; mass ESI [M + H]+: 395.1.
3-((1-Methyl-5-(5-nitrofuran-2-yl)-3-propyl-1H-pyrazolo[4,3-d]pyrimidin-7-yl)amino)phenol ( 34 )
Orange-colored solid; m.p.: 227–229 °C; 1H NMR (400 MHz, acetone d6) δ 7.64 (d, J = 3.8 Hz,1H) 7.50–7.48 (m, 1H), 7.39–7.34 (m, 2H), 7.246–7.22 (m, 1H), 6.97 (dd, J = 2.02, 8.08 Hz, 1H), 4.45 (s, 3H) 2.94 (t, J = 7.6 Hz, 2H), 1.93–1.83 (m, 2H), 1.02 (t, J = 7.3 Hz, 3H); 13C NMR (126 MHz, DMSO) δ 157.48, 154.30, 151.62, 147.39, 147.13, 144.75, 142.36, 139.61, 129.14, 121.33, 114.76, 113.90, 112.87, 111.04, 109.07, 39.31, 27.09, 21.48, 13.84; HRMS (ESI) calcd for C19H19N6O4 [M + H]+: 395.1468, found 395.1455.
4-((1-Methyl-5-(5-nitrofuran-2-yl)-3-propyl-1H-pyrazolo[4,3-d]pyrimidin-7-yl)amino)phenol ( 35 )
Yellow solid; m.p.: 251–253 °C; 1H NMR (400 MHz, acetone d6) δ 7.66 (d, J = 9.2 Hz, 2H), 7.6 (d, J = 4 Hz, 1H), 7.25 (d, J = 4 Hz, 1H), 6.93 (d, J = 9.2 Hz, 2H), 4.4 (s, 3H), 2.93 (t, J = 7.6 Hz, 2H), 1.91–1.82 (m, 2H), 1.01 (t, J = 7.2 Hz, 3H); 13C (126 MHz, acetone) δ 156.06, 155.36,152.94, 149.03, 148.73, 146.19, 143.76, 131.27, 125.41, 122.11, 115.91, 114.47, 114.06, 39.76, 29.36, 22.35, 14.26; HRMS (ESI) calcd for C19H19N6O4 [M + H]+: 395.1468, found 395.1463.
N-(4-Methoxyphenyl)-1-methyl-5-(5-nitrofuran-2-yl)-3-propyl-1H-pyrazolo[4,3-d]pyrimidin-7-amine ( 36 )
Orange-colored solid; m.p.: 152–154 °C; 1H NMR (400 MHz, CDCl3) δ 7.63 (d, J = 9.0 Hz, 2H), 7.39 (d, J = 3.8 Hz, 1H), 7.22 (d, J = 3.7 Hz, 1H), 6.99 (d, J = 9.0 Hz, 2H), 6.92 (s, 1H), 4.31 (s, 3H), 3.86 (s, 3H), 2.94 (t, J = 7.6 Hz, 2H), 1.94–1.82 (m, 2H), 1.05 (t, J = 7.4 Hz, 3H). 13C NMR (126 MHz, CDCl3) δ 156.95, 154.68, 151.98, 148.05, 147.55, 146.73, 143.37, 130.63, 123.33, 121.02, 114.30, 113.34, 113.14, 55.59, 39.36, 27.64, 22.08, 14.09; HRMS (ESI) calcd for C20H21N6O4 [M + H]+: 409.1624, found 409.1613.
1-Methyl-5-(5-nitrofuran-2-yl)-3-propyl-N-(4-(trifluoromethoxy)phenyl)-1H-pyrazolo[4,3-d]pyrimidin-7-amine ( 37 )
Yellow solid; m.p.: 198–200 °C; 1H NMR (400 MHz, CDCl3) δ 7.83 (d, J = 8.6 Hz, 2H), 7.42 (d, J = 4 Hz, 1H), 7.34 (d, J = 8.6 Hz, 2H), 7.26 (d, J = 4 Hz, 1H), 7.03 (s, 1H), 4.37 (s, 3H), 3.02 (t, J = 7.6 Hz, 2H), 1.96–1.87 (m, 2H), 1.05 (t, J = 7.2 Hz, 3H); 13C NMR (126 MHz, DMSO) δ 154.06, 151.57, 147.10, 146.90, 144.81, 144.01, 142.54, 137.81, 123.60, 121.16, 121.11, 119.15, 114.66, 113.77, 39.33, 27.09, 21.42, 13.82; HRMS (ESI) calcd for C20H18F3N6O4 [M + H]+: 463.1342, found 463.1332.
N-(3-Fluorophenyl)-1-methyl-5-(5-nitrofuran-2-yl)-3-propyl-1H-pyrazolo[4,3-d]pyrimidin-7-amine ( 38 )
Yellow solid; m.p.: 179–181 °C; 1H NMR (400 MHz, CDCl3) δ 7.85–7.82 (m, 1H), 7.41 (d, J = 4 Hz, 1H), 7.39–7.34 (m, 2H), 7.27 (d, J = 4 Hz, 1H), 7.06 (s, 1H), 6.93–6.88 (m, 1H), 4.36 (s, 3H), 2.99 (t, J = 7.5 Hz, 2H), 1.93–1.84 (m, 2H), 1.04 (t, J = 7.3 Hz, 3H); 13C NMR (126 MHz, acetone) δ 164.60, 162.68, 155.55, 148.41, 148.21, 146.36, 144.32, 141.50, 130.89, 122.15, 118.33, 114.54, 114.13, 111.18, 109.74, 39.78, 28.25, 22.68, 14.27; HRMS (ESI) calcd for C19H18FN6O3 [M + H]+: 397.1424, found 397.1417.
N-(4-Fluorophenyl)-1-methyl-5-(5-nitrofuran-2-yl)-3-propyl-1H-pyrazolo[4,3-d]pyrimidin-7-amine ( 39 )
Yellow solid; m.p.: 201–203 °C; 1H NMR (400 MHz, CDCl3) δ 7.72 (m, 2H), 7.41 (d, J = 3.7 Hz, 1H), 7.23 (d, J = 3.7 Hz, 1H), 7.18 (d, J = 8.5 Hz, 2H), 6.98 (s, 1H), 4.36 (s, 3H), 3.00 (t, J = 7.7 Hz, 2H), 1.95–1.84 (m, 2H), 1.05 (t, J = 7.4 Hz, 3H). 13C NMR (126 MHz, acetone) δ 161.20, 155.73, 148.61, 146.29, 143.99, 135.79, 125.31, 125.20, 122.03, 115.96, 115.78, 114.49, 114.17, 39.79, 28.27, 22.67, 14.29; HRMS (ESI) calcd for C19H18FN6O3 [M + H]+: 397.1424, found 397.1415.
N-(4-Chlorophenyl)-1-methyl-5-(5-nitrofuran-2-yl)-3-propyl-1H-pyrazolo[4,3-d]pyrimidin-7-amine ( 40 )
Orange-colored solid; m.p.: 209–211 °C; 1H NMR (400 MHz, CDCl3) δ 7.74 (d, J = 8.9 Hz, 2H), 7.42 (m, 3H), 7.24 (d, J = 3.8 Hz, 1H), 6.98 (s, 1H), 4.36 (s, 3H), 2.99 (t, J = 7.6 Hz, 2H), 1.93–1.84 (m, 2H), 1.04 (t, J = 7.2 Hz, 4H); 13C NMR (101 MHz, CDCl3) δ 154.41, 152.07, 147.89, 146.95, 146.84, 143.89, 136.41, 129.80, 129.20, 122.36, 120.90, 113.19, 113.05, 39.30, 27.63, 21.98, 14.03. HRMS (ESI) calcd for C19H18ClN6O3 [M + H]+: 413.1129, found 413.1119.
N-(4-Bromophenyl)-1-methyl-5-(5-nitrofuran-2-yl)-3-propyl-1H-pyrazolo[4,3-d]pyrimidin-7-amine ( 41 )
Orange-colored solid; m.p.: 212–214 °C; 1H NMR (400 MHz, CDCl3) δ 7.70 (d, J = 8.8 Hz, 2H), 7.58 (d, J = 8.8 Hz, 2H), 7.42 (d, J = 3.7 Hz, 1H), 7.25 (mer with CDCl3, 1H), 6.97 (s, 1H), 4.37 (s, 3H), 3.00 (t, J = 7.6 Hz, 2H), 1.94–1.84 (m, 2H), 1.05 (t, J = 7.3 Hz, 3H); 13C NMR (126 MHz, CDCl3) δ 154.73, 152.40, 148.20, 147.27, 147.11, 144.19, 137.26, 132.50, 122.99, 121.25, 117.69, 113.65, 113.49, 39.71, 28.00, 22.39, 14.44; HRMS (ESI) calcd for C19H18BrN6O3 [M + H]+: 457.0624, found 457.0610.
1-Methyl-5-(5-nitrofuran-2-yl)-3-propyl-N-(4-(trifluoromethyl)phenyl)-1H-pyrazolo[4,3-d]pyrimidin-7-amine ( 42 )
Yellow solid; m.p.: 216–218 °C; 1H NMR (400 MHz, CDCl3) δ 7.98 (d, J = 8.4 Hz, 2H), 7.74 (d, J = 8.4 Hz, 2H), 7.44 (d, J = 3.7 Hz, 1H), 7.31 (d, J = 3.7 Hz, 1H), 7.16 (s, 1H), 4.42 (s, 3H), 3.02 (t, J = 7.6 Hz, 2H), 1.95–188 (m, 2H), 1.07 (t, J = 7.4 Hz, 3H); 13C NMR (101 MHz, DMSO) δ 153.92, 151.61, 146.80, 144.94, 142.89, 142.50, 125.63, 123.57, 123.26, 123.09, 121.53, 121.30, 114.64, 113.91, 39.31, 27.08, 21.38, 13.80; HRMS (ESI) calcd for C20H18F3N6O3 [M + H]+: 447.1392, found 447.1382.
1-Methyl-5-(5-nitrofuran-2-yl)-3-propyl-N-(pyridin-4-yl)-1H-pyrazolo[4,3-d]pyrimidin-7-amine ( 43 )
4-Aminopyridine (50 mg, 0.53 mmol 1 eq.) was dissolved in dry DMF (2 ml) and to it was added K2CO3 (220 mg 1.59 mmol 3q); then, 7-chloro-1-methyl-5-(5-nitrofuran-2-yl)-3-propyl-1H-pyrazolo[4,3-d] pyrimidine (170 mg, 0.53 mmol, 1 eq.) was added to the mixture. The reaction mixture was stirred for 3–4 h at 80 °C. After completion, 20 ml EtOAc and 30 ml water were added and the organic layer was separated. The water layer was re-extracted with EtOAc (2 × 20 ml) and the combined organic layers were washed with brine solution, dried over anhydrous sodium sulfate, and concentrated. The residue was purified by column chromatography. Compound 43 was obtained as an orange-colored solid.
Orange-colored solid; m.p.: 265–267 °C; 1H NMR (400 MHz, CDCl3) δ 8.65 (d, J = 4.3 Hz, 2H), 7.83 (d, J = 4.3 Hz, 2H), 7.46 (d, J = 3.8 Hz, 1H), 7.35 (d, J = 3.8 Hz, 1H), 4.41 (s, 3H), 3.02 (t, J = 7.6 Hz, 2H), 1.95–1.85 (m, 2H), 1.05 (t, J = 7.3 Hz, 3H); 13C NMR (101 MHz, DMSO) δ 153.91, 151.67, 149.46, 146.80, 144.99, 143.17, 121.75, 115.06, 114.68, 113.89, 39.28, 27.08, 21.39, 13.81; HRMS (ESI) calcd for C18H18N7O3 [M + H]+: 380.1471, found 380.1461.
1-Methyl-5-(5-nitrofuran-2-yl)-3-propyl-N-(pyridin-4-ylmethyl)-1H-pyrazolo[4,3-d] pyrimi-din-7-amine ( 44 )
Yellow solid; m.p.: 218–220 °C; 1H NMR (400 MHz, DMSO) δ 8.51 (d, J = 5.0 Hz, 2H), 8.24 (t, J = 5.7 Hz, 1H), 7.74 (d, J = 3.9 Hz, 1H), 7.55 (d, J = 5.0 Hz, 2H), 7.19 (d, J = 3.9 Hz, 1H), 4.82 (d, J = 5.6 Hz, 1H), 4.29 (s, 3H), 2.84 (t, J = 7.5 Hz, 1H), 1.84–1.70 (m, 2H), 0.95 (t, J = 7.4 Hz, 3H).13C NMR (126 MHz, DMSO) δ 154.49, 151.50, 149.31, 149.16, 148.89, 147.38, 144.44, 141.28, 122.72, 120.90, 114.69, 113.60, 43.16, 39.23, 27.10, 21.53, 13.83; HRMS (ESI) calcd for C19H20N7O3 [M + H]+: 394.1628, found 394.1620.
1-Methyl-5-(5-nitrofuran-2-yl)-3-propyl-N-(pyridin-3-ylmethyl)-1H-pyrazolo[4,3-d]pyrimidin-7-amine ( 45 )
Yellow solid; m.p. 243–245 °C; 1H NMR (400 MHz, DMSO) δ 8.77 (s, 1H), 8.43 (d, J = 3.7 Hz, 1H), 8.19 (t, J = 5.7 Hz, 1H), 7.96 (d, J = 7.8 Hz, 1H), 7.77 (d, J = 3.8 Hz, 1H), 7.33 (dd, J = 7.7, 4.8 Hz, 1H), 7.29 (d, J = 3.8 Hz, 1H), 4.80 (d, J = 5.6 Hz, 2H), 4.26 (s, 3H), 2.83 (t, J = 7.5 Hz, 2H), 1.82–1.70 (m, 2H), 0.94 (t, J = 7.4 Hz, 3H); 13C NMR (101 MHz, DMSO) δ 154.61, 151.49, 149.33, 149.12, 147.98, 147.42, 144.42, 141.23, 135.63, 134.90, 123.33, 120.89, 114.69, 113.53, 41.69, 39.24, 27.10, 21.48, 13.80; HRMS (ESI) calcd for C19H20N7O3 [M + H]+: 394.1628, found 394.1626.
1-Methyl-5-(5-nitrofuran-2-yl)-3-propyl-7-(4-(pyridin-4-yl)piperazin-1-yl)-1H-pyrazolo[4,3-d]pyrimidine ( 46 )
Yellow solid; m.p.: 249–251 °C; 1H NMR (400 MHz, CDCl3) δ 8.33 (d, J = 4.2 Hz, 2H), 7.44 (d, J = 3.8 Hz, 1H), 7.34 (d, J = 3.8 Hz, 1H), 6.80 (d, J = 5.8 Hz, 2H), 4.19 (s, 3H), 3.83–3.74 (m, 4H), 3.68–3.60 (m, 4H), 3.02 (t, J = 7.6 Hz, 2H), 1.96–1.84 (m, 2H), 1.06 (t, J = 7.4 Hz, 3H). 13C NMR (126 MHz, CDCl3) δ 154.84, 154.50, 153.74, 152.13, 150.49, 148.26, 147.77, 144.98, 124.38, 113.41, 113.21, 108.76, 49.02, 45.59, 38.89, 27.79, 22.02, 14.12; HRMS (ESI) calcd for C22H25N8O3 [M + H]+: 449.2050, found 449.2039.
The HepG2, RAW 264.7 and J774A.7 cells were acquired from the American Type Culture Collection (ATCC, Manassas, VA) and cultured in Dulbecco's modified Eagle's medium (DMEM, Gibco, Life Technologies, NY) supplemented with 10% fetal bovine serum (FBS) at 37 °C and 5% CO2. MTT dye (HiMedia) was used to detect cell viability. The antibiotics ciprofloxacin hydrochloride monohydrate (HiMedia), vancomycin (HiMedia), and rifampicin (HiMedia) were used in different assays.
The minimum bactericidal concentration is the lowest concentration at which complete killing of bacterial cells is observed. The MBC was determined using 10 μl of the suspensions from the wells of the MIC plate where no growth was visible on the MHA plate. The agar plates were incubated for 24 h and the growth of bacterial colony was visualized in the respective concentrations.
The fraction inhibitory concentration index (FICI) is used to categorize the values for the interaction of two antibiotics. It is calculated by using the following formulas:
FICI = FIC of identified compound + FIC of standard drug |
Compound 35 was tested further using RAW 264.7 and J774A.1 macrophage/monocyte cells with changed incubation treatment. The cells were plated at a density of 1 × 104 cells per well in a 96-well flat-bottom plate in DMEM containing 10% FBS and incubated for 48 h and 72 h (37 °C; 5% CO2). The treated cells were then analyzed using OD at 570 nm.
Footnotes |
† Electronic supplementary information (ESI) available. See DOI: https://doi.org/10.1039/d4md00826j |
‡ Equal contribution as first author to this work. |
This journal is © The Royal Society of Chemistry 2025 |