Next Article in Journal
A Comprehensive Review on the Phytochemical Constituents and Pharmacological Activities of Pogostemon cablin Benth.: An Aromatic Medicinal Plant of Industrial Importance
Previous Article in Journal
Isoindolinones as Michael Donors under Phase Transfer Catalysis: Enantioselective Synthesis of Phthalimidines Containing a Tetrasubstituted Carbon Stereocenter
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Microwave-Assisted Synthesis of Novel Pyrazolo[3,4-g][1,8]naphthyridin-5-amine with Potential Antifungal and Antitumor Activity

1
Heterocyclic Compounds Research Group, Department of Chemistry, Universidad del Valle, A.A. 25360 Cali, Colombia
2
Farmacognosia, Facultad de Ciencias Bioquímicas y Farmacéuticas, Universidad Nacional de Rosario, Suipacha 531, CP 2000 Rosario, Argentina
*
Author to whom correspondence should be addressed.
Molecules 2015, 20(5), 8499-8520; https://doi.org/10.3390/molecules20058499
Submission received: 17 April 2015 / Revised: 5 May 2015 / Accepted: 6 May 2015 / Published: 12 May 2015
(This article belongs to the Section Medicinal Chemistry)

Abstract

:
The microwave assisted reaction between heterocyclic o-aminonitriles 1 and cyclic ketones 2 catalyzed by zinc chloride led to new series of pyrazolo[3,4-b][1,8]naphthyridin-5-amines 3 in good yields. This procedure provides several advantages such as being environmentally friendly, high yields, simple work-up procedure, broad scope of applicability and the protocol provides an alternative for the synthesis of pyrazolonaphthyridines. The whole series showed antifungal activities against Candida albicans and Cryptococcus neoformans standardized strains, being compounds with a 4-p-tolyl substituent of the naphthyridin scheleton (3a, 3d and 3g), the most active ones mainly against C. albicans, which appear to be related to their comparative hydrophobicity. Among them, 3d, containing a cyclohexyl fused ring, showed the best activity. The anti-Candida activity was corroborated by testing the three most active compounds against clinical isolates of albicans and non-albicans Candida strains. These compounds were also screened by the US National Cancer Institute (NCI) for their ability to inhibit 60 different human tumor cell lines. Compounds 3a and 3e showed remarkable antitumor activity against cancer cell lines, with the most important GI50 values ranging from 0.62 to 2.18 μM.

Graphical Abstract

1. Introduction

Naphthyridines are fused nitrogen heterocycles present in many natural and synthetic compounds of particular interest in Medicinal Chemistry due to their diverse biological activities. They have showed a broad range of interesting pharmacological activities, such as anti-inflammatory [1,2,3,4], analgesic, antiaggressive [5], anticancer [6], antibacterial [7], antitumor [8], antihypertensive [9] and antiallergic [10] ones. They also showed to be useful starting materials for the synthesis of various policyclic heterocycles of biological interest. Due to their biological and synthetic importance, the development of effective routes to synthesize naphthyridines continues to be an active area of research for synthetic organic chemists [11]. A survey of the literature shows that the major synthetic approaches used to prepare the naphthyridine system involved condensation of 2-aminopyridine derivatives with carbonyl compounds containing an activated methylene group [12,13,14,15,16,17,18,19] or with β-ketoesters [20].
Microwave irradiation (MWI) is a technique that has been employed in a number of applications in synthetic chemistry and has been observed to have several advantages compared to traditional methods of synthesis. Due to selective heating in the microwave, the occurrence of side reactions is avoided. Microwave assisted organic synthesis (MAOS) has also emerged as a powerful tool for high-throughput procedures. This can improve the yield and purity of the final compounds in short reaction times through the precise control of parameters such as power irradiation, pressure and temperature [21,22,23,24,25,26,27].
On the other hand, the Friedländer annulation is one of the most simple and straightforward approaches to the synthesis of poly-substituted pyridines and related aza-heterocycles or aza-aromatic compounds [28,29,30]. For this kind of Friedländer reaction, two points are of concern. One is the catalyst, which belongs to one of two categories: the proton acid [31,32,33,34] or the Lewis acid [35,36,37,38,39]. The other point is that the structures of a normal Friedländer condensation product are either a pyridine or a quinoline skeleton.
Cancer is the major health problem that threatens people worldwide. Since many of the current pharmacotherapeutic drugs have problems with toxicity and drug resistance, there is a strong need for the discovery and development of effective new anticancer drugs [40]. Among the wide range of compounds tested as potential anticancer agents, compounds containing the six-membered heterocyclic pyridine and/or pyrimidine, have attracted significantly attention.
In the last years, fungi have emerged as major cause of human infections especially among immunocompromised hosts having an enormous impact on morbidity and mortality [1,2,3,4]. Because most patients with invasive fungal infections are immunocompromised, the success of treatments is more dependent on the efficacy of the antifungal agent than on the immune system [41,42]. Unfortunately, the available antifungal agents are limited and it took 30 years for the newest class of antifungal drugs, the echinocandins [43,44], to appear on the market. Furthermore, the usual therapy for cryptococcal meningitis, a disease produced by Cryptococcus neoformans that kills most AIDs patients worldwide, is based on amphotericin B and flucytosine, which were discovered nearly 50 years ago [45]. There is, therefore, an urgent need for new antifungal chemical structures for treating infections produced by these fungi, alternatives to the existing ones [46].
Because of this great need for new antifungal and/or anticancer structures, and considering that other naphthyridines-bearing structures have shown antimicrobial and antitumor activities [4,5], herein we report the synthesis of a novel series of pyrazolo[3,4-b][1,8]naphthyridin-5-amines by the Friedländer condensation of o-aminonitrile 1 with cyclic ketones 2. The whole series was tested for antifungal activity against standardized as well clinical strains of the clinically important fungi C. neoformans and species of Candida genus. All compounds were also evaluated to determine antitumor activity.

2. Results and Discussion

2.1. Chemistry

In our study, several conditions were tested for the first time, including diverse solvents, temperatures, heating source and catalysts in order to find the best reaction conditions for the synthesis of 3.
Scheme 1. Synthesis of pyrazolo[3,4-b][1,8]naphthyridin-5-amine 3.
Scheme 1. Synthesis of pyrazolo[3,4-b][1,8]naphthyridin-5-amine 3.
Molecules 20 08499 g003
The reactions were carried out from o-aminonitrile 1a (R = CH3) and cyclohexanone as a model reaction (Scheme 1). When ethanol was used as the solvent and the mixture was subjected to reflux, the desired product, 3d, was obtained in low yields (30%, 35% and 40%, entries 2, 3 and 4) after 10 h. In a modified protocol, the reaction was performed under MWI, obtaining significant improvements evidenced by better yields of the target product (40%, 45% and 75%, entries 6, 7 and 8) and shorter reaction times. As it is clearly observed in entries 1 and 5, the reaction did not take place without a catalyst. So, we performed the reaction with a Lewis acid catalyst. Results showed that both the aluminum chloride and the p-toluenesulfonic acid, in either conventional refluxing or MWI, favored the product of Friedländer condensation 3 (Table 1), but the yields were not very high, whereas anhydrous zinc chloride favored the formation of the product in higher yields in both conditions. The best catalyst, anhydrous zinc chloride, was therefore chosen as the catalyst for this new transformation.
Table 1. Optimization of reaction conditions for the synthesis of compounds 3d.
Table 1. Optimization of reaction conditions for the synthesis of compounds 3d.
EntriesCatalystConditionsYield (%)
1EtOH, reflux
2AlCl3EtOH, reflux30
3pMeC6H4SO3HEtOH, reflux35
4ZnCl2EtOH, reflux40
5EtOH, MW (120 °C, 300 W)
6AlCl3EtOH, MW (120 °C, 300 W)40
7pMeC6H4SO3HEtOH, MW (120 °C, 300 W)45
8ZnCl2EtOH, MW (120 °C, 300 W)75
To determine the extent of application of the cyclization reaction, the same conditions were used for some selected cyclic ketones (of five, six and seven members) to get the pyrazolo[3,4-b][1,8]naphthyridin-5-amines (Scheme 1, Table 2).
Table 2. Scope of the reaction.
Table 2. Scope of the reaction.
Compound 3StructureYield (%)
3a Molecules 20 08499 i00165
3b Molecules 20 08499 i00263
3c Molecules 20 08499 i00362
3d Molecules 20 08499 i00480
3e Molecules 20 08499 i00575
3f Molecules 20 08499 i00665
3g Molecules 20 08499 i00760
3h Molecules 20 08499 i00860
3i Molecules 20 08499 i00955
All the structures 3ai were characterized by IR (Infrared), 1H-NMR, 13C-NMR (Proton- and carbon-Nuclear Magnetic Resonance), MS (mass-spectrometry) spectra and elemental analyses. Formation of the pyrazolo[3,4-b][1,8]naphthyridin-5-amines 3 was unequivocally established by NMR data of the products. The chemical shifts and multiplicities of the protons were in accordance with the expected values. For example, signals for the protons of the phenyl of compounds 3 were found between 7.20 and 8.30 ppm. The signal for NH2 appears as a broad singlet between 6.10 and 6.80 ppm, the signals of methyl protons of CH3 appears as singlets between 1.74 and 1.84 ppm and all aliphatic protons, corresponding to the five-, six- and seven-membered, appear between 1.46 and 3.87.
A possible mechanism of the cyclization reaction between 1 and 2 is depicted in Scheme 2. The reaction seemed to proceed via the initial formation of the imine intermediate 5 by the normal Friedländer reaction [47,48], which subsequently affords the final compound 3 by an intramolecular nucleophilic cyclization on the nitrile.
Scheme 2. Proposed mechanism.
Scheme 2. Proposed mechanism.
Molecules 20 08499 g004

2.2. Antifungal Activity

The antifungal properties of compounds 3a3i were tested first against two clinically important fungal species, Candida albicans and C. neoformans, which were selected due to the following facts: C. albicans is among the most common cause of opportunistic fungal infections in immunocompromised hosts, although recently, non-albicans Candida species have been increasingly identified among Candida-infected patients [49].
In turn, C. neoformans is the most frequent cause of meningitis and is one of the most important HIV-related fatal opportunistic mycosis, which has killed more than 650,000 immunocompromised patients worldwide up to date [46]. Although the incidence of disease tends to decline in countries with highly active anti-retroviral therapy, the outcome of infection is influenced by a variety of factors including the antifungal resistance and new strategies including new structural types with anti-cryptococcal activity are highly welcome [50].
For a more comprehensive analysis of the antifungal results, we grouped the compounds in two series, (i) and (ii). Series (i) includes compounds with different rings (cyclopentyl, cyclohexyl or cycloheptyl) fused to the naphthyridines scheleton and the same R (sub-series i.1 with CH3, i.2 with Cl or i.3 with OCH3), which allowed having a look at the influence of the size of the fused ring on the antifungal activity. Series (ii) includes compounds with different R (CH3, Cl or OCH3), but the same fused ring moiety (subseries ii.1 with cyclopentyl; ii.2 with cyclohexyl and ii.3 with cycloheptyl), which allowed analyzing the role played by the different R substituents in the antifungal activity.
Compounds were evaluated by using the standardized microbroth dilution method M-27A3 for yeasts of Clinical and Laboratory Standards Institute [51], which assures confident and reproducible results.
Results of the whole series are expressed as the percentages of inhibition of each fungus in the range 250–3.9 µg∙mL−1 and are presented as Supplementary Table 1 (Table S1).
For the sake of clarity, Table S1 was summarized in Table 3 by using the MICs at different endpoints, such as MIC100 MIC80 and MIC50 (minimum concentration that inhibits 100%, 80% and 50% of growth) that have showed to consistently represent the in vitro activity of compounds [52].
From Table 3, it is clear that all compounds displayed some degree of activity against C. albicans and C. neoformans. However, C. albicans showed to be more sensitive for the whole series than C. neoformans, since eight of the nine compounds showed MIC100 against C. albicans below 250 µg∙mL−1 (range = 31.2–250 µg∙mL−1), while only four of the nine compounds showed MICs100 below 250 µg∙mL−1 against C. neoformans (range = 125–250 µg∙mL−1). The same analysis can be performed with MIC80 and MIC50.
Regarding the activity against C. albicans, the comparison of the activity of compounds with same ring and different R (compare 3a/3b/3c, 3d/3e/3f or 3g/3h/3i) showed that the type of ring does not play a crucial role in the activity, since the most active compounds, 3a, 3d and 3g, possess different rings, cyclopentyl, cyclohexyl and cycloheptyl, respectively. Instead, the three compounds share the feature of having a 4-p-tolyl moiety.
The comparison of the activity of the three compounds possessing a methyl groups as R can be clearly observed in Figure 1.
From Figure 1, it is clear that 3d, possessing a cyclohexyl ring, is the most active compound, while 3a and 3g (with cyclopentyl and cycloheptyl, respectively) possessed lower activities than 3d, but similar to each other.
Table 3. Minimum inhibitory concentrations (MIC100, MIC80 and MIC50) and minimum fungicidal concentrations (MFC) of 3ai grouped by their structural features against standardized strains of Candida albicans and C. neoformans. Molecules 20 08499 i010
Table 3. Minimum inhibitory concentrations (MIC100, MIC80 and MIC50) and minimum fungicidal concentrations (MFC) of 3ai grouped by their structural features against standardized strains of Candida albicans and C. neoformans. Molecules 20 08499 i010
R1Fused RingCompC. albicans ATCC 10231C. neoformans ATCC 32264
MIC100MIC80MIC50MIC100MIC80MIC50
A3a12562.562.52502507.8
CH3B3d31.231.231.2250125125
C3g12562.562.5125125125
A3b>250250125>250250250
ClB3e250125125>250>250250
C3h125125125250125125
A3c250250250>25025031.2
OCH3B3f250125125250125125
C3i250125125>250>250250
Amph B0.12 0.25
Figure 1. Comparative curves of the growth inhibition of C. albicans ATCC 10231 produced by compounds 3a, 3d and 3g at different concentrations. Inhibition percentages are the means ± SD obtained from experiments in triplicate.
Figure 1. Comparative curves of the growth inhibition of C. albicans ATCC 10231 produced by compounds 3a, 3d and 3g at different concentrations. Inhibition percentages are the means ± SD obtained from experiments in triplicate.
Molecules 20 08499 g001
It is known that logP (the logarithm of the partition coefficient in a biphasic system, e.g., n-octanol/water) describes the macroscopic hydrophobicity of a molecule, which is a factor that determines its ability to penetrate fungal cell membranes and to reach the interacting sites, thus influencing the antifungal activity of compounds [53,54]. In order to establish a correlation between logP and the activity of 3a–i, if any, logP of each compound was calculated and correlated to the percentage of inhibition of each compound at a selected concentration (125 µg∙mL−1). Table 4 shows the values of logP and Figure 2 plots logP vs. activity of all compounds of the series. For the calculation of logP, we used quantum mechanical at semi-empirical level using Mopac, with the parametric method 3 (PM3). The molecular modeling were prepared using CS Chem-Office Software version 9.0 (Cambridge software) [55]. The models were minimizedation until the root mean square (RMS) gradient value reached a value smaller than 0.0001 kcal∙mol−1. The lowest energy structure was used for each molecule to calculate logP values.
Table 4. In vitro activity of compounds 3ai expressed as % inhibition of Candida albicans (C.a.) ATCC 10231 growth at 125 µg∙mL−1.
Table 4. In vitro activity of compounds 3ai expressed as % inhibition of Candida albicans (C.a.) ATCC 10231 growth at 125 µg∙mL−1.
CompoundLog P% Ihn C.a.
3a5.61803100
3b3.5088443.63
3c3.7833435.58
3d5.87099100
3e3.0558156.19
3f4.3363156.35
3g6.42396100
3h6.60877100
3i3.8892750.55
Figure 2. LogP vs. inhibition percentage of C. albicans growth, by 3ai at 125 µg∙mL−1.
Figure 2. LogP vs. inhibition percentage of C. albicans growth, by 3ai at 125 µg∙mL−1.
Molecules 20 08499 g002
Interesting enough, the most active compounds, 3a, 3d, 3g and 3h, possess logP values between 5.62 and 6.61. The rest of these compounds possess values of logP lower than 4.8 (mean value). The results showed above suggest that the antifungal activity of compounds 3 would be related to their hydrophobicity.

Second-Order Studies with Clinical Isolates

In order to gain insight into the actual inhibitory capacity of 3a, 3d and 3g against C. albicans, the three compounds were tested not only against the ATCC standardized strain but also on six clinical strains of C. albicans (isolated from patients suffering from mycoses) and on four non-albicans Candida strains, such as C. glabrata, C. parapsilopsis, C. krusei and C. tropicalis, all of them provided by CEREMIC (see Experimental). The selection of these non-Candida spp. was due to that fact that these four spp, along with C. albicans, are responsible for more than 90% of all Candida infection all over the world and also in Latin American countries [49].
The minimum inhibitory concentration (MIC) values of 3a, 3d and 3g were determined against this new panel by determining MIC100, MIC80 and MIC50. These results are shown in Table 5.
Table 5. The 100%, 80% and 50% inhibitory concentrations (MIC100, MIC80 and MIC50) of 3a, 3d, and 3g against clinical isolates of C. albicans and non-albicans Candida strains. For the sake of comparison, MIC100, MIC80 and MIC50 of all compounds against the ATCC 10231 standardized strain of C. albicans are included.
Table 5. The 100%, 80% and 50% inhibitory concentrations (MIC100, MIC80 and MIC50) of 3a, 3d, and 3g against clinical isolates of C. albicans and non-albicans Candida strains. For the sake of comparison, MIC100, MIC80 and MIC50 of all compounds against the ATCC 10231 standardized strain of C. albicans are included.
StrainVoucher Specimen3a3d3gAmph.
MIC100MIC80MIC50MIC100MIC80MIC50MIC100MIC80MIC50MIC100
C. albicansATCC 1023112562.562.531.231.231.212562.562.51.00
C. albicansCCC 12562.562.562.531.231.231.212562.531.20.78
C. albicansCCC 12612562.562.531.231.215.612562.562.51.56
C. albicansCCC 12762.531.231.231.231.231.212562.531.20.78
C. albicansCCC 12812562.531.231.231.215.612562.562.51.56
C. albicansCCC 12912531.262.562.531.231.225062.531.20.78
C. albicansCCC 13012562.562.531.231.215.612531.231.20.50
C. glabrataCCC 115>250>250>250>250>250>250>250>2502500.39
C. parapsilopsisCCC 12412562.562.531.231.231.212512562.50.78
C. kruseiCCC 11712512562.515.615.67.812562.562.50.39
C. tropicalisCCC 13112512562.531.215.615.612512562.50.50
ATCC = American Type Culture Collection (Illinois, USA); CCC = Center of Mycological Reference (Rosario, Argentina), C. albicans = Candida albicans; C. glabrata = Candida glabrata; C. parapsilopsis = Candida parapsilopsis; C. krusei = Candida krusei; C. tropicalis = Candida tropicalis; Amph. = Amphotericin B.
As it can be seen in Table 5, clinical isolates of Candida genus showed similar sensitivity to 3a, 3d and 3g than the standardized strain C. albicans ATCC 10231, corroborating the anti-Candida activity of the three compounds.

2.3. Anticancer Activity

As a preliminary screening, structures of all new compounds (i.e., 3aj) were submitted to the Developmental Therapeutics Program (DTP) at National Cancer Institute (NCI) for evaluation of their anticancer activity against different human cell lines. All the submitted structures (3aj) were selected and subjected to the preliminary evaluation against the 60 tumor cell lines at a single dose of 10 mM after 48 h of incubation. The output from the single dose screening was reported as a mean graph available for analysis by the COMPARE program (data not shown). The results of this first assay showed that compounds 3a and 3e were active.
Then, the second screening was made in order to determine cytostatic activity of active compound against the 60 tumor cell lines represented in leukemia, melanoma, lung, colon, brain, breast, ovary, kidney and prostate panels; where the testing results were expressed according to the following three parameters: GI50, which is the molar concentration of the compounds required to inhibit the growing of the cell lines to 50% (relative to untreated cells). TGI as the molar concentration that causes total growth inhibition, and LC50, which is a parameter of cytotoxicity and reflects the molar concentration needed to kill 50% of the cells [56]. The active compounds were evaluated at five concentration levels (100, 10, 1.0, 0.1, and 0.01 mM) and the test consisted of a 48 h continuous drug exposure protocol using sulforhodamine B (SRB) protein assay to estimate cell growth. Details of this evaluation method, and the complementary information related with the activity pattern over all cell lines, have been published [57,58,59,60,61]. The compound 3a show a remarkable activity against 39 human tumor cell lines (Table 6), with low values of GI50 ≈ 10−6 μM, being SR (Leukemia; GI50 = 0.62 μM, LC50 > 100 μM), HCT-15 (Colon Cancer; GI50 = 1.98 μM, LC50 > 100 μM) and MCF7 (Breast Cancer; GI50 = 1.62 μM, LC50 > 100 μM) the most sensitive strains. In a similar way, compound 3e also showed an interesting activity against 41 human tumor cell lines, remarking that for SR and CCRF-CEM (Leukemia) with GI50 Values of 1.84, 2.18 μM and LC50 values of 79.5 μM and >100 μM, respectively. The cytotoxic effects associated with compounds 3a and 3e were measured as LC50 goes from 6.79 to >100 μM, indicating a low toxicity of these compounds for normal human cell lines, as required for development of potential antitumor agents.
Table 6. In vitro testing expressed as growth inhibition of cancer cell lines for compounds 3a and 3e a.
Table 6. In vitro testing expressed as growth inhibition of cancer cell lines for compounds 3a and 3e a.
Panel/Cell LineCompounds
3a3e
GI50 b (µM)LC50 c (µM)GI50 b (µM)LC50 c (µM)
Leukemia
CCRF-CEM2.56>1002.18>100
HL-60(TB)2.24>1002.128.71
K-5622.78>1002.3049.1
MOLT-43.17>1002.3042.4
RPMI-82262.25>1001.929.72
SR0.62>1001.8479.5
Non-small Cell Lung Cancer
A549/ATCC2.2240.23.0146.7
HOP-623.3258.714.160.9
HOP-92
NCI-H22621.6>10019.6>100
NCI-H2316.394.0
NCI-H322M3.2633.22.9233.1
NCI-H4601.928.643.2847.5
NCI-H52220.193.02.9156.0
Colon Cancer
COLO 2051.747.905.9131.3
HCC-29983.5153.8
HCT-1161.682.1525.9
HCT-151.98>1003.0855.7
HT2914.373.22.6841.9
KM124.8350.43.1640.0
SW-6202.4848.93.0550.3
CNS Cancer
SF-2683.1372.85.3354.6
SF-2952.9958.310.154.0
SF-5391.886.4914.558.8
SNB-193.4690.610.866.4
SNB-752.4346.514.072.1
U2511.786.851.727.36
Melanoma
LOX IMVI1.886.7912.060.8
MALME-3M21.574.5
M143.5059.03.7446.6
MDA-MB-43517.167.33.8353.4
SK-MEL-221.489.22.6335.3
SK-MEL-2811.362.66.7570.2
SK-MEL-51.7767.31.848.08
UACC-25714.361.62.4333.3
UACC-6213.756.8
Ovarian Cancer
IGROV14.4395.34.2681.5
OVCAR-32.069.474.1743.3
OVCAR-43.95>1003.2296.0
OVCAR-515.672.410.153.2
OVCAR-82.32>1002.8248.0
NCI/ADR-RES15.0>1003.41>100
SK-OV-319.961.812.150.3
Renal Cancer
786-02.034.5550.4
A49816.264.111.962.7
ACHN1.947.584.3246.3
CAKI-12.5631.53.8843.4
RXF 3938.4876.0
SN12C1.847.403.7255.9
TK-10
UO-311.847.332.4641.3
Prostate Cancer
PC-37.0567.32.5442.0
DU-1452.4017.54.7245.6
Breast Cancer
MCF71.62>1002.5774.0
MDA-MB-231/ATCC1.938.474.1547.5
HS 578T12.1>10012.5100
BT-54924.4>10017.266.5
T-47D2.21>100
MDA-MB-4682.0330.22.3340.5
a Data obtained from NCI’s in vitro disease-oriented human tumor cell lines screen [57]; b GI50 was the drug concentration resulting in a 50% reduction in the net protein increase (as measured by SRB staining) in control cells during the drug incubation. Determined at five concentration levels (100, 10, 1.0, 0.1, and 0.01 μM); c LC50 is a parameter of cytotoxicity and reflects the molar concentration needed to kill 50% of the cells.

3. Experimental Section

3.1. General Information

All the reagents were commercially available and used without any further purification. The solvents used were of analytical grade. Microwave experiments were carried out on a focused microwave reactor (300W CEM Discover). TLC analyses were performed on Merck TLC-plates aluminum silica gel 60 F254. Melting points were determined in a Büchi Melting Point Apparatus and are uncorrected. The IR analysis was performed on a Shimadzu FTIR 8400 spectrophotometer in KBr disks. 1H- and 13C-NMR spectra were run on a Bruker AVANCE 400 spectrometer operating at 400 MHz and 100 MHz, respectively, using dimethylsulfoxide-d6 as solvents and tetramethylsilane as internal standard. The mass spectra were scanned on a Shimadzu GCMS-QP 2010 and Hewlett-Packard HP Engine-5989 spectrometer (equipped with a direct inlet probe) and operating at 70 eV. The elemental analyses were obtained using a LECO CHNS-900 elemental analyzer and the values are within ± 0.4% of the theoretical values.

3.2. Chemistry

General Procedure for the Synthesis of Pyrazolo[4,3-g][1,8]naphthyridin-5-amine

All experiments were carried out using a focused microwave reactor (CEM Discover TM). A mixture of ortho-aminonitrile 1 ( 0.3 mmol), an excess of cyclic ketone 2 and ZnCl2 (10 mol %), were subjected to microwave irradiation, at 120 °C for 5–10 min and a maximum power of 300 W. Then, the solvent in the reaction mixture was removed under reduced pressure. Purification of products was performed using column chromatography in a mixture CHCl3/EtOH (20:1) as eluent.
3-Methyl-1-phenyl-4-(p-tolyl)-6,7-dihydro-8H-cyclopenta[g]pyrazolo[3,4-b][1,8]naphthyridin-5amine (3a). Yellow solid, yield 65%, mp 224–225. FTIR (KBr) ν(cm−1): 3446 (NH), 3030 (=C-H), 1620, 1569 (C=N and C=C). 1H-NMR (400 MHz, DMSO-d6): 1.86 (s, 3H), 2.21 (t, 2H, J = 7.2 Hz), 2.77 (s, 3H), 2.75 (t, 2H, J = 7.3 Hz), 3.22 (t, 2H, J = 7.6 Hz), 7.39 (t, 1H, J = 7.4 Hz), 7.56–7.64 (m, 6H), 8.30 (d, 2H, J = 7.8 Hz). Not observed (brs, 2H, NH2). 13C-NMR (100 MHz DMSO-d6) δ: 14.0 (CH3), 21.5 (CH3), 22.1 (CH2), 28.2 (CH2), 32.9 (CH2), 104.7 (C), 114.7 (C), 116.6 (C), 120.3 (CH), 126.2 (C), 128.0 (CH), 129.3 (CH), 130.2 (CH), 130.8 (CH), 138.2 (C), 140.4 (C), 145.3 (C), 147.1 (C), 148.1 (C), 149.2 (C), 156.5 (C), 161.1 (C). HR-MS calcd for C26H23N5 405, 1953, found [M+K]+ 443.9281. [M+H]+ 405.8378.
4-(4-Chlorophenyl)-3-methyl-1-phenyl-6,7-dihydro-8H-cyclopenta[g]pyrazolo[3,4b][1,8]naphthyridin-5-amine (3b). Yellow solid, yield 63%, mp 206–207. FTIR (KBr) ν(cm−1): 3467 (NH), 3035 (=C-H), 1622, 1568 (C=N and C=C).1H-NMR (400 MHz, DMSO-d6): 1.84 (s, 3H), 2.14–224 (m, 2H), 2.70–2.78 (m, 2H), 3.17–3.22 (m, 2H), 6.61 (brs, 2H, NH2), 7.37 (t, 1H, J = 7.4 Hz), 7.59 (t, 2H, J = 7.7 Hz), 7.73 (d, 2H, J = 8.0 Hz), 7.84 (d, 2H, J = 8.4 Hz), 8.30 (d, 2H, J = 6.7 Hz). 13C-NMR (100 MHz DMSO-d6) δ: 14.2 (CH3), 21.7 (CH2), 27.9 (CH2), 32.9 (CH2), 104.9 (C), 115.0 (C), 118.1 (C), 120.3 (CH), 126.1 (CH), 129.3 (CH), 129.8 (CH), 130.4 (CH), 132.9 (C), 135.5 (C), 138.3 (C), 144.9 (C), 145.4 (C), 147.5 (C), 149.1 (C), 156.4 (C), 160.1 (C). EI MS (70 eV): m/z: 425/427(M+, 1/0.35), 410/412(3/1), 236 (17). Anal. Calcd for C25H20ClN5: C, 70.50; H, 4.73; N, 16.44; found: C, 70.55; H, 4.68; N, 16.49.
4-(4-Methoxyphenyl)-3-methyl-1-phenyl-6,7dihydro-8H-cyclopenta[b]pyrazolo[3,4-g][1,8]naphthyridin-5-amine (3c). Yellow solid, yield 62%, mp 164–165. FTIR (KBr) ν(cm−1): 3450 (NH), 3035 (=C-H), 1625, 1565 (C=N and C=C). 1H-NMR (400 MHz, DMSO-d6): 1.78 (s, 3H), 2.09–2.20 (m, 2H), 2.67 (s, 2H), 3.19 (s, 2H,), 3.87 (s, 3H), 7.25 (d, 2H, J = 8.6 Hz), 7.31 (t, 1H, J = 7.1 Hz), 7.51 (s, 4H), 8.21 (d, 2H, J = 8.6 Hz). Not observed (brs, 2H, NH2). 13C-NMR (100 MHz DMSO-d6) δ: 14.4 (CH3), 21.8 (CH2), 28.1 (CH2), 32.8 (CH2), 56.0 (CH3), 105.4 (C), 109.8 (C), 115.3 (CH), 120.4 (C), 120.8 (CH), 125.4 (C), 126.1 (C), 126.4 (CH), 129.6 (CH), 130.0 (CH), 136.3 (C), 145.4 (C), 146.6 (C), 148.2 (C), 149.4 (C), 155.0 (C), 161.0 (C). m/z: 421 (M+, 1), 406 (3), 236 (10). Anal. Calcd for C26H23N5O: C, 74.09; H, 5.50; N, 16.62; found: C, 74.05; H, 5.56; N, 16.59.
3-Methyl-1-phenyl-4-(p-tolyl)-6,7,8,9-tetrahydro-1H-benzo[g]pyrazolo[3,4-b][1,8]naphthyridin-5-amine (3d). Yellow solid, yield 80%, mp 161-162. FTIR (KBr) ν(cm−1): 3444 (NH2), 2953 (=C-H), 1626, 1589 (C=N and C=C). 1H-NMR (400 MHz, DMSO-d6): 1.82 (s, 7H), 2.34 (s, 2H), 2.51 (s, 3H), 2.98 (s, 2H), 6.71 (brs, 2H, NH2), 7.36 (t, 1H, J = 7.4 Hz), 7.53–7.63 (m, 6H), 8.31 (d, 2H, J = 7.8 Hz). 13C-NMR (100 MHz DMSO-d6) δ: 14.5 (CH3), 21.2 (CH2), 21.5 (CH3), 21.6 (CH2), 23.0 (CH2) 29.7 (CH2), 104.8 (C), 109.3 (C), 116.9 (C), 120.5 (CH), 126.3 (C), 128.5 (CH), 129.7 (CH), 130.7 (CH), 131.7 (CH), 138.9 (C), 140.7 (C), 145.6 (C), 147.1 (C), 149.2 (C), 149.8 (C), 156.6 (C), 156.9 (C). HR-MS calcd for C27H25N5 419.2110, found. [M+H]+ 420.005.
4-(4-Chlorophenyl)-3-methyl-1-phenyl-6,7,8,9-tetrahydro-1H-benzo[g]pyrazolo[3,4-b][1,8]naphthyridin-5-amine (3e). Yellow solid, yield 75%, mp 180–181. FTIR (KBr) ν(cm−1): 3448 (NH2), 2960 (=C-H), 1628, 1579 (C=N and C=C).1H-NMR (400 MHz, DMSO-d6): 1.84 (s, 4H), 1.88 (s, 3H), 2.38 (s, 2H), 3.02 (s, 2H), 7.40 (t, 1H, J = 7.2 Hz), 7.62 (t, 2H, J = 7.4 Hz), 7.75 (d, 2H, J = 8.4 Hz), 7.87 (d, 2H, J = 8.4 Hz), 8.29 (d, 2H, J = 8.0 Hz). Not observed (brs, 2H, NH2). 13C-NMR (100 MHz DMSO-d6) δ: 14.3 (CH3), 20.4 (CH2), 20.9 (CH2), 22.4 (CH2) 28.1 (CH2), 104.0 (C), 109.1 (C), 116.9 (C), 120.4 (CH), 126.3 (CH), 129.4 (CH), 129.9 (CH), 130.3 (CH), 132.5 (C), 135.7 (C), 135.9 (C), 138.2 (C), 145.3 (C), 145.7 (C), 149.4 (C), 153.6 (C), 157.6 (C). m/z: 439/441 (M+,1/0.35), 424/426 (4/1.3), 236 (20). Anal. Calcd for C26H22ClN5: C, 70.98; H, 5.08; N, 15.92; found: C, 70.94; H, 5.12; N, 15.88.
4-(4-Methoxyphenyl)-3-methyl-1-phenyl-6,7,8,9-tetrahydro-1H-benzo[g]pyrazolo[3,4-b][1,8]naphthyridin-5-amine (3f). Yellow solid, yield 65%, mp 135–136. FTIR (KBr) ν(cm−1): 3450 (NH2), 2965 (=C-H), 1617, 1520 (C=N and C=C). 1H-NMR (400 MHz, DMSO-d6): 1.84 (s, 4H), 1.88 (s, 3H), 2.38 (s, 2H), 3.02 (s, 2H), 3.93 (s, 3H), 7.40 (t, 1H, J = 7.2 Hz), 7.62 (t, 2H, J = 7.4 Hz), 7.75 (d, 2H, J = 8.4 Hz), 7.87 (d, 2H, J = 8.4 Hz), 8.29 (d, 2H, J = 8.0 Hz). Not observed (brs, 2H, NH2). 13C-NMR (100 MHz DMSO-d6) δ: 14.2 (CH3), 20.3 (CH2), 20.9 (CH2), 22.3 (CH2) 27.9 (CH2), 55.6 (CH3), 104.2 (C), 108.9 (C), 115.3 (CH), 117.3 (CH), 120.3 (CH), 125.3 (C), 126.2 (C), 129.3 (CH), 129.7 (CH), 138.3 (C), 145.6 (C), 147.2 (C), 147.3 (C), 149.3 (C), 154.2 (C), 158.1 (C), 160.8 (C). HR-MS calcd for C27H25N5O 435.2059, found [M+H]+ 436.1918.
3-Methyl-1-phenyl-4-(p-tolyl)-6,7,8,9-tetrahydro-10H-cyclohepta[g]pyrazolo[3,4-b][1,8]naphthyridin-5-amine (3g). Yellow solid, yield 60%, mp 177–178. FTIR (KBr) ν(cm−1): 3489 (NH2), 3.040 (=C-H), 1630, 1569 (C=N and C=C). 1H-NMR (400 MHz, DMSO-d6): 1.46 (s, 2H), 1.68 (s, 2H), 1.74 (s, 3H), 1.77 (s, 2H), 2.46 (s, 3H), 2.63 (s, 2H), 3.11 (s, 2H), 7.33 (t, 1H, J = 7.4 Hz), 7.60–7.46 (m, 6H), 8.20 (d, 2H, J = 7.9 Hz). Not observed (brs, 2H, NH2). 13C-NMR (100 MHz DMSO-d6) δ: 14.7 (CH3), 21.8 (CH3), 24.9 (CH2), 25.8 (CH2), 26.3 (CH2), 31.3 (CH2), 33.8 (CH2), 100.3 (C), 105.3 (C), 114.9 (C), 117.8 (C), 121.0 (CH), 127.0 (CH), 128.7 (CH), 130.0 (CH), 131.0 (CH), 131.3 (C), 138.8 (C), 141.2 (C), 144.4 (C), 146.1 (C), 147.8 (C), 149.6 (C), 157.4 (C). m/z: 433 (M+,1), 424 (4/1.3), 236 (25). Anal. Calcd for C28H27N5: C, 77.57; H, 6.28; N, 15.16; found: C, 77.52; H, 6.33; N, 15.11.
4-(4-Chlorophenyl)-3-methyl-1-phenyl-6,7,8,9-tetrahydro-10H-cyclohepta[g]pyrazolo[3,4-b][1,8]naphthyridin-5-amine (3h). Yellow solid, yield 60%, mp 146–147. FTIR (KBr) ν(cm−1): 3480 (NH2), 3035 (=C-H), 1620, 1518 (C=N and C=C). 1H-NMR (400 MHz, DMSO-d6): 1.51–1.59 (m, 2H), 1.71–1.79 (m, 2H), 1.81–1.87 (m, 2H), 1.88 (s, 3H), 2.72–2.77 (m, 2H), 3.17–3.23 (m, 2H), 7.41 (t, 1H, J = 7.4 Hz), 7.63 (t, 2H, J = 8.0 Hz), 7.76 (d, 2H, J = 8.4 Hz), 7.86 (d, 2H, J = 8.4 Hz). 8.32 (d, 2H, J = 7.8 Hz). Not observed (brs, 2H, NH2). 13C-NMR (100 MHz DMSO-d6) δ: 14.4 (CH3), 24.3 (CH2), 25.3 (CH2), 25.9 (CH2), 30.8 (CH2), 42.6 (CH2), 104.2 (C), 109.3 (C), 117.1 (C), 120.5 (CH), 126.4 (CH), 129.5 (CH), 129.8 (CH), 130.5 (CH) 132.3 (C), 135.4 (C), 135.6 (C), 138.7 (C), 145.8 (C), 145.4 (C), 149.7 (C), 153.8 (C), 157.3 (C). m/z: 453/455 (M+,3/1), 438 (1/0.33), 342 (15), 236 (30). Anal. Calcd for C27H24ClN5: C, 71.43; H, 5.33; N, 15.43; found: C, 71.47; H, 5.28; N, 15.47.
4-(4-Methoxyphenyl)-3-methyl-1-phenyl-6,7,8,9-tetrahydro-10H-cyclohepta[g]pyrazolo[3,4-b][1,8]naphthyridin-5-amine (3i). Yellow solid, yield 55%, mp 125–126. FTIR (KBr) ν(cm−1): 3489 (NH2), 3038 (=C-H), 1618, 1525 (C=N and C=C). 1H-NMR (400 MHz, DMSO-d6): 1.48–1.53 (m, 2H), 1.68–1.75 (m, 2H), 1.77–1.85 (m, 5H), 2.66–2.72 (m, 2H), 3.13–3.20 (m, 2H), 3.89 (s, 3H), 7.30 (d, 2H, J = 8.6 Hz), 7.46 (t, 1H, J = 7.4 Hz), 7.54–7.62 (m, 4H), 8.25 (d, 2H, J = 8.1 Hz). Not observed (brs, 2H, NH2). 13C-NMR (100 MHz DMSO-d6) δ: 14.4 (CH3), 24.4 (CH2), 25.5 (CH2), 26.1 (CH2), 31.1 (CH2), 33.6 (CH2), 52.0 (CH3), 105.1 (C), 114.4 (C), 115.5 (CH), 117.9 (C), 120.8 (CH), 125.4 (C), 126.6 (CH), 129.6 (CH), 130.0 (CH), 138.5 (C), 145.8 (C), 147.5 (C), 147.6 (C), 149.5 (C), 152.9 (C), 157.4 (C), 161.0 (C). m/z: 449 (M+,1), 434 (3), 236 (10). Anal. Calcd for C28H27N5O: C, 74.81; H, 6.05; N, 15.58; found: C, 74.84; H, 6.01; N, 15.62.

3.3. Antifungal Activity

3.3.1. Microorganisms and Media

For the antifungal evaluation, standardized strains from the American Type Culture Collection (ATCC), Rockville, MD, USA, Reference Center in Mycology (CEREMIC, CCC, Rosario, Argentina) were used. Standardized strains: C. albicans ATCC 10231 and C. neoformans ATCC 32264; clinical isolates of Candida genus were provided by CCC. Voucher specimens of the isolated are presented in Table 5. Strains were grown on Sabouraud-chloramphenicol agar slants for 48 h at 30 °C, were maintained on slopes of Sabouraud-dextrose agar (SDA, Oxoid) and sub-cultured every 15 days to prevent pleomorphic transformations. Inocula were obtained according to reported procedures, [51] and adjusted to 1 × 103–5 × 103 cells with colony forming units (CFU)/mL.

3.3.2. Fungal Growth Inhibition Percentage Determination

Yeasts broth microdilution technique M27-A3 of CLSI [51] was performed in 96-well microplates. For the assay, compound test wells (CTWs) were prepared with stock solutions of each compound in DMSO (maximum concentration ≤ 1%), diluted with RPMI-1640, to final concentrations of 250–3.9 μg∙mL−1. An inoculum suspension (100 μL) was added to each well (final volume in the well = 200 μL). A growth control well (GCW) (containing medium, inoculum, and the same amount of DMSO used in a CTW, but compound-free) and a sterility control well (SCW) (sample, medium, and sterile water instead of inoculum) were included for each fungus tested. Microtiter trays were incubated in a moist, dark chamber at 30 °C for 48 h for both yeasts. Microplates were read in a VERSA Max microplate reader (Molecular Devices, Sunnyvale, CA, USA). Amphotericin B was used as positive control. Tests were performed in triplicate. Reduction of growth for each compound concentration was calculated as follows: % of inhibition = 100 − (OD 405 CTW − OD 405 SCW)/(OD 405 GCW − OD 405 SCW). The means ± SD (standard deviations) were used for constructing the dose-response curves representing % inhibition vs. concentration of each compound. Dose-response curves were constructed with SigmaPlot 11.0 software.

3.3.3. MIC100, MIC80 and MIC50 Determinations

Three endpoints were defined from the dose-response curves. Minimum Inhibitory concentration (MIC) resulting in total fungal growth inhibition was named MIC100, while MIC80 and MIC50 were defined as the minimum concentration that inhibits 80% or 50% of the fungal growth, respectively.

3.4. Anticancer Activity

The human tumor cell lines of the cancer-screening panel were grown in RPMI 1640 medium containing 5% fetal bovine serum and 2 mM L-glutamine. For a typical screening experiment, cells are inoculated into 96 well microtiter plates. After cell inoculation, the microtiter plates were incubated at 37 °C, 5% CO2 and 95% air, and 100% relative humidity for 24 h prior to addition of tested compounds. After 24 h, two plates of each cell line were fixed in situ with TCA, to represent a measurement of the cell population for each cell line at the time of sample addition (Tz). The samples were solubilized in dimethyl sulfoxide (DMSO) at 400-fold the desired final maximum test concentration and stored frozen prior to use. At the time of compounds addition, an aliquot of frozen concentrate was thawed and diluted to twice the desired final maximum test concentration with complete medium containing 50 µg∙mL−1 gentamicin. Additionally, four 10-fold or ½ log serial dilutions were made to provide a total of five drug concentrations plus control. Aliquots of 100 µL of these different sample dilutions were added to the appropriate microtiter wells already containing 100 µL of medium, resulting in the required final sample concentrations [58]. After the tested compounds were added, the plates were incubated for an additional 48 h at 37 °C, 5% CO2 and 95% air, and 100% relative humidity. For adherent cells, the assay was terminated by the addition of cold TCA. Cells were fixed in situ by the gentle addition of 50 µL of cold 50% (w/v) TCA (final concentration, 10% TCA) and incubated for 60 min at 4 °C. The supernatant was discarded, and plates were washed five times with tap water and air-dried. Sulforhodamine B (SRB) solution (100 µL) at 0.4% (w/v) in 1% acetic acid was added to each well, and plates were incubated for 10 min at room temperature. After staining, unbound dye was removed by washing five times with 1% acetic acid and the plates were air-dried. Bound stain was subsequently solubilized with 10 mM trizma base, and the absorbance was read on an automated plate reader at a wavelength of 515 nm. Using the seven absorbance measurements (time zero (Tz), control growth in the absence of drug (C), and test growth in the presence of drug at the five concentration levels (Ti)), the percentage growth was calculated at each of the drug concentrations levels. Percentage growth inhibition was calculated as: [(Ti − Tz)/(C − Tz)] × 100 for concentrations for which Ti > Tz, and [(Ti − Tz)/Tz] × 100 for concentrations for which Ti < Tz. Three dose response parameters were calculated for each compound. Growth inhibition of 50% (GI50) was calculated from [(Ti − Tz)/(C − Tz)] × 100 = 50, which is the drug concentration resulting in a 50% lower net protein increase in the treated cells (measured by SRB staining), as compared to the net protein increase seen in the control cells. The drug concentration resulting in total growth inhibition (TGI) was calculated from Ti = Tz. The LC50 (concentration of drug resulting in a 50% reduction in the measured protein at the end of the drug treatment as compared to that at the beginning), indicating a net loss of cells following treatment, was calculated from [(Ti − Tz)/Tz] × 100 = −50. Values were calculated for each of these three parameters if the level of activity is reached; however, if the effect was not reached or was exceeded, the value for that parameter was expressed as greater or less than the maximum or minimum concentration tested [58,59,60,61].

4. Conclusions

In this article we described the microwave-assisted synthesis of novel pyrazolo[3,4-g][1,8]naphthyridin-5-amine 3 by the Friedländer condensation of o-aminonitrile 1 with cyclic ketones 2 using zinc chloride as a catalyst. The presented synthetic procedure is an environmentally friendly, simple, and high yielding method for the preparation of compounds 3. The whole series of compounds 3 were tested against standardized strains of the clinically important fungi, C. albicans and C. neoformans, showing C. albicans to be more sensitive to the whole synthetic series than C. neoformans. Compounds 3a, 3d and 3g, all possessing a p-tolyl substituent, but different fused rings, were the most active structures. They showed good antifungal activity against a second panel of clinical isolates of albicans and non-albicans Candida species and appear as good models for the development of new analogues with improved activity. The antitumor evaluation data revealed that compounds 3a and 3e exhibited remarkable activity with GI50 values in the range from 10−6 M against different cancer cell lines.

Supplementary Materials

Supplementary materials can be accessed at: https://www.mdpi.com/1420-3049/20/05/8499/s1.

Acknowledgments

Authors wish to thank the COLCIENCIAS and Universidad del Valle for financial support. S.Z. and E.B. acknowledge ANPCyT and CONICET. E.B. is a doctoral fellowship of CONICET.

Author Contributions

The authors P.A., E.B., B.I., A.O., R.A., S.Z. and J.Q. designed and accomplished research. Also, they analyzed data and wrote the paper together. Finally, all authors read and approved the final manuscript.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Vanov, A.S.; Tugusheva, N.Z.; Granik, V.G. Benzo[b]naphthyridines. Russ. Chem. Rev. 2005, 74, 915–936. [Google Scholar] [CrossRef]
  2. Litvinov, V.P. Chemistry and biological activities of 1,8-naphthyridines. Russ. Chem. Rev. 2004, 73, 637–669. [Google Scholar] [CrossRef]
  3. Litvinov, V.P.; Roman, S.V.; Dyachenko, V.D. Naphthyridines. Structure, physicochemical properties and general methods of synthesis. Russ. Chem. Rev. 2000, 69, 201–220. [Google Scholar] [CrossRef]
  4. Roma, G.; Grossi, G.; Braccio, M.D.; Piras, D.; Ballabeni, V.; Tognolini, M.; Bertoni, S.; Barocelli, E. 1,8-Naphthyridines VII. New substituted 5-amino[1,2,4]triazolo[4,3-a][1,8]naphthyridine-6-carboxamides and their isosteric analogues, exhibiting notable anti-inflammatory and/or analgesic activities, but no acute gastrolesivity. Eur. J. Med. Chem. 2008, 43, 1665–1680. [Google Scholar] [CrossRef] [PubMed]
  5. Roma, G.; Braccio, M.D.; Grossi, G.; Mattioli, F.; Ghia, M. 1,8-Naphthyridines IV. 9-Substituted N,N-dialkyl-5-(alkylamino or cycloalkylamino) [1,2,4]triazolo[4,3-a][1,8]naphthyridine-6-carboxamides, new compounds with anti-aggressive and potent anti-inflammatory activities. Eur. J. Med. Chem. 2000, 35, 1021–1035. [Google Scholar] [CrossRef] [PubMed]
  6. Atanasova, M.; Ilieva, S.; Galabov, B. QSAR analysis of 1,4-dihydro-4-oxo-1-(2-thiazolyl)-1,8-naphthyridines with anticancer activity. Eur. J. Med. Chem. 2007, 42, 1184–1192. [Google Scholar] [CrossRef] [PubMed]
  7. Kuramoto, Y.; Ohshita, Y.; Yoshida, J.; Yazaki, A.; Shiro, M.; Koike, T. A novel antibacterial 8-chloroquinolone with a distorted orientation of the N-1-(5-Amino-2,4-difluorophenyl) group. J. Med. Chem. 2003, 46, 1905–1917. [Google Scholar] [CrossRef] [PubMed]
  8. Chen, K.; Kuo, S.C.; Hsieh, M.C.; Mauger, A.; Lin, C.M.; Hamel, E.; Lee, K.H. Antitumor agents. 174. 2',3',4',5,6,7-substituted 2-phenyl-1,8-naphthyridin-4-ones: Their synthesis, cytotoxicity, and inhibition of tubulin polymerization. J. Med. Chem. 1997, 40, 2266–2275. [Google Scholar] [CrossRef] [PubMed]
  9. Ferrarini, P.L.; Mori, C.; Badawneh, M.; Calderone, V.; Greco, R.; Manera, C.; Martinelli, A.; Nieri, P.; Saccomanni, G. Synthesis and β-blocking activity of (R,S)-(E)-oximeethers of 2,3-dihydro-1,8-naphthyridine and 2,3-dihydrothiopyrano[2,3-b]pyridine: Potential antihypertensive agents Part IX. Eur. J. Med. Chem. 2000, 35, 815–826. [Google Scholar] [CrossRef] [PubMed]
  10. Sherlock, M.H.; Kaminski, J.J.; Tom, W.C.; Lee, J.F.; Wong, S.C.; Kreutner, W.; Bryant, R.W.; Mcphail, A.T. Antiallergy agents. 1. Substituted 1,8-naphthyridin-2(1H)-ones as inhibitors of SRS-A release. J. Med. Chem. 1988, 31, 2108–2121. [Google Scholar] [CrossRef] [PubMed]
  11. Barlin, G.; Tan, W. 2-Chloro-7-methyl-12-phenyldibenzo[b,g][1,8]naphthyridin-11(6H)-one. Aust. J. Chem. 1984, 37, 1065–1073. [Google Scholar] [CrossRef]
  12. Sampathkumar, N.; Kumar, N.V.; Rajendran, S.P. A simple synthesis of dibenzo[b,g][1,8]naphthyridines. Synth. Commun. 2004, 34, 2019–2014. [Google Scholar] [CrossRef]
  13. Naik, T.R.R.; Naik, H.S.B.; Raghavendra, M.; Naik, S.G.K. Synthesis of thieno[2,3-b]benzo[1,8]naphthyridine-2-carboxylic acids under microwave irradiation and interaction with DNA studies. ARKIVOC 2006, 15, 84–94. [Google Scholar] [CrossRef]
  14. Ahn, S.H.; Jang, S.S.; Kim, Y.H.; Lee, K.J.; Baylis, H. Morita-Baylis-Hillman Route to 8,9,9a,10-Tetrahydrobenzo[b][1,8]naphthyridine-6(7H)-ones and 3,4,4a,5-Tetrahydrodibenzo[b,g][1,8]naphthyridine-1(2H)-ones. Bull. Korean Chem. Soc. 2011, 32, 3145–3148. [Google Scholar] [CrossRef]
  15. Manoj, M.; Prasad, K.J.R. Synthesis of linear dibenzo[1,8]naphthyridines using 2-chloro-4-methylquinolines. ARKIVOC 2011, 9, 289–307. [Google Scholar] [CrossRef]
  16. Yamuna, E.; Zeller, M.; Prasad, K.J.R. Microwave assisted synthesis of indolo[2,3-b]dibenzo[b,g][1,8]naphthyridines. Tetrahedron Lett. 2012, 53, 1514–1517. [Google Scholar] [CrossRef]
  17. Chen, K.; Kuo, S.C.; Hsieh, M.C.; Mauger, A.; Lin, C.M.; Hamel, E.; Lee, K.H. Antitumor agents. 178. Synthesis and biological evaluation of substituted 2-aryl-1,8-naphthyridin-4(1H)-ones as antitumor agents that inhibit tubulin polymerization. J. Med. Chem. 1997, 40, 3049–3056. [Google Scholar] [CrossRef] [PubMed]
  18. Mekheimer, R.A.; Hameed, A.M.A.; Sadek, K.U. 1,8-Naphthyridines II: Synthesis of novel polyfunctionally substituted 1,8-naphthyridinones and their degradation to 6 aminopyridones. ARKIVOC 2007, 20, 269–281. [Google Scholar] [CrossRef]
  19. Mohamed, E.A.; Abdel-Rahman, R.M.; El-Gendy, Z.; Ismail, M.M. Synthesis and reactions of 3-halo-1,8-naphthyridin-2,4-diones. J. Indian Chem. Soc. 1994, 71, 765–768. [Google Scholar]
  20. Ferrarini, P.L.; Mori, C.; Primofiore, G.; Gazlolari, L. One step synthesis of pyrimido[1,2-a][1,8]naphthyridinones, pyrido[1,2-a]pyrimidinones and 1,8-naphthyridinones. J. Heterocycl. Chem. 1990, 27, 881–886. [Google Scholar] [CrossRef]
  21. Strauss, C.R.; Varma, R.S. Microwaves in green and sustainable chemistry. Top. Curr. Chem. 2006, 266, 199–231. [Google Scholar]
  22. Kappe, C.O. Controlled microwave heating in modern organic synthesis. Angew. Chem. Int. Ed. 2004, 43, 6250–6284. [Google Scholar] [CrossRef]
  23. Quiroga, J.; Sanchez, N.; Acosta, P.; Insuasty, B.; Abonia, R. Microwave-assisted synthesis of fused pyrazolo[3,4-b]pyrazines by the reaction of ortho-aminonitrosopyrazoles and cyclic β-diketones. Tetrahedron Lett. 2012, 53, 3181–3187. [Google Scholar] [CrossRef]
  24. Martins, M.; Frizzo, C.; Moreira, D.; Buriol, L.; Machado, P. Solvent-free heterocyclic synthesis. Chem. Rev. 2009, 109, 4140–4182. [Google Scholar] [CrossRef] [PubMed]
  25. Quiroga, J.; Portilla, J.; Abonia, R.; Insuasty, B.; Nogueras, M.; Cobo, J. Regioselective synthesis of novel substituted pyrazolo[1,5-a]pyrimidines under solvent-free conditions. Tetrahedron Lett. 2008, 49, 6254–6256. [Google Scholar] [CrossRef]
  26. Quiroga, J.; Trilleras, J.; Insuasty, B.; Abonia, R.; Nogueras, M.; Cobo, J. Microwave-assisted synthesis of pyrazolo[3,4-d]pyrimidines from 2-amino-4,6-dichloropyrimidine-5-carbaldehyde under solvent-free conditions. Tetrahedron Lett. 2008, 49, 3257–3259. [Google Scholar] [CrossRef]
  27. Quiroga, J.; Portilla, J.; Abonia, R.; Insuasty, B.; Nogueras, M.; Cobo, J. Regioselective synthesis of novel polyfunctionally substituted pyrazolo[1,5-a]pyrimidines under solvent-free conditions. Tetrahedron Lett. 2007, 48, 6352–6355. [Google Scholar] [CrossRef]
  28. Wang, M.; Wu, A.; Pan, X.; Yang, H. Total synthesis of two naturally occurring bicyclo[3.2.1]octanoid neolignans. J. Org. Chem. 2002, 67, 5405–5409. [Google Scholar] [CrossRef] [PubMed]
  29. Ishihara, K.; Nakamura, S.; Yamamoto, H. The first enantioselective biomimetic cyclization of polyprenoids. J. Am. Chem. Soc. 1999, 121, 4906–5001. [Google Scholar] [CrossRef]
  30. Quiroga, J.; Trilleras, J.; Abonia, R.; Insuasty, B.; Nogueras, M.; Cobo, J.; de la Torre, J.M. 4-Aminopyrimidine-5-carbaldehydes as intermediates in a Friedländer type synthesis of 7-arylpyrido[2,3-d]pyrimidines. ARKIVOC 2009, 14, 9–27. [Google Scholar]
  31. Groundwater, P.W.; Munawar, M.A. Synthesis of pyrido[2,3-c]acridines. J. Chem. Soc. Perkin Trans.1 1997, 22, 3381–3386. [Google Scholar] [CrossRef]
  32. Tabarrini, O.; Cecchetti, V.; Temperini, A.; Filipponi, E.; Lamperti, M.G.; Fravolini, A. Synthesis and application of a novel, crystalline phosphoramidite monomer with thiol terminus, suitable for the synthesis of DNA conjugates. Bioorg. Med. Chem. 2001, 9, 2921–2929. [Google Scholar] [CrossRef] [PubMed]
  33. Munawar, M.A.; Groundwater, P.W. Synthesis of thieno[2,3-c]acridines. J Chem. Soc. Pak. 2004, 26, 264–269. [Google Scholar]
  34. Khalilzadeh, M.A.; Hosseini, A.; Tajbakhsh, M. Synthesis of tacrine derivatives under solventless conditions. J. Heterocycl. Chem. 2007, 44, 535–542. [Google Scholar] [CrossRef]
  35. Marco, J.L.; De los Rios, C.; Carreiras, M.C.; Banos, J.E.; Badia, A.; Vivas, N.M. Synthesis and acetylcholinesterase/butyrylcholinesterase inhibition activity of new tacrine-like analogues. Bioorg. Med. Chem. 2001, 9, 727–738. [Google Scholar] [CrossRef] [PubMed]
  36. Marco, J.L.; De los Rios, C.; Garcia, A.G.; Villarroya, M.; Carreiras, M.C.; Martins, C.; Eleuterio, A.; Morreale, A.; Orozco, M.; Luque, F. Synthesis, biological evaluation and molecular modelling of diversely functionalized heterocyclic derivatives as inhibitors of acetylcholinesterase/butyrylcholinesterase and modulators of Ca2+ channels and nicotinic receptors. Bioorg. Med. Chem. 2004, 12, 2199–2209. [Google Scholar] [CrossRef] [PubMed]
  37. Leon, R.; Contelles, J.; Garcia, A.G.; Villarroya, M. Synthesis, acetylcholinesterase inhibition and neuroprotective activity of new tacrine analogues. Bioorg. Med. Chem. 2005, 13, 1167–1176. [Google Scholar] [CrossRef] [PubMed]
  38. Barreiro, E.J.; Camara, C.A.; Verli, H.; Brazil-Mas, L.; Castro, N.G.; Cintra, W.M.; Aracava, Y.; Rodrigues, C.R.; Fraga, C.A.M. Design, synthesis, and pharmacological profile of novel fused pyrazolo[4,3-d]pyridine and pyrazolo[3,4-b][1,8]naphthyridine isosteres: A new class of potent and selective acetylcholinesterase inhibitors. J. Med. Chem. 2003, 46, 1144–1149. [Google Scholar] [CrossRef] [PubMed]
  39. Luo, F.T.; Ravi, V.K.; Xue, C.H. The novel reaction of ketones with o-oxazoline-substituted anilines. Tetrahedron 2006, 62, 9365–9371. [Google Scholar] [CrossRef]
  40. Lawen, A. Apoptosis—An introduction. Bioessays 2003, 25, 888–896. [Google Scholar] [CrossRef] [PubMed]
  41. Mathew, B.; Nath, M. Recent approaches to antifungal therapy for invasive mycoses. ChemMedChem 2009, 4, 310–323. [Google Scholar] [CrossRef] [PubMed]
  42. Chen, S.; Playford, E.; Sorrell, T. Antifungal therapy in invasive fungal infections. Curr. Opin. Pharmacol. 2010, 10, 522–530. [Google Scholar] [CrossRef] [PubMed]
  43. Brown, E.; Wright, G. New targets and screening approaches in antimicrobial drug discovery. Chem. Rev. 2005, 105, 759–774. [Google Scholar] [CrossRef] [PubMed]
  44. Vicente, M.F.; Basilio, A.; Cabello, A.; Peláez, F. Microbial natural products as a source of antifungals. Clin. Microbiol. Infect. 2003, 9, 15–32. [Google Scholar] [CrossRef] [PubMed]
  45. Park, B.; Wannemuehler, K.; Marston, B.; Govender, N.; Pappas, P. Estimation of the current global burden of cryptococcal meningitis among persons living with HIV/AIDS. Aids 2009, 23, 525–530. [Google Scholar] [CrossRef] [PubMed]
  46. Butts, A.; Krysan, D. Antifungal drug discovery: Something old and something new. PLoS Pathog. 2012, 8, e1002870. [Google Scholar] [CrossRef] [PubMed]
  47. Tang, I.; Li, J.; Zhang, L.; Ma, S.; Shi, D.; Zhang, O.; Yang, L.; Wang, X.; Liu, X.; Liu, C. The divergent transformations of aromatic o-aminonitrile with carbonyl compound. J. Heterocycl. Chem. 2012, 49, 533–542. [Google Scholar] [CrossRef]
  48. Silva, D.; Chioua, M.; Samadi, A.; Carreiras, M.; Jimeno, M.; Mendes, E.; De los Ríos, C.; Romero, A.; Villarroya, M.; López Contellesb, M.J. Synthesis and pharmacological assessment of diversely substituted pyrazolo[3,4-b]quinoline, and benzo[b]pyrazolo[4,3-g][1,8]naphthyridine derivatives. Eur. J. Med. Chem. 2011, 46, 4676–4681. [Google Scholar] [CrossRef] [PubMed]
  49. Pfaller, M.A.; Diekema, D.J. Epidemiology of invasive candidiasis: A persistent public health problem. Clin. Microbiol. Rev. 2007, 20, 133–163. [Google Scholar] [CrossRef] [PubMed]
  50. Trpkovic, M.; Pekmezovic, A.; Barac, L.; Crncevic, R.; Arsic Arsenijevic, V. In vitro antifungal activities of amphotericin B, 5-fluorocytosine, fluconazole and itraconazole against Cryptococcus neoformans isolated from cerebrospinal fluid and blood from patients in Serbia. J. Mycol. Med. 2012, 22, 243–248. [Google Scholar] [CrossRef] [PubMed]
  51. Clinical and Laboratory Standards Institute (CLSI). Reference Method for Broth Dilution Antifungal Susceptibility Testing for YeastsCLSI Document M27A3, Approved Standard—Third Edition; CLSI: Wayne, PA, USA, 2008; Volume 28, pp. 1–25. [Google Scholar]
  52. Ernst, E.J.; Roling, E.E.; Petzold, R.; Keele, D.J.; Klepser, M.E. In vitro activity of micafungin (fk-463) against Candida spp.: Microdilution, time-kill, and postantifungal-effect studies. Antimicrob. Agents Chemother. 2002, 46, 3846–3853. [Google Scholar] [CrossRef] [PubMed]
  53. Wang, X.; Dong, Y.; Wang, L.; Han, S. Acute toxicity of substituted phenols to Rana japonica tadpoles and mechanism-based quantitative structure-activity relationship (QSAR) study. Chemosphere 2001, 44, 447–455. [Google Scholar] [CrossRef] [PubMed]
  54. Leal, P.C.; Mascarello, A.; Derita, M.; Zuljan, F.; Nunes, R.J.; Zacchino, S.; Yunes, R.A. Relation between lipophilicity of alkyl gallates and antifungal activity against yeasts and filamentous fungi. Bioorg. Med. Chem. Lett. 2009, 19, 1793–1796. [Google Scholar] [CrossRef] [PubMed]
  55. C.S. Chem, Office; Version 9.0; Cambridge Soft Corporation: 100 Cambridge Park Drive, Cambridge, MA, USA, 2005.
  56. Boyd, M.R.; Paull, K.D. Some practical considerations and applications of the National Cancer Institute in vitro anticancer drug discovery screen. Drug Dev. Res. 1995, 34, 91–109. [Google Scholar] [CrossRef]
  57. Hubbard, W.C.; Alley, M.C.; Gray, G.N.; Green, K.C.; McLemore, T.L.; Boyd, M.R. Evidence for prostanoid biosynthesis as a biochemical feature of certain subclasses of non-small cell carcinomas of the lung as determined in established cell lines derived from human lung tumors. Cancer Res. 1989, 49, 826–832. [Google Scholar] [PubMed]
  58. Monks, A.; Scudiero, D.; Skehan, P.; Shoemaker, R.; Paull, K.; Vistica, D.; Hose, C.; Langley, J.; Cronise, P.; Vaigro-Wolff, A.; et al. Feasibility of a heigh-flux anticancer drug screen using a diverse panel of cultured human tumor cell lines. J. Natl. Cancer Inst. 1991, 83, 757–766. [Google Scholar] [CrossRef] [PubMed]
  59. Weinstein, J.N.; Myers, T.G.; O’Connor, P.M.; Friend, S.H.; Fornace, A.J.; Kohn, K.W.; Fojo, T.; Bates, S.E.; Rubinstein, L.V.; Anderson, N.L.; et al. An information-intensive approach to the molecular pharmacology of cancer. Science 1997, 275, 343–349. [Google Scholar] [CrossRef] [PubMed]
  60. Grever, M.R.; Sehepartz, S.A.; Chabners, B.A. The national cancer institute: Cancer drug discovery and development program. Semin. Oncol. 1992, 19, 622–638. [Google Scholar] [PubMed]
  61. Collins, J.M. Developmental Therapeutics Program NCI/NIH. Available online: http://dtp.cancer.gov/branches/btb/ivclsp.html (accessed on 12 March 2015).
  • Sample Availability: Samples of the compounds 3ai are available from the authors.

Share and Cite

MDPI and ACS Style

Acosta, P.; Butassi, E.; Insuasty, B.; Ortiz, A.; Abonia, R.; Zacchino, S.A.; Quiroga, J. Microwave-Assisted Synthesis of Novel Pyrazolo[3,4-g][1,8]naphthyridin-5-amine with Potential Antifungal and Antitumor Activity. Molecules 2015, 20, 8499-8520. https://doi.org/10.3390/molecules20058499

AMA Style

Acosta P, Butassi E, Insuasty B, Ortiz A, Abonia R, Zacchino SA, Quiroga J. Microwave-Assisted Synthesis of Novel Pyrazolo[3,4-g][1,8]naphthyridin-5-amine with Potential Antifungal and Antitumor Activity. Molecules. 2015; 20(5):8499-8520. https://doi.org/10.3390/molecules20058499

Chicago/Turabian Style

Acosta, Paola, Estefanía Butassi, Braulio Insuasty, Alejandro Ortiz, Rodrigo Abonia, Susana A. Zacchino, and Jairo Quiroga. 2015. "Microwave-Assisted Synthesis of Novel Pyrazolo[3,4-g][1,8]naphthyridin-5-amine with Potential Antifungal and Antitumor Activity" Molecules 20, no. 5: 8499-8520. https://doi.org/10.3390/molecules20058499

Article Metrics

Back to TopTop