ISSN : 0970 - 020X, ONLINE ISSN : 2231-5039
     FacebookTwitterLinkedinMendeley

Design, Synthesis, in Silico Study and Biological Evaluation of 1,8-Naphthyridine Derivatives as Potential Antibacterial Agents

Vinod Kumar Gurjar1*, Shruti Shukla2, Nirav Gondaliya2 and Nityaraj Puwar2

1School of Pharmacy, Faculty of Pharmacy, Parul University, Vadodara, Gujarat, India.

2Department of Chemistry and Industrial Chemistry, Parul Institute of Applied Sciences, Parul University, Vadodara, Gujarat, India.

Corresponding Author E-mail: vinodkumar.gurjar121130@paruluniversity.ac.in

DOI : http://dx.doi.org/10.13005/ojc/390211

Article Publishing History
Article Received on : 03 Feb 2023
Article Accepted on :
Article Published : 11 Apr 2023
Article Metrics
Article Review Details
Reviewed by: Dr. Sangeeta Jha
Second Review by: Dr. S Anand
Final Approval by: Dr. Ioana Stanciu
ABSTRACT:

The purpose of this article is to synthesize some novel 1,8-Naphthyridine-3-Carboxylic acid derivatives, analyze them, and assess their antibacterial potential. With the help of elemental analysis, IR, NMR, and mass spectrum data, the synthesized derivatives were identified. Derivatives' antibacterial activity was determined using the cup and plate diffusion method. At doses of 50 μg/ml and 100 μg/ml, the substance demonstrated substantial antibacterial potential against the tested strains. To predict the pharmacokinetic properties (ADME) of these derivatives, in silico investigations were also carried out. For the current study, the in silico Swiss ADME assisted results were shown to be suitable for the derivation and synthesis of efficient antibacterial drugs.

KEYWORDS:

Antimicrobial; ADMET; Ciprofloxacin; 1,8- Naphthyridine

Download this article as: 

Copy the following to cite this article:

Gurjar V. K, Shukla S, Gondaliya N, Puwar N. Design, Synthesis, in Silico Study and Biological Evaluation of 1,8-Naphthyridine Derivatives as Potential Antibacterial Agents. Orient J Chem 2023;39(2).


Copy the following to cite this URL:

Gurjar V. K, Shukla S, Gondaliya N, Puwar N. Design, Synthesis, in Silico Study and Biological Evaluation of 1,8-Naphthyridine Derivatives as Potential Antibacterial Agents. Orient J Chem 2023;39(2). Available from: https://bit.ly/3zKZIV9


Introduction

Microbes are the primary cause of many different diseases, including viral disease, coughing, amoebic dysentery, typhoid, malaria, and other diseases1. In the third world, communicable diseases are the main cause of illness. These illnesses are managed with a variety of medications, both synthetic and natural. Drugs or other chemicals that kill or inhibit the growth of microorganisms are known as antimicrobial agents. They consist of antibiotic, anti-quorum sensitive, antiviral, fungicidal, and anthelmintic substances2-3. In recent years, the harm caused by bacterial and fungal infections has greatly increased. Due to the widespread occurrence of antibiotic resistance, infectious diseases brought on by bacterial infections have emerged as a major public health concern. Health issues have developed as a result of antibiotic resistance, and treatment failures have led to mortality and morbidity4. Quinolones are effective antibacterial medications used to treat a variety of infectious diseases. The discovery of 1,8-naphthyridine derivative, nalidixic acid in 1962 marked the beginning of the quinolone era. Subsequently, first-generation quinolones were found, including enoxacin, lomefloxacin, and norfloxacin5. Because they contain a fluorine atom connected to the C5 on the core ring structure, the 2nd, 3rd, and 4th generation quinolones are known as fluoroquinolones6. The most popular fluoroquinolones include ciprofloxacin, levofloxacin, and moxifloxacin, all of which were created with a wide range of Gram-positive and Gram-negative bacterial activities, against enterococci, streptococci, and staphylococci7. The majority of Enterobacteriaceae bacteria as well as numerous species of bacteria that are multi-resistant to aminoglycoside and beta-lactam antibiotics are also significantly active against them8-9. Even though fluoroquinolones are very effective antibacterial agents, fluoroquinolone-resistant bacteria have unavoidably arisen as a result of their widespread use. Antibiotic resistance is a rapidly expanding public concern, and there is a high likelihood that people will get diseases that cannot be treated. The synthesis of new antibiotics has been the subject of extensive study to address this problem. The idea of adding structural alterations to current antibiotics is also being researched. These techniques have ran to the development of several new medications. The second tactic is more popular than the first because it is the most feasible one10-11. To stop this rapid rise in drug resistance, new drugs should preferably have chemical features that are distinct from those of currently available agents. The synthesis of compounds that are unique but resemble known physiologically active molecules due to the presence of important structural properties is a crucial part of the search for fresh leads in drug discovery programmes12-13.

Fluoroquinolone-type medicines have some drawbacks despite their effectiveness against infectious illnesses. In rare circumstances, drug permeability changes in fluoroquinolones have been linked to multidrug resistance14. Additionally, quinolone effects like plasmid curability, the encouragement of deteriorations and frontward mutations, and successful outcomes in genotoxicity testing 15-16 imply that quinolones may be mutagenic for bacteria. Moreover, it has been observed that a key concern for fluoroquinolones is the generation of DNA mutations and DNA diminishing in both eukaryotic and prokaryotic cells17. Introducing new fluoroquinolones without mutagenesis potential may help resolve these troubling problems. To create effective and non-mutagenic derivatives, we developed a novel series of derivatives in this class based on the information provided above.

Therefore, the necessity to develop new chemotherapeutic drugs will always be critical to prevent the establishment of resistance and, preferably, cut the length of therapy. Due to the well-established pharmacokinetics and pharmacodynamics of these medications, a change to the established medication is an additional option for innovative drug discovery. According to reports, 1,8-naphthyridine analogs exhibit a variety of pharmacological actions, including those of A2A adenosine receptor ligands18, antimycobacterial19, A1 adenosine antagonists20, anti-inflammatory21, Cannabinoid receptor ligands22, antibacterial activity23-24. Well-known antibiotics that fall within the category of antibacterial chemicals contain the 1,8-naphthyridine nucleus25, called quinolones (Figure 1). The quinolones are well-known antibacterial substances that also serve as DNA gyrase inhibitors26. The present work’s objective was to develop potential naphthyridine derivative by modification at the C3 carboxylic acid end (Figure 2, Scheme). The antibacterial efficacy of the produced compounds was evaluated in-vitro against a group of reference bacterial strains, containing Gram-positive and Gram-negative bacteria. In the current study, we demonstrated the preparation, in-silico ADME study and antibacterial activity of several 1,8-naphthyridine derivatives that may be used to create “hybrid” molecules having antibacterial capabilities.

Figure 1: Marketed antibiotics having the 1, 8-naphthyridine nucleus.

Click here to View Figure

Figure 2: Design of Molecules.

Click here to View Figure

Scheme 1: Representative Scheme for the preparation of 1,8-naphthyridine-3-cabxylic acid derivatives, Reagents and conditions:

Click here to View Scheme

Material and Methods

Reagents were often the highest-grade commercial items and were utilized without additional purification. Melting points of the derivatives were measured in open capillaries, using an X4 apparatus and were uncorrected. 1H NMR spectra of derivatives were recorded on a Bruker Avance II 400 MHz system. The coupling constants (J) for 1H were given in Hz and stated as (s) for a singlet, (d) for a doublet, (t) for a triplet, and (m) for a multiplate. All chemical shifts were expressed using the ppm scale. Mass spectra were recorded using Electrospray Ionization (ESI) technique on a Bruker’s. All the reactions were supervised via TLC using silica gel GF254 purchased from Himedia and solvent systems methanol 4%, chloroform 96%, whereas spots were visualized using an iodine chamber

Synthesis protocol

Synthesis of malonate (1a-c)

2-Aminopyridine and substituted 2-aminopyridine (6-picoline, for Compounds a, and 5-Bromo 2-aminopyridine for Compounds b) (1m mole) and diethyl ethoxy methylene malonate (1m mole) and phenoxy ether 5 ml was put under microwave oven for 5–20 min at 150–155 °C, 250 Watt power, and pressor 35 PSI. After completion, the reaction mixture was cooled at 25 oC temperature, and the solid crude producs were saperate out by filtration, washed with petroleum ether, and dried in the air.  No further purification was done to get 1,8-naphthyridine-3-carboxylate (compounds 2a-c)

Synthesis of (3a1-3a6, 3b1-3b6 and 3c1-3c6)

A mixture of the 1,8-naphthyridine derivative 2a-c (1 mmol) and cyclic amines (10 mmol) like N-ethyl piperazine, 3-Chloro aniline, pyrrolidine, pyrrolidine, morpholine, piperazine and piperidine heated in a tightly closed glass tube at 50-120 oC for 24 hrs, then reaction mixture cooled at room temperature, after cooling ether was added to the mixture to get compounds 3a1-3a6, 3b1-3b6, and 3c1-3c6 as a pure solid.

Assessment of Antimicrobial activity

Biological activity or pharmacological activity in pharmacology refers to the favorable or unfavorable effects of medication on living things. This activity is exhibited by the substance’s active ingredient or pharmacophore when medicine is a complex chemical combination, albeit it can be altered by the other ingredients. Pharmacological/biological activity is one of the many characteristics of chemical compounds that is important since it proposes uses for the compounds in medical applications. Chemical substances, however, may exhibit some unfavorable and harmful consequences, which may exclude their usage in medical procedures. In general, dose affects activity.

The test compounds (3a1-3a6, 3b1-3b6 and 3c1-3c6) were evaluated for their antibacterial activity by using cup plate method according to the method prescribed against reference bacterial strains. This traditional approach provides a measurable outcome for the number of antimicrobial drugs required to prevent the growth of particular microorganisms27-28.

This study is carried out in Petri plates, S. Aureus from the Gram-positive group of bacteria and E. Coli from the Gram-negative group of bacteria for assessment of antibacterial activity were used. To find the drug effect on these bacteria we use the cup and plate method. In which the bacteria first are spread out on a nutrient agar media present in a plate and then in that plate a well-made borer adds drug solution which is made by dissolving drug compounds at different concentrations in DMSO solvent by a micropipette. The inoculation culture plates were cultured for the bacteria at 37° for 24 hours and the fungus at 30° for 48 hours. By measure the diameter (mm) of the inhibitory zone that formed around the cylinder after incubation, the antibacterial activity was determined. Fluoroquinolone antibiotic ciprofloxacin was used as the standard  drug for antimicrobial activity of the newly synthesized derivatives.

Sample test solutions of (3a1-3a6, 3b1-3b6, and 3c1-3c6) were prepared in dimethyl sulphoxide (DMSO) at 10 times concentrations and comprehensively covering a series of dilution from 50–100 μg ml-1. This technique was practical and affordable for pipetting practice. Bacto agar in distilled water was used in preparing microbiological culture media for microbial experiments. The test analogs were mixed in to agar medium that was liquified at 45–50 °C, combined, and then put into Petri plates to set. The reference methods for determining the susceptibility of antimicrobial drugs are dilution procedures, which are used to calculate the minimum inhibitory concentrations (MICs) of antibacterial agents. The MIC was determined to be the lowest concentration that prevented the organism’s development. Before incubation, the growth rate from the control disc-which serves as the initial inoculum was compared. Spot inoculations of up to two different strains were made on each plate using the inoculating applicator. Putting the petri plate on a dark surface and determining which derivatives have the lowest concentration, preventing visible growth allowed us to calculate the minimum inhibitory concentration (MIC) endpoint after overnight incubation. All the antimicrobial activity experiments were repeated and representative data are presented29-30.

Each synthetic derivative was diluted to a stock solution of 2000 mg/ml concentration. Concentrations of the synthesized derivatives 1000, 500, and 250 mg/ml were used in the initial screening. The initial screening’s active ingredients were further diluted to provide secondary screening concentrations of 100, 50, 25, 12.5, and 6.25 μg/ml. The highest dilution in the investigation that demonstrated a least 99% inhibition zone was chosen as the MIC 31.

 In Silico Study

ADMET analysis

Through the use of the Swiss online services Swiss ADMET the generated ligands were screened physiochemically in the lab. Molecular weight, broad nitrogen and oxygen range, hydrogen bond donor/acceptor, solubility, % human absorbance, wide rotor range, and background polar area are some examples of descriptive terms (PSA). To choose drug-like molecules, Lipinski’s parameter was also taken into consideration.

The ADME investigation for this experiment was done using the SWISS ADME predictor. This is a free online tool for evaluating the properties of small molecules used in ADMET, including their solubility in water (log S), synthetic accessibility (SA), pharmacokinetics (PKs), permeability to the skin (log Kp),  percentage absorption, and drug-likeness. The following parameters were selected for the current investigation: molecular weight, hydrogen bond acceptors (HBAs), hydrogen bond donors (HBDs), and rotatable bonds (RBs)32.

Results and Discussion

Chemistry

The reaction procedure employed for synthesis of 1,8-naphthyridine derivatives are shown in Scheme. The initial steps of the synthesis of target compounds (3a1-3a6, 3b1-3b6 and 3c1-3c6) from commercially available 2-Aminopyridine, 5-Bromo-2-Aminopyridine and 6-methyl-2-Aminopyridine were carried out essentially as described in scheme20,22,33.The initial 2-aminopyridines are heated with diethylethoxymethylen malonate (EEMM) gives a malonates 1a-c. This crude ester gives naphthyridine-3-carboxylic acids 2a-c by alkaline hydrolysis. The carboxylic acids 2a-c was then treated with several primary and secondary cyclic amines like 3-Chloroaniline, morpholine, piperazine, etc. to get the target compounds (3a1-3a6, 3b1-3b6 and 3c1-3c6).

Physical and spectral data of compounds

Compound (1a)

Yield: 63%; mp 80-82 oC; IR peaks: 3271.45 (N-H), 3076.03-2980.17 (C-H Aryl), 2896.03-2780.17 (C-H methyl)  1676.86 (C=O ester), 1645.44 (C=O ring), 1582.28(C=N), 1552.39 (C=C), 847.71 (C-Br), 795.03, 737.05 (C-H).1H NMR: δ 1.30 (3H-methyl, t), 4.26 (2H-methylen, q), 7.99-8.19 (2H-Ar, 8.04 (d, J = 1.7 Hz, Aromatic-H), 8.13 (d, J = 1.7 Hz, H5), 8.53 (1H-Ar, s). m/z: 296.00, Chem. Formula: C11H9BrN2O3, mol. wt:  297.11, Elemnt. Anal:  (Calc) C, 44.47; H, 3.05; N, 9.43; O, 16.15 Elemnt. Anal:  (Found)  C, 44.44; H, 3.00; N, 9.33; O, 16.25.

Compound (1b)

Yield: 63%; mp 124-126 oC; IR peaks: 3267.72 (N-H), 3082.39-2979.91 (C-H aryl), 2896.03-2780.17 (C-H Alkyl),  1676.89 (C=O ring), 1645.12 (C=O ester), 1553.89, (C=N) 1467.92 ( C=C), 795.13, 737.79 (C-H aryl). 1H NMR: δ 1.29 (3H-Methyl, t), 2.59 (3H-Methyl, s), 4.27 (2H-methylene, q), 7.12 (1H-Ar, d, J = 7.7 Hz, Aromatic-H), 8.39-8.58 (2H-Ar, 8.44 (d, J = 7.71 Hz, Aromatic-H), 8.50 (1H-Ar, s). m/z: 232.08, Chem. Formula: C12H12N2O3, mol. wt:  232.24, Elemnt. Anal: (Calc.)  C, 62.07; H, 5.19; N, 12.06; O, 20.65. Elemnt. Anal: (Found) C, 62.16; H, 5.22; N, 12.05; O, 20.63.

Compound (1c)

Yield: 97%; mp 72-74oC; IR peaks: 3398.21 (N-H), 3071.8 (C-H, Ar ) 1740.01 (C=O ester), 1673.9 (C=O keto ring) and 1572.9, 1491.6 (C=N and C=C), 791.0 and 776.61. 1H NMR: δ 1.33 (3H-methyl, t,), 4.27 (2H-methylene, q), 7.01 (1H-Ar, d, J = 7.2, 4.7 Hz, Aromatic-H), 7.82 (1Ar, d, J = 7.2, 1.9 Hz, Aromatic-H), 8.39 (1H-Ar, d, J = 4.71, 1.9 Hz, Aromatic-H), 8.52 (1H-Ar, s). m/z: 218.07, Chem. Formula: C11H10N2O3, mol. wt:  218.21, Elemnt. Anal:  (Calc.)  C, 60.56; H, 4.61; N, 12.83; O, 22.01. Elemnt. Anal:  (Found)  C, 60.50; H, 4.52; N, 12.80; O, 22.09.

Compound (2a)

Yield: 63%; mp 86-88oC; IR peaks: 3275.40 (N-H), 3069.13-2982.27 (C-H Aryl), 2890.03-2782.17 (C-H methyl),  1672.86 (C=O acid), 1646.44 (C=O ring), 1581.21(C=N), 1550.31 (C=C), 847.71 (C-Br), 797.03, 73778.05 (C-H). 1H NMR: δ 7.97-8.19 (2H-Ar, 8.01 (d, J = 1.70 Hz, Aromatic-H), 8.14 (d, J = 1.72 Hz, Aromatic-H), 8.50 (1H-Ar, s). 8.53 (1H-Ar, s). m/z: 267.95, Chem. Formula: C9H5BrN2O3, mol. wt:  269.95, Elemnt. Anal: (Calc.)  C, 40.18; H, 1.87; N, 10.41; O, 17.84. Elemnt. Anal: (Found) C, 40.16; H, 1.80; 29.71; N, 10.42; O, 17.80.

Compound (2b)

Yield: 92%; mp 132-134 oC; IR peaks: 3269.72 (N-H), 3080.39-2972.91 (C-H aryl), 1671.89 (C=O ring), 1641.12 (C=O acid), 1551.89, (C=N) 1461.92 ( C=C), 791.13, 731.79 (C-H aryl). 1H NMR: δ 2.59 (3H-Methyl, s), 7.12 (1H-Ar, d, J = 7.7 Hz, Aromatic-H), 8.39-8.57 (2H-Ar, 8.45 (d, J = 7.7 Hz, Aromatic-H), 8.52 (s)). 8.50 (1H-Ar, s). m/z: 204.01, Chem. Formula: C10H8N2O3, mol. wt:  204.18, Elemnt. Anal: (Calc.)  C, 58.80; H, 3.94; N, 13.70; O, 23.50. Elemnt. Anal: (Found) C, 58.81; H, 3.91; N, 13.71; O, 23.48.

Compound (2c)

Yield: 73%; mp 120-122 oC; IR peaks: 3268.88(N-H), 3080-3000 (C-H Aryl), 2980.16, 2898.76(C-H alkyl), 1675.28 (C=O acid) 1645.32(C=O ring), 1486.32 (C=N) 1470.07 (C=C), 792.01 (C-H Aryl). 1H NMR: δ 7.12 (1H-Ar, d, J = 7.21, 4.69 Hz, Aromatic-H), 7.82 (1H-Ar, d, J = 7.21, 1.9 Hz, Aromatic-H), 8.38 (1H-Ar, d, J = 4.71, 1.9 Hz, Aromatic-H), 8.51 (1H-Ar, s), 10.52 (1H, s). m/z: 190.01, Chem. Formula: C9H6N2O3, mol. wt:  190.15, Elemnt. Anal:  (Calc.)  C, 56.84; H, 3.20; N, 14.70; O, 25.20, Elemnt. Anal:  (Found)  C, 56.80; H, 3.19; N, 14.70; O, 25.18.

Graph 1: IR spectra of compound 1A

Click here to View Graph

Graph 2: IR spectra of compound 1B

Click here to View Graph

Graph 3: IR spectra of compound 1C

Click here to View Graph

Graph 4: IR spectra of compound 2C

Click here to View Graph 

Graph 5: IR spectra of compound 2B

Click here to View Graph 

Graph 6: IR spectra of compound 2C

Click here to View Graph

Physical and spectral data of compounds 3a1-a6, 3b1-b6, 3c1-c6.

Compound (3a1)

Yield: 82%; mp 197-199 oC; IR peaks: 3270.56 (N-H), 3071.36-2979.48 (C-H Aryl), 2890.03-2782.17 (C-H Alkyl), 1676.63 (C=O ring), 1645.83 (C=O), 1552.69 (C=N), 1466.28 (C=C), 847.56 (C-Br), 794.80, 736.99 (C-H Aryl). 1H NMR: δ 1.31 (3H-Methyl, t), 2.36 (2H-Methylene, q), 2.61 (4H, m, J = 13.2, 6.7, 2.8 Hz, Piperazine), 3.56 (4H, m, J = 14.1, 6.7, 2.8 Hz, Piperazine), 8.07-8.19 (2H-Ar, 8.10 (d, J = 1.7 Hz, Aromatic-H), 8.13 (d, J = 1.73 Hz, Aromatic-H), 8.43 (1H-Ar, s). m/z: 364.05, Chem. Formula: C15H17BrN4O2, mol. wt:  365.23, Elemnt. Anal:  (Calc.)  C, 49.33; H, 4.69; Br, 21.88; N, 15.34; O, 8.77, Elemnt. Anal:  (Found)  C, 49.24; H, 4.61; N, 15.34; O, 8.70.

Compound (3a2)

Yield: 79%; mp 138-140 oC; IR peaks: 3271.21 (N-H), 3071.18-3050.54 (C-H Aryl), 1671.07 (C=O ring), 1645.39 (C=O), 1582.26 (C=N), 1552.75 (C=C), 847.56 (C-Br), 795.03 (C-Cl),  736.87, 703.99 (C-H Aryl). 1H NMR: δ 7.15 (1H-Ar, d, J = 8.11, 1.7 Hz, Aromatic-H), 7.34 (1H-Ar, d, J = 8.0, 0.5 Hz, Aromatic-H), 7.58 (1H-Ar, d, J = 8.2, 1.6 Hz, Aromatic-H), 7.77 (1H-Ar, d, J = 1.7, 0.5 Hz, Aromatic-H), 8.07-8.20 (2H-Ar, 8.11 (d, J = 1.7 Hz, Aromatic-H), 8.13 (d, J = 1.7 Hz, Aromatic-H), 8.49 (1H-Ar, s). m/z: 376.96, Chem. Formula: C15H9BrClN3O2, mol. wt:  378.61, Elemnt. Anal:  (Calc.)  C, 47.59; H, 2.41; Br, 21.12; Cl, 9.36; N, 11.11; O, 8.46. Elemnt. Anal:  (Found)  C, 47.52; H, 2.39; N, 11.11; O, 8.30.

Compound (3a3)

Yield: 60 %; mp 130-132 oC; IR peaks: 3271.65 (N-H), 3078.28-3050.65 (C-H Aryl), 1677.09 (C=O ring), 1645.44 (C=O), 1582.51 (C=N), 1553.09 (C=C), 847.65 (C-Br),  795.39, 736.90 (C-H Aryl). 1H NMR: δ 2.00 (4H-pyrrolidine, m, J = 13.1, 7.4, 3.9, 3.5 Hz, pyrrolidine), 3.43 (4H- pyrrolidine, m, J = 15.0, 7.3, 4.01 Hz, pyrrolidine), 8.06-8.19 (2H-Ar, 8.11 (d, J = 1.70 Hz, Aromatic-H), 8.13 (d, J = 1.71 Hz, Aromatic-H), 8.43 (1H-Ar, s). m/z: 321.01, Chem. Formula: C13H12BrN3O2, mol. wt:  322.16, Elemnt. Anal:  (Calc.)  C, 48.47; H, 3.76; Br, 24.81; N, 13.05; O, 9.92. Elemnt. Anal:  (Found)  C, 48.36; H, 3.75; N, 13.14; O, 9.91.

Compound (3a4)

Yield: 75 %; mp 145-147 oC; IR peaks: 3271.40 (N-H), 3077.03-3050.48 (C-H Aryl), 1676.98 (C=O ring), 1645.26 (C=O), 1582.48 (C=N), 1552.75 (C=C), 847.52 (C-Br),  794.90, 736.79 (C-H Aryl). 1H NMR: δ 3.53-3.69 (8H-Morpholine, 3.61 (m, J = 15.2, 6.7, 2.8 Hz, Morpholine), 3.62 (m, J = 12.0, 6.7, 2.8 Hz, Morpholine)), 8.05-8.19 (2H-Ar, d, J = 1.7 Hz, Aromatic-H), 8.13 (d, J = 1.71 Hz, Aromatic-H), 8.43 (1H-Ar, s). m/z: 337.01, Chem. Formula: C13H12BrN3O3, mol. wt:  338.16, Elemnt. Anal:  (Calc.)  C, 46.17; H, 3.58; Br, 23.63; N, 12.42; O, 14.19. Elemnt. Anal:  (Found)  C, 46.14; H, 3.54; N, 12.47; O, 14.15.

Compound (3a5)

Yield: 84 %; mp 156-158 oC; IR peaks: 3271.04 (N-H), 3077.20-3050.51 (C-H Aryl), 1677.08 (C=O ring), 1645.35 (C=O), 1582.33 (C=N), 1553.14 (C=C), 847.68 (C-Br),  794.76, 736.91 (C-H Aryl). 1H NMR: δ 1.38-1.67 (6H-piperazine, m,), 3.34 (4H, m), 8.06-8.18 (2H-Ar, d, J = 1.71 Hz, Aromatic-H), 8.13 (d, J = 1.73 Hz, Aromatic-H), 8.43 (1H-Ar, s). m/z: 335.03, Chem. Formula: C14H14BrN3O2. mol. wt:  336.19. Elemnt. Anal:   (Calc.) C, 50.03; H, 4.21; Br, 23.76; N, 12.51; O, 9.53. Elemnt. Anal:    (Found)  C, 49.97; H, 4.21; N, 12.55; O, 9.51.

Compound (3a6)

Yield: 70 %; mp 150-152 oC; IR peaks: 3268.57 (N-H), 3049.88-2980.38 (C-H Aryl), 1676.79 (C=O ring), 1643.92 (C=O), 1604.36 (C=N), 1554.49 (C=C), 847.80 (C-Br),  794.62, 737.20 (C-H Aryl). 1H NMR: δ 2.80 (4H- piperidine, m), 3.61 (4H-piperidine, m), 8.07-8.19 (2H-Ar, 8.12 (d, J = 1.71 Hz, Aromatic-H), 8.13 (d, J = 1.72 Hz, Aromatic-H), 8.43 (1H-Ar, s). m/z: 336.02, Chem. Formula: C13H13BrN4O2. mol. wt:  337.18. Elemnt. Anal:   (Calc.)  C, 46.30; H, 3.87; Br, 23.69; N, 16.59; O, 9.49. Elemnt. Anal:  (Found)  C, 46.29; H, 3.84; N, 16.58; O, 9.44.

Compound (3b1)

Yield: 63%; mp 146-148 oC; IR peaks: 3265.18 (N-H), 3080.94-2979.81 (C-H Aryl), 2890.03-2782.17 (C-H Alkyl), 1672.99 (C=O ring), 1643.50 (C=O), 1610.01 (C=N), 1556.35 (C=C),  788.70 (C-H Aryl). 1H NMR: δ 0.96 (3H-methyl, t), 2.45 (2H-methylene, q), 2.6 (4H-Piperazine, m), 3.55 (4H-Piperazine, m), 7.12 (1H-Ar, d, J = 7.7 Hz, Aromatic-H), 8.38-8.51 (2H-Ar, 8.43 (1H-Ar, s), 8.44 (d, J = 7.73 Hz, Aromatic-H). m/z: 300.16, Chem. Formula: C16H20N4O2, mol. wt:  300.36, Elemnt. Anal:  (Calc.)  C, 63.97; H, 6.72; N, 18.61; O, 10.66. Elemnt. Anal:  (Calc.)  (Found)  C, 63.90; H, 6.68; N, 18.62; O, 10.60.

Compound (3b2)

Yield: 74%; mp 156-158oC; IR peaks: 3264.92 (N-H), 3081.26-2980.13 (C-H Ar), 2929.69-2904.61(C-H methyl), 1673.10 (C=O ring), 1642.88 (C=O), 1609.89 (C=N), 1555.95 (C=C),  788.87 (C-Cl). 1H NMR: δ 2.59 (3H-methyl, s), 7.06-7.21 (2H-Ar, 7.10 (d, J = 7.71 Hz, Aromatic-H), 7.15 (d, J = 8.1 Hz, Aromatic-H), 7.34 (1H-Ar, d, J = 8.1 Hz, Aromatic-H), 7.58 (1H-Ar, d, J = 8.2 Hz, Aromatic-H), 7.77 (1H-Ar, d, J = 1.7, 0.5 Hz, Aromatic-H), 8.39-8.53 (2H-Ar, 8.44 (d, J = 7.7 Hz, Aromatic-H), 8.48 (1H-Ar, s). m/z: 313.06, Chem. Formula: C16H12ClN3O2, mol. wt:  313.74, Elemnt. Anal:  (Calc.)  C, 61.26; H, 3.88; Cl, 11.29; N, 13.40; O, 10.19. Elemnt. Anal:  (Found)  C, 61.21; H, 3.82; N, 13.31; O, 10.19.

Compound (3b3)

Yield: 63%; mp 141-143 oC; IR peaks: 3266.12 (N-H), 3081.30-2979.78 (C-H Ar), 2929.69-2894.61(C-H methyl), 1672.95 (C=O ring), 1642.95 (C=O), 1610.15 (C=N), 1558.00 (C=C), 1H NMR: δ 2.04 (4H-pyrrolidine, m), 2.58 (3H-methyl, s), 3.45 (4H-mrthylene, m), 7.12 (1H-Ar, d, J = 7.7 Hz, Aromatic-H), 8.37-8.51 (2H-Ar, d), 8.42 (1H-Ar, s), 8.45 (1H-Ar, d, J = 7.71 Hz, Aromatic-H). m/z: 257.12, Chem. Formula: C14H15N3O2, mol. wt:  257.29, Elemnt. Anal:   (Calc.)  C, 65.33; H, 5.87; N, 16.32; O, 12.45. Elemnt. Anal:  (Found)  C, 65.27; H, 5.81; N, 16.26; O, 12.37.

Compound (3b4)

Yield: 72%; mp 130-132 oC; IR peaks: 3258.67 (N-H), 3081.06-2979.49 (C-H Aryl), 2904.07-2894.61(C-H Alkyl), 1673.23 (C=O ring), 1642.61 (C=O), 1609.95 (C=N), 1555.91 (C=C). 1H NMR: δ 2.59 (3H-mrthyl, s), 3.52-3.69 (4H-Morpholine, m), 2.62 (4H-Morpholine, m), 7.12 (1H-Ar, d, J = 7.7 Hz, Aromatic-H), 8.38-8.51 (2H-Ar, 8.43 (s), 8.45 (1H-Ar, s). m/z: 273.11, Chem. Formula: C14H15N3O3, MWt: 273.29, Elemnt. Anal:   (Calc.)  C, 61.53; H, 5.54; N, 15.39; O, 17.56. Elemnt. Anal:  (Found)  C, 61.47; H, 5.49; N, 15.31; O, 17.51.

Compound (3b5)

Yield: 56%; mp 151-153 oC; IR peaks: 3258.67 (N-H), 3081.06-2979.49 (C-H Aryl), 2904.07-2894.61(C-H Alkyl), 1673.23 (C=O ring), 1642.61 (C=O), 1609.95 (C=N), 1555.91 (C=C). 1H NMR: δ 2.59 (3H-methyl, s), 2.79 (4H-piperazine, m), 3.61 (4H, m), 7.12 (1H-Ar, d, J = 7.7 Hz, Aromatic-H), 8.37-8.51 (2H-Ar, d, J = 7.73 Hz, Aromatic-H), 8.45 (1H-Ar, s). m/z: 271.12, Chem. Formula: C14H16N4O2, mol. wt:  271.30, Elemnt. Anal:  (Calc.)  C, 61.98; H, 5.57; N, 20.65; O, 11.79. Elemnt. Anal:  (Found)  C, 62.02; H, 5.60; N, 20.60; O, 11.82.

Compound (3b6)

Yield: 65%; mp 153-155oC; IR peaks: 3258.19 (N-H), 3080.87-2979.46 (C-H Aryl), 2904.23-2894.61(C-H Alkyl), 1673.19 (C=O ring), 1642.94 (C=O), 1610.06 (C=N), 1555.85 (C=C). 1H NMR: δ 1.35-1.67 (6H-Piperidine, m, J = 12.1, 6.6, 2.7 Hz, Piperidine), 1.58 (3H-Methyl, s), 3.34 (4H- Piperidine, d, J = 14.9, 6.81, 2.8 Hz, Piperidine), 8.01-8.19 (2H-Ar, d, J = 1.70 Hz, Aromatic-H), 8.12 (d, J = 1.7 Hz, Aromatic-H), 8.43 (1H-Ar, s). m/z: 271.13, Chem. Formula: C15H17N3O2, mol. wt:  271.32, Elemnt. Anal:   (Calc.)  C, 66.41; H, 6.33; N, 15.50; O, 11.78. Elemnt. Anal:  (Calc) C, 66.38; H, 6.30; N, 15.50; O, 11.81.

Compound (3c1)

Yield: 65%; mp 145-147 oC; IR peaks: 3235.46(N-H), 3120-3000 (C-H Aryl), 1664.48, 1622.70 (C=O), 1486.26 (C=N) 1486.32 (C=C), 773.15, 733.61 (C-H Aryl).   1H NMR: δ 0.96 (3H-methyl, t), 2.45 (2H-piperazine, q, J = 6.01 Hz, piperazine), 2.61 (4H-Piperazine, d), 3.56 (4H- Piperazine, d), 7.11 (1H-Ar, d, J = 7.82, 4.7 Hz, Aromatic-H), 7.81 (1H-Ar, d, J = 7.81 Hz, Aromatic-H), 8.34-8.47 (2H-Ar, d, J = 4.71, 1.9 Hz, Aromatic-H), 8.42 (1H-Ar, s). m/z: 286.14, Chem. Formula: C15H18N4O2, mol. wt:  286.34, Elemnt. Anal:  (Calc.)   C, 62.91; H, 6.34; N, 19.58; O, 11.19. Elemnt. Anal:  (Found)  C, 62.87; H, 6.31; N, 19.54; O, 11.15.

Compound (3c2)

Yield: 70%; mp 146-148 oC; IR peaks: 3236.11 (N-H), 3118.19-3067.31 (C-H Aryl), 1664.55, 1622.46 (C=O), 1480.00 (C=N) 1431.19 (C=C), 866.59 (C-Cl), 772.80, 733.18 (C-H Aryl). 1H NMR: δ 7.03 (1H-Ar, d, J = 7.81 Hz, Aromatic-H), 7.15 (1H-Ar, d, J = 8.1Hz, Aromatic-H), 7.34 (1H-Ar, d, J = 8.11 Hz, Aromatic-H), 7.58 (1H-Ar, d, J = 8.2 Hz, Aromatic-H), 7.72-7.88 (2H-Ar, d, J = 1.7 Hz, Aromatic-H), 7.81 (1H-Ar, d, J = 7.8 Hz, Aromatic-H), 8.34-8.53 (2H-Ar, d, J = 4.7 Hz, Aromatic-H), 8.48 (1H-Ar, s). m/z: 299.05, Chem. Formula: C15H10ClN3O2, mol. wt:  299.71, Elemnt. Anal:   (Calc.)  C, 60.11; H, 3.36; N, 14.02; O, 10.68. Elemnt. Anal:  (Found)  C, 59.99; H, 3.31; N, 13.98; O, 10.62.

Compound (3c3)

Yield: 69%; mp 149-151 oC; IR peaks: 3236.71 (N-H), 3124.96-3088.74 (C-H Aryl), 1685.84, 1624.35 (C=O), 1496.17 (C=N) 1431.19 (C=C), 788.73, 735.15 (C-H Aryl)..  

1H NMR: δ 2.00 (4H-Pyrrolidine, t), 3.43 (4H- Pyrrolidine, t), 7.01 (1H-Ar, d, J = 7.81, 4.7 Hz, Aromatic-H), 7.81 (1H-Ar, d, J = 7.82, 1.9 Hz, Aromatic-H), 8.34-8.47 (2H-Ar, d, J = 4.71, 1.81 Hz, Aromatic-H), 8.40 (1H-Ar, s). m/z: 243.10 (100.0%), Chem. Formula: C13H13N3O2, mol. wt:  243.27, Elemnt. Anal:  (Calc.)  C, 64.20; H, 5.38; N, 17.27; O, 13.15. Elemnt. Anal:  (Found)  C, 64.15; H, 5.35; N, 17.21; O, 13.10.

Compound (3c4)

Yield: 73%; mp 140-142 oC; IR peaks: 3236.20 (N-H), 3123.96-3069.11 (C-H Aryl), 1686.68, 1623.67 (C=O), 1487.77 (C=N) 1434.01 (C=C), 774.42, 734.93 (C-H Aryl). 1H NMR: δ 3.52-3.68 (4H-Morpholine, m), 2.42-2.46 (4H-Morpholine, m), 7.00 (1H-Ar, d, J = 7.83, 4.7 Hz, Aromatic-H), 7.81 (1H-Ar, d, J = 7.80 Hz, Aromatic-H), 8.34-8.45 (2H-Ar, d, J = 4.71 Hz, Aromatic-H), 8.45 (1H-Ar, s). m/z: 259.10, Chem. Formula: C13H13N3O3, mol. wt:  259.27, Elemnt. Anal:   (Calc.)  C, 59.98; H, 5.05; N, 16.21; O, 18.51. Elemnt. Anal: (Found)  C, 59.89; H, 4.99; N, 16.17; O, 18.50.

Compound (3c5)

Yield: 70%; mp 145-147 oC; IR peaks: 3256.67 (N-H), 3080.06-2977.49 (C-H Aryl), 2906.07-2895.61(C-H Alkyl), 1671.23 (C=O ring), 1640.61 (C=O), 1608.95 (C=N), 1554.91 (C=C). 1H NMR: δ 2.79 (4H-Piperazine, m), 3.62 (4H-Piperazine, m), 7.00 (1H-Ar, d, J = 7.83 Hz, Aromatic-H), 7.82 (1H-Ar, d, J = 7.87 Hz, Aromatic-H), 8.34-8.47 (2H-Ar, d, J = 4.7 Hz, Aromatic-H), 8.40 (1H-Ar, s). m/z: 258.11, Chem. Formula: C13H14N4O2, mol. wt:  258.28, Elemnt. Anal:  (Calc.)  C, 59.96; H, 5.45; N, 21.70; O, 12.39. Elemnt Analysis (Found)  C, 59.91; H, 5.44; N, 20.90; O, 12.40.

Compound (3c6)

Yield: 67%; mp 145 oC; IR peaks: 3237.42(N-H), 3125.55-3069.15 (C-H Aryl), 1684.84, 1623.68(C=O), 1470.07 (C=C). 1486.47 (C=N) 1433.87 (C=C), 773.16, 733.82 (C-H Aryl). 1H NMR: δ 1.44 (2H-piperidine, m), 1.66 (4H- piperidine, m), 3.34 (4H- piperidine, d), 7.10 (1H-Ar, d, J = 7.82 Hz, Aromatic-H), 7.81 (1H-Ar, d, J = 7.81 Hz, Aromatic-H), 8.34-8.47 (2H-Ar, d, J = 4.7, Aromatic-H), 8.42 (1H-Ar, s). m/z: 257.12, Chem. Formula: C14H15N3O2, mol. wt:  258.20, Elemnt. Anal: (Calc.)  C, 65.30; H, 5.86; N, 16.31; O, 12.41. Elemnt. Anal: (Found)  C, 65.29; H, 5.81; N, 16.30; O, 12.42.

Graph 7: IR Spectra of compound 3A1

Click here to View Graph

Graph 8: IR Spectra of compound 3A2

Click here to View Graph

Graph 9: IR Spectra of compound 3A3

Click here to View Graph

Graph 10: IR Spectra of compound 3A4

Click here to View Graph

Graph 11: IR Spectra of compound 3A5

Click here to View Graph

Graph 12: IR Spectra of compound 3A6

Click here to View Graph

Graph 13: IR Spectra of compound 3B1

Click here to View Graph

Graph 14: IR Spectra of compound 3B2

Click here to View Graph 

Graph 15: IR Spectra of compound 3B3

Click here to View Figure

Graph 16: IR Spectra of compound 3B4

Click here to View Graph

Graph 17: IR Spectra of compound 3B5.

Click here to View Graph

Graph 18: IR Spectra of compound 3B6

Click here to View Graph

Graph 19: IR Spectra of compound 3C1

Click here to View Graph

Graph 20: IR Spectra of compound 3C2

Click here to View Graph

Graph 21: IR Spectra of compound 3C3

Click here to View Graph

Graph 22: IR Spectra of compound 3C4

Click here to View Graph

Graph 23: IR Spectra of compound 3C5.

Click here to View Graph

Graph 24: IR Spectra of compound 3C6

Click here to View Graph

Biological evaluation

Antimicrobial Assay

An agar diffusion well technique was used to test the antibacterial activity of each of the recently synthesized derivatives (3a1-3a6, 3b1-3b6, and 3c1-3c6)34-35. The zone of inhibition was determined by a serial dilution method36-37. The newly synthesized compounds 3a1-3a6, 3b1-3b6, and 3c1-3c6 were found to have the greatest impact and the broadest spectrum of antibacterial activity against all tested reference bacterial strains. The observed inhibition zone (IZ) is shown in Fig. 3, 4, and Table 2. Derivatives were less effective against Gram-positive bacteria, while good to very strong bactericidal action toward Gram-negative bacteria. Minimum doses that prevented these microbes from growing ranged from 50 to 100 μg/ml (Table 1).

Figure 3: ZOI for 3a1-3a6, 3b1-3b6 and 3c1-3c6 against bacterial strain S. Aureus & E-coli NCTC 5933

Click here to View Figure 

Table 1: MIC In μg / ml against bacterial strain E-coli and S. Aureus

MIC in μg/ml

Compounds

E-coli

S. Aureus

A1

>50

>70

A2

>70

>60

A3

>60

>80

A4

>100

>70

A5

>80

>50

A6

>50

100

B1

>30

>40

B2

>60

>50

B3

>40

>20

B4

>20

>30

B5

>40

>40

B6

>30

>50

C1

>70

>80

C2

>60

>70

C3

>80

>70

C4

>70

>80

C5

>80

>60

C6

>80

>60

Ciprofloxacin

>20

>30

 

Table 2: ZOI In mm against bacterial strain E-coli and S. Aureus

ZOI  in mm

Compound

E-coli

S.Aureus

A1

16

18

A2

19

21

A3

17

20

A4

20

21

A5

18

15

A6

16

17

B1

15

13

B2

14

15

B3

13

16

B4

21

19

B5

17

21

B6

22

15

C1

20

18

C2

14

17

C3

16

20

C4

19

19

C5

17

21

C6

18

20

Ciprofloxacin

21

20

 

Figure 4: Bar Graph for ZOI of Antibacterial Activity

Click here to View Figure

In Silico Study

ADMET analysis and Target prediction: 

The in-silico research findings unmistakably demonstrated that the compounds have drug-like candidate homes without straying from the aforementioned drug-likeness standards. Intriguingly, leading drug similarity recommendations and the data get from the Swiss ADME prediction value for Log P, molar potency, and overall pole position within Molecule One were in great agreement. The halogen spin-off with high lipophilicity is anxiously expecting the opening of reasonable GI absorption, despite the compounds’ good hydrophilic-lipophilic stability and equivalent anticipated bioavailability. We also computed the Topological polar surface area (TPSA), which is another crucial element associated with medication bioavailability. Environment (TPSA), is one of the other important factors affecting a drug’s bioavailability. Therefore, it is believed that compounds that are passively absorbed and have a TPSA > 100 and 40 have limited oral bioavailability. Table 3 lists the outcomes from the Swiss search engine ADME. The radar charts and the chart for hard-boiled eggs (Fig. 5, 6) provided evidence in the evaluation that derivatives were in the permitted range of the classical capsules.

Table 3: Drug likeness and ADME parameters

Compd

HBA

HBD

MR

TPSA

Log P

GI
absorption

BBB
permeant

Lipinski
 violations

Bioavailability
 Score

PAINS
alerts

Synthetic
Accessibility

A1

4

1

95.8

69.3

2.69

High

No

0

0.55

0

2.38

A2

3

2

89.29

74.85

2.1

High

Yes

0

0.55

0

2.14

A3

3

1

79.38

66.06

2.1

High

Yes

0

0.55

0

1.98

A4

4

1

80.46

75.29

2.22

High

No

0

0.55

0

2.15

A5

3

1

84.18

66.06

2.44

High

Yes

0

0.55

0

2.08

A6

4

2

86.09

78.09

1.98

High

No

0

0.55

0

2.17

B1

5

2

93.81

72.46

2.93

High

No

0

0.55

0

3.02

B2

3

3

87.29

78.01

-0.01

High

Yes

0

0.55

0

2.81

B3

4

2

77.38

69.22

2.48

High

No

0

0.55

0

2.6

B4

5

2

78.47

78.45

2.42

High

No

0

0.55

0

2.82

B5

5

3

84.1

81.25

2.03

High

No

0

0.55

0

2.81

B6

4

2

82.19

69.22

2.64

High

No

0

0.55

0

2.69

C1

5

2

88.84

72.46

2.69

High

No

0

0.55

0

2.87

C2

3

3

82.33

78.01

2.35

High

No

0

0.55

0

2.67

C3

4

2

72.42

69.22

1.81

High

No

0

0.55

0

2.43

C4

5

2

73.5

78.45

1.71

High

No

0

0.55

0

2.68

C5

5

3

79.13

81.25

1.52

High

No

0

0.55

0

2.65

C6

4

2

77.22

69.22

2.07

High

No

0

0.55

0

2.53

CFL

5

2

95.25

74.57

2.24

High

No

0

0.55

0

2.51

CFL -Ciprofloxacin, HBA- Hydrogen bond acceptors, HBA- Hydrogen bond donors, TPSA-Topological polar surface area

 

Figure 5: ADME properties of derivatives and ciprofloxacin by Graphical picture (BOILED- Egg) (Predict GI Absorption and BBB Penetration of small molecules).

Click here to View Figure

Figure 6: Bioavailability Radar graph of derivatives and reference drug (pink area reflects the allowed values of drug-likeness properties of a small molecule)

Click here to View Figure

Structure Activity Relationship (SAR)

According to this research, 1,8 naphthyridine is a potential scaffold for antibacterial activity. The pyridine ring must be fused with an aromatic ring. The isotactic replacement of nitrogen atom for carbon atoms at positions 2, 5, 6, and 8 leads to the relation of antibacterial activity. The substituted position of 5, 6, 7 & 8 of the annulated ring produces good effects. Ex. The amino group at 5 positions increases the activity. Aryl substitution at the 1-position is also consistent with antibacterial activity. 1,8- positions also lead to produce active compounds. Increase the joining of a molecule the drug molecule antibacterial biological activity is an increase

Conclusion

With differently substituted amines, several 1,8-Naphthyridine derivatives were synthesized. The yield of the synthetic chemicals was discovered to be between 50 and 85%. Based on their physical and spectral data, all the newly synthesized substances were described. Analysis was done on the representative compounds’ FT-IR, 1H NMR, and mass spectral data. Antimicrobial tests were conducted on synthetic substances. All of the compounds that were synthesized had an antibacterial activity that was between average and good. The 1,8-Naphthyridine ring has been substituted in the current investigation with a variety of amines at C3, and produced compounds have been tested for antibacterial activity. Synthesized substances showed inhibition of E. coli growth. In a similar vein, the derivative inhibited S. aureus.

Acknowledgement

The author acknowledges the Parul Institute of Pharmacy and Research, Parul University Vadodara for IR spectra and Parul Sevashram hospital, Vadodara for the bacterial strain of this research work.

Conflict of Interest

None declared.

References

  1. Black, J. G., Microbiology: Principles and Applications. Prentice-Hall, Englewood Cliffs, New Jersey, 1993.
  2. Pelczar, M. J.; Chan, E. C. S.; Krieg, N. R., Microbiology. Tata Mcgraw–Hill Publishing Company Ltd., New Delhi, 2005.
  3. Singh, S.; Wanjari, P. J.; Bhatia, S.; Sonwane, V. C.; Chakraborti, A. K.; Bharatam, P. V., Med Chem Res., 2015, 5, 28.
  4. Zha, G. F.; Leng, J.; Darshini, N.; Shubhavathi, T.; Vivek, H. K.; Asiri A. M.; Marwani, H. M.; Rakesh, K.P.; Mallesha, N.; Qin, H. L., Bioorg Med Chem Lett., 2017, 27, 3148.
    CrossRef
  5. Ball, P., Quinolone generations: natural history or natural selection? J Antimicrob Chemother, 2000, 46,17.
    CrossRef
  6. Oliphant, C. M., & Green, G.M., Am Fam Physician, 2002, 455-465.
  7. Blondeau, J. M., Clin Ther, 1999, 21, 3.
  8. Vyas, S.; Parmar, K.; Patel, B.; Chaudhari, M.; Umrigar, N.; Goswami, K., Orient J Chem, 2022, 38(4), 1064-1068.
    CrossRef
  9. Jones, R. N.; Low, D. E.; Pfaller, M. A., Diagn Microbial Infect Dis, 1999, 33, 101-112.
    CrossRef
  10. Bisacchi, G. S., J Med Chem, 2015, 58, 4874-4882.
    CrossRef
  11. Chugunova, E.; Akylbekov, N.; Bulatova, A.; Gavrilov, N.; Voloshina, A.; Kulik, N.; Zobov, V.; Dobrynin, A.; Syakaev, V.; Burilov, A., Eur J Med Chem, 2016, 116, 165-172.
    CrossRef
  12. Thompson, L. A.; Ellman, J. A., Chem Rev, 1996, 96, 555-600.
    CrossRef
  13. Andriote, V. T., J Antimicrob Chemother, 1999, 44, 151-162.
    CrossRef
  14. Mamber, S. W.; Kolek, B.; Brookshire, K. W.; Bonner, D. P.; Fung–Tomc, J., Antimicrob Agents Chemother, 1993, 37, 213.
    CrossRef
  15. Albertini, S.; Chételat, A. A.; Miller, B.; Muster, W.; Pujadas, E.; Strobel, R.; Gocke, E., Mutagenesis, 1995, 10, 343-351.
    CrossRef
  16. Yim, G.; McClure, J.; Surette, M. G.; Davies, J. E., J Antibio,2011,  64, 73-78.
    CrossRef
  17. Fort, F. L., Drug Saf, 1992, 7, 214-222.
    CrossRef
  18. Manera, C.; Betti, L.; Cavallini, T.; Giannaccini, G.; Martinelli, A.; Ortore, G.; Saccomanni, G.; Trincavelli, L.; Tuccinardia, T.; Ferrarini, P. L., Bioorg Med Chem Lett, 2005, 15, 4604-4610.
    CrossRef
  19. Ferrarini, P. L.; Manera, C.; Mori, C.; Badawneh, M.; Saccomanni, G., Il Farmaco, 1998, 53, 741-746.
    CrossRef
  20. Ferrarini, P. L.; Betti, L.; Cavallini, T.; Giannaccini, G.; Lucacchini, A.; Manera, C.; Martinelli, A., J Med Chem, 2004, 47, 3019-3031.
    CrossRef
  21. Dianzani, C.; Collino, M.; Gallicchio, M.; Braccio, M. D.; Roma, G.; Fantozzi, R., J Inflam (Lond), 2006, 3,4.
    CrossRef
  22. Ferrarini, P. L.; Calderone, V.; Cavallini, T.; Manera, C.; Saccomanni, G.; Pani, L.; Ruiu, S.; Gessa, G. L., Bioorg Med Chem, 2004, 15, 1921-1933.
    CrossRef
  23. Lv, K.; Liu, M. L.; Feng, L. S.; Sun, L. Y.; Sun, Y. X.; Wei, Z. Q.; Guo, H. Q., Eur J Med Chem, 2012, 47, 619-625.
    CrossRef
  24. Palabindela, R.; Myadaravenia, P.; Banothu, D.; Korra, R.; Mekala, H.; Kasula. M.,. Orient J Chem, 2022, 38(1), 137-143.
    CrossRef
  25. Saleh, M. Y., & Ayoub, A. I., Eur J Chem, 2014, 5, 475.
    CrossRef
  26. Smith, J., J Antimicrob Chemother, 1986 (Supl D), 18, 21-29.
    CrossRef
  27. Isenberg, H. D., Clinical Microbiology Procedures Handbook, vol. 1, American society for microbiology/Washington, D.C. 1992.
  28. Schwalve, R.; Moore, L.S.; Goodwin, A. C., Antimicrobial Susceptibility Testing Protocols, Crc. Press, Taylor, and Francis. 2007.
    CrossRef
  29. Grover, G., & Kini, S. G., Eur J Med Chem, 2006, 41, 256-262.
    CrossRef
  30. Wiegand, I.; Hilpert, K.; Hancock, R. E. W., Nat Protoc, 2008, 3, 163-175.
    CrossRef
  31. O’Donnell, F.; Smyth, T. J.; Ramachandran, V. T.; Smyth, W. F., Int J Antimicrob Agents, 2010, 35, 30-38.
    CrossRef
  32. Daina, A.; Michielin, O.; Zoete, V., Sci Rep, 2017, 7, 42717.
    CrossRef
  33. Li, W.; Hong, G.; Mao, L.; Xu, Z.; Wang, J.; Wang, W.; Liu, T., Med Chem Res, 2022, 31, 705-719.
  34. Manera, C.; Benetti, V.; Castelli, M. P.; Cavallini, T.; Lazzarotti, S.; Pibiri, F.; Saccomanni, G.; Tuccinardi, T.; Vannacci, A.; Martinelli, A.; Ferrarini, P. L., J Med Chem, 2006, 49, 59475957.
  35. Patel, D. M,; Patel, N. K.; Narolkar, S.; Mishra, A., Orient J Chem, 2022, 38(2), 432-438.
    CrossRef
  36. Sarıkavaklı, N.; Koç, E.; Aljohani, M. M. H.; Mustafa, S. K., Orient J Chem, 2020, 36(6), 1194-1204
    CrossRef
  37. Cappuccino, J. G., & Sherman, N., Microbiology: a laboratory manual Benjamin-Cummings Pub Co, California. 1999.

Creative Commons License
This work is licensed under a Creative Commons Attribution 4.0 International License.

About The Author