Next Article in Journal
Accuracy of 3D Tooth Movements in the Fabrication of Manual Setup Models for Aligner Therapy
Previous Article in Journal
Sugar Palm Fibre-Reinforced Polymer Composites: Influence of Chemical Treatments on Its Mechanical Properties
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Relationship between Structure and Antibacterial Activity of α-Aminophosphonate Derivatives Obtained via Lipase-Catalyzed Kabachnik−Fields Reaction

by
Dominik Koszelewski
1,*,
Paweł Kowalczyk
2,*,
Paweł Śmigielski
1,
Jan Samsonowicz-Górski
1,
Karol Kramkowski
3,
Aleksandra Wypych
4,
Mateusz Szymczak
5 and
Ryszard Ostaszewski
2
1
Institute of Organic Chemistry PAS, Kasprzaka 44/52, 01-224 Warsaw, Poland
2
Department of Animal Nutrition, The Kielanowski Institute of Animal Physiology and Nutrition, Polish Academy of Sciences, Instytucka 3, 05-110 Jabłonna, Poland
3
Department of Physical Chemistry, Medical University of Bialystok, Kilińskiego 1 Str., 15-089 Białystok, Poland
4
Centre for Modern Interdisciplinary Technologies Nicolaus Copernicus University in Torun ul. Wileńska 4, 87-100 Toruń, Poland
5
Department of Molecular Virology, Institute of Microbiology, Faculty of Biology, University of 7 Warsaw, Miecznikowa 1, 02-096 Warsaw, Poland
*
Authors to whom correspondence should be addressed.
Materials 2022, 15(11), 3846; https://doi.org/10.3390/ma15113846
Submission received: 24 March 2022 / Revised: 29 April 2022 / Accepted: 25 May 2022 / Published: 27 May 2022

Abstract

:
We reported a new method dealing with the synthesis of novel pharmacologically relevant α-aminophosphonate derivatives via a lipase-catalyzed Kabachnik−Fields reaction with yields of up to 93%. The advantages of this protocol are excellent yields, mild reaction conditions, low costs, and sustainability. The developed protocol is applicable to a range of H-phosphites and organic amines, providing a wide substrate scope. A new class of α-aminophosphonate analogues possessing P-chiral centers was also synthesized. The synthesized compounds were characterized on the basis of their antimicrobial activities against E. coli. The impact of the various alkoxy groups on antimicrobial activity was demonstrated. The crucial role of the substituents, located at the aromatic rings in the phenylethyloxy and benzyloxy groups, on the inhibitory action against selected pathogenic E. coli strains was revealed. The observed results are especially important because of increasing resistance of bacteria to various drugs and antibiotics.

1. Introduction

α-Aminophosphonates appear to be a very important class of organic compounds because of their potential biological activities [1,2,3,4]. The distinctive character of bioactive organophosphorus compounds has established their wide applicability in agricultural and medicinal chemistry [5,6,7,8,9,10,11,12]. α-Aminophosphonates (Figure 1) play a crucial role as a platform to design new drugs [13,14,15,16,17,18,19,20]. Among other advantages, there are several reports regarding their antimicrobial activity; alafosfalin, for example, a simple dialkyl α-aminophosphonate, exhibits activity against pathogenic E. coli, S. aureus, Bacillus, and K. pneumonia strains (Figure 1) [21,22,23,24,25,26,27,28,29,30]. However, the application of alafosfalin in medicine is limited due to its instability. It is shown that the correct design of the alkoxyl groups in the H-phosphite used for these compounds may significantly increase their antimicrobial properties (Figure 1) [1,2,3,31,32,33,34,35,36].
The aim of the work is to develop a metal-free protocol for the preparation of α-aminophosphonate derivatives with P-chiral centers on bacterial strains K12 and R2−R4.

2. Materials and Methods

2.1. Microorganisms and Media

All microorganisms and media were accurately described in detail in the previous work [37,38,39,40,41,42,43,44,45,46,47,48,49,50,51,52,53,54,55,56,57,58,59,60,61,62,63,64,65,66,67,68,69,70,71,72,73,74] and analyzed by a Tukey test.

2.2. General Methods of Synthesis α-Aminophosphonate Derivatives

All the chemicals were described in detail in the previous work [74]. All specific strains, such as Pseudomonas cepacia (PcL) and wheat germ, were provided by Sigma-Aldrich (Merck). The bovine acetone powder was prepared in our laboratory according to the literature procedure [38]. Symmetrical and unsymmetrical H-phosphites were obtained via alcoholysis of the dimethyl phosphite with the appropriate alcohol according to the literature procedure [40,41,42,43,44,45,46] (see Supplementary Materials).

3. Results

3.1. Chemistry

Organophosphorus compounds show a variety of relevant biological activities [47,48,49]. The Kabachnik–Fields reaction is the most efficient method for the formation of carbon−phosphorus bonds using an aldehyde, amine, and H-phosphite. A number of other synthetic approaches have also been reported for the preparation of α-aminophosphonates [7,50,51,52,53,54,55,56,57,58,59,60,61,62,63,64,65,66,67,68,69,70,71,72,73,74,75,76,77,78,79,80,81,82,83,84,85,86,87,88,89,90,91,92,93,94,95]. These methods are generally conducted in the presence of various organic and inorganic bases [56,57,58] as well as Lewis and Bronsted acids, such as zirconium tetrachloride (ZrCl4), aluminum chloride (AlCl3), tantalum pentachloride (TaCl5), or lanthanide triflates [59,60,61,62,63]. Therefore, it is necessary to further develop an efficient one-pot, multicomponent synthesis of α-aminophosphonates that is devoid of these problems and fulfils requirements of the pharmaceutical industry. Enzymes, which are natural catalysts with high catalytic activity, seem to be the best alternative leading to the development of new synthesis methods that meet the requirements related to safety and environmental protection. In addition, enzymes enable the synthesis of compounds without metal contamination, which is especially appreciated by the pharmaceutical industries. Among other uses, hydrolases are most often used as biocatalysts in organic synthesis. Our work is more focused on discovering new unnatural catalytic activities of hydrolases. This phenomenon was defined as enzymatic promiscuity. Recently, a number of unnatural reactions catalyzed by hydrolases have been reported, such as the aza-Henry reaction [64], Michael additions [65,66], 1,2-addition of thiols to imines [67], and Morita–Baylis–Hillman reaction [68,69,70]. Although some chemical strategies work towards the synthesis of α-aminophosphonates, the biocatalytic preparation of target α-aminophosphonates remains unexploited. It was shown that some selected α-aminophosphonates could be obtained from aniline derivatives by the Kabachnik–Fields reaction using Candida antarctica lipase B (CAL-B) as a catalyst [71,72,73].
As a continuation of our research on seeking new catalytic activities of hydrolases [74,75,76,77,78,79,80,94,95], we focused our efforts on elaborating a sustainable metal-free method towards desired α-aminophosphonates 116 (Figure 2).
Regarding the promiscuous activity of lipases, [71] the model Kabachnik–Fields reaction of benzyl amine (1 mmol), benzaldehyde (1 mmol), and dimethyl phosphite (1 mmol) was conducted in neat at 25 °C (Scheme 1 and Table 1, entry 1).
As shown in Table 1, lipase from a porcine pancreas (PpL) was found as the best catalyst among the tested lipases for this addition reaction (Table 1, entry 2). The α-aminophosphonate 1 obtained agood yield (73%) after 24 h in neat at 25 °C. The yield did not increase substantially after 24 h. In the absence of enzyme only traces of the target product 1 was formed (Table 1, entry 1). In addition, four different nonenzymatic catalysts were reported in the literature as sustainable promoters of the Kabachnik–Fields reaction [81,82,83,94,95]; copper(I) iodide, copper(I) oxide, copper(II) acetate, and phenylboronic acid were tested under similar reaction conditions, leading to the target product 1 with up to a 39% yield (Table 1, entries 19–22). It is well recognized that the type of solvent used has a great impact on enzyme stability and activity [84]. Product 1 was provided with the highest yield of 88% in TBME (Table 1, entry 7); therefore, this solvent was applied in the following optimization. Furthermore, the model reaction was carried out at elevated temperatures; however, the yield of product 1 was reduced at temperatures above 30 °C (Table 1, entries 8 and 9). Next, we studied if the amount of enzyme used had any impact on the reaction yield, and we found out that the yield of target compound 1 increased slightly by raising the amount of PpL from 50 mg to 80 mg. Thus, the 80 mg of PpL was the optimal amount for the further investigations [71,94,95].
Finally, we used the elaborated enzymatic protocol with various aromatic and aliphatic amines, aldehydes, and symmetrical as well as unsymmetrical H-phosphites [41] (Figure 2). The enzymatic Kabachnik–Fields reaction with aliphatic aldehydes and amines as well as 2-phenylethylamine provided products 4, 5, and 911 with lower yield ranges from 51% to 71% (Figure 2). A similar reduction in the reaction yield was observed for sterically bulky electron-rich aldehyde and amine with methoxy groups located at the phenyl ring, which resulted in product 8 with a 69% yield. Finally, the application of unsymmetrical H-phosphonates provided P-chiral products 1416 as a mixture of diasteroisomers (1:1) with yields up to 76%. The structures of all obtained compounds 116 are presented in the experimental section (Supplementary Materials Figures S4–S65).
Additional experiments were performed to gather insights on the reaction pathway. Under developed conditions, N-(4-methoxylbenzylidene)benzylamine was used together with dimethyl H-phosphite in the presence of PpL as a catalyst, which resulted in an excellent yield of 95% of the target α-aminophosphonate 1. This observation constitutes the initial formation of an imine in the presence of lipase (Scheme 2).

3.2. Cytotoxic Studies of the Library of α-Hydroxy Phosphonate Derivatives

It is worth noting that the introduction of a fluorine atom into the structure of all 16 tested compounds did not have a significant effect on the activity of 2 and 12, which is often observed for various types of compounds exhibiting antibacterial activity [12] (Figure 3, Figure 4, Figure 5, Figure 6 and Figure 7).
The analyzed bacterial strains used in the experiments were used in 48-well plates. (Figure 3, Figure 4 and Figure 5 and Table 2).

3.3. Analysis of R2–R4 E. coli Strains Modified with α-Aminophosphonate Derivatives

The obtained MIC values as well as our previous studies with various types of the analyzed compounds [85,86,87,88,89,90,91,92,93,94,95] indicate that α-aminophosphonate derivatives also show a strong toxic effect on the analyzed bacterial model strains. The three analyzed compounds were selected for further analysis by modifying their DNA. Modified bacterial DNA was digested with Fpg as described earlier [85,86,87,88,89,90,91,92,93]. All selected analyzed α-aminophosphonate derivatives (Figure 6), including different types of alkoxy groups, substituents located at the phenyl ring, and the length of the alkyl chain, can strongly change the topology of bacterial DNA. After digestion with Fpg, approximately 3.5% of oxidative damage was identified, which very strongly indicates oxidative damage in bacterial DNA, similar to the previous observations [85,86,87,88,89,90,91,92,93]. The different types of alkoxy groups, substituents located at the phenyl ring, and the length of the alkyl affected this outcome (Figure 6).
Figure 6. Percentage of plasmid DNA recognized by the Fpg enzymes (y-axis) with model bacterial, K12, and R2–R4 strains (x-axis). All analyzed compounds numbered were statistically significant at <0.05 (see Table 2).
Figure 6. Percentage of plasmid DNA recognized by the Fpg enzymes (y-axis) with model bacterial, K12, and R2–R4 strains (x-axis). All analyzed compounds numbered were statistically significant at <0.05 (see Table 2).
Materials 15 03846 g006

3.4. R2-R4 E. coli Strains with Tested α-Aminophosphonate Derivatives

The performed studies prove that the analyzed and newly synthesized compounds can potentially be used as “substitutes” for the currently used antibiotics in hospital and clinical infections, (Figure 7 and Figure 8 and Supplementary Materials Figure S3).
Figure 7. Examples of MIC with model bacterial strains K12, R2, R3, and R4 for studying the antibiotics ciprofloxacin (cipro), bleomycin (bleo), and cloxacillin (clox). The x-axis features antibiotics used sequentially. The y-axis features the MIC value in µg/mL−1.
Figure 7. Examples of MIC with model bacterial strains K12, R2, R3, and R4 for studying the antibiotics ciprofloxacin (cipro), bleomycin (bleo), and cloxacillin (clox). The x-axis features antibiotics used sequentially. The y-axis features the MIC value in µg/mL−1.
Materials 15 03846 g007
Large modifications of plasmid DNA were observed for the three analyzed compounds numbered 13, 15, and 16, showing high superselectivity.

4. Conclusions

Our developed protocol provides an efficient mild and metal-free synthesis of the target products with a high yield (51–93%). Among the studied derivatives, the compounds possessing alkoxy groups with halogen atoms or nitro groups in phosphate moieties 13, 15, and 16 turned out to be the most active compared to derivatives with the dimethyl groups (Figure 2). Finally, the reported α-aminophosphonate derivatives are more cytotoxic in the model bacterial cells than the following commonly used antibiotics: ciprofloxacin, bleomycin, and cloxacillin.

Supplementary Materials

The supporting information can be downloaded at the following: https://www.mdpi.com/article/10.3390/ma15113846/s1. Figure S1: examples of MIC and MBC on microplates with different concentrations of studied compounds (mg L−1). Figure S2: an example of an agarose gel electrophoresis separation of isolated plasmids DNA from R4 strains modified with selected compounds. Figure S3: an example of an agarose gel electrophoresis separation of isolated plasmids DNA from R4 strains modified with the following antibiotics: cloxacilline, ciprofloxaclinie, and bleomycine digested (or not) with repair Fpg enzymes. NMR spectra of α-aminophosphonate derivatives 116. Figure S4: 1HNMR (400 MHz, CDCl3) spectra of compound 1. Figure S5: 13CNMR (100 MHz, CDCl3) spectra of compound 1. Figure S6: 31PNMR (162 MHz, CDCl3) spectra of compound 1. Figure S7: 1HNMR (400 MHz, CDCl3) spectra of compound 2. Figure S8: 13CNMR (100 MHz, CDCl3) spectra of compound 2. Figure S9: 31PNMR (162 MHz, CDCl3) spectra of compound 2. Figure S10: 1HNMR (400 MHz, CDCl3) spectra of compound 3. Figure S11: 13CNMR (100 MHz, CDCl3) spectra of compound 3. Figure S12: 31PNMR (162 MHz, CDCl3) spectra of compound 3. Figure S13: 1HNMR (400 MHz, CDCl3) spectra of compound 4. Figure S14: 13CNMR (100 MHz, CDCl3) spectra of compound 4. Figure S15: 31PNMR (162 MHz, CDCl3) spectra of compound 4. Figure S16: 1HNMR (400 MHz, CDCl3) spectra of compound 5. Figure S17: 13CNMR (100 MHz, CDCl3) spectra of compound 5. Figure S18: 31PNMR (162 MHz, CDCl3) spectra of compound 5. Figure S19: 1HNMR (400 MHz, CDCl3) spectra of compound 6. Figure S20: 13CNMR (100 MHz, CDCl3) spectra of compound 6. Figure S21: 31PNMR (162 MHz, CDCl3) spectra of compound 6. Figure S22: 1HNMR (400 MHz, CDCl3) spectra of compound 7. Figure S23: 13CNMR (100 MHz, CDCl3) spectra of compound 7. Figure S24: 31PNMR (162 MHz, CDCl3) spectra of compound 7. Figure S25: 1HNMR (400 MHz, CDCl3) spectra of compound 8. Figure S26: 13CNMR (100 MHz, CDCl3) spectra of compound 8. Figure S27: 31PNMR (162 MHz, CDCl3) spectra of compound 8. Figure S28: 1HNMR (400 MHz, CDCl3) spectra of compound 9. Figure S29: 13CNMR (100 MHz, CDCl3) spectra of compound 9. Figure S30: 31PNMR (162 MHz, CDCl3) spectra of compound 9. Figure S31: 1HNMR (400 MHz, CDCl3) spectra of compound 10. Figure S32: 13CNMR (100 MHz, CDCl3) spectra of compound 10. Figure S33: 31PNMR (162 MHz, CDCl3) spectra of compound 10. Figure S34: 1HNMR (400 MHz, CDCl3) spectra of compound 11. Figure S35: 13CNMR (100 MHz, CDCl3) spectra of compound 11. Figure S36: 31PNMR (162 MHz, CDCl3) spectra of compound 11. Figure S37: 1HNMR (400 MHz, CDCl3) spectra of compound 12. Figure S38: 13CNMR (100 MHz, CDCl3) spectra of compound 12. Figure S39: 31PNMR (162 MHz, CDCl3) spectra of compound 12. Figure S40: 1HNMR (400 MHz, CDCl3) spectra of compound 13. Figure S41: 13CNMR (100 MHz, CDCl3) spectra of compound 13. Figure S42: 31PNMR (162 MHz, CDCl3) spectra of compound 13. Figure S43: 1HNMR (400 MHz, CDCl3) spectra of compound 14. Figure S44: 13CNMR (100 MHz, CDCl3) spectra of compound 14. Figure S45: 31PNMR (162 MHz, CDCl3) spectra of compound 14. Figure S46: 1HNMR (400 MHz, CDCl3) spectra of compound 15. Figure S47: 13CNMR (100 MHz, CDCl3) spectra of compound 15. Figure S48: 31PNMR (162 MHz, CDCl3) spectra of compound 15. Figure S49: 1HNMR (400 MHz, CDCl3) spectra of compound 16. Figure S50: 13CNMR (100 MHz, CDCl3) spectra of compound 16. Figure S51: 31PNMR (162 MHz, CDCl3) spectra of compound 16. Figure S52: 1HNMR (400 MHz, CDCl3) spectra of bis(4-fluorophenylethyl)phosphite. Figure S53: 13CNMR (100 MHz, CDCl3) spectra of bis(4-fluorophenylethyl)phosphite. Figure S54: 31PNMR (162 MHz, CDCl3) spectra of bis(4-fluorophenylethyl)phosphite. Figure S55: 1HNMR (400 MHz, CDCl3) spectra of methyl (phenylethyl) phosphite. Figure S56: 13CNMR (100 MHz, CDCl3) spectra of methyl (phenylethyl) phosphite. Figure S57: 31PNMR (162 MHz, CDCl3) spectra of methyl (phenylethyl) phosphite. Figure S58: 1HNMR (400 MHz, CDCl3) spectra of methyl (4-chlorophenylethyl) phosphite. Figure S59: 13CNMR (100 MHz, CDCl3) spectra of methyl (4-chlorophenylethyl) phosphite. Figure S60: 31PNMR (162 MHz, CDCl3) spectra of methyl (4-chlorophenylethyl) phosphite. Figure S61: 1HNMR (400 MHz, CDCl3) spectra of methyl (4-nitrobenzyl) phosphite. Figure S62: 13CNMR (100 MHz, CDCl3) spectra of methyl (4-nitrobenzyl) phosphite. Figure S63: 31PNMR (162 MHz, CDCl3) spectra of methyl (4-nitrobenzyl) phosphite. Figure S64: 1HNMR (400 MHz, CDCl3) spectra of N-(4-methoxylbenzylidene)benzylamine. Figure S65: 13CNMR (100 MHz, CDCl3) spectra of N-(4-methoxylbenzylidene)benzylamine.

Author Contributions

Conceptualization, P.K. and D.K.; methodology, P.K., D.K., P.Ś., J.S.-G. and A.W.; software P.Ś., J.S.-G., K.K., investigation, P.K., A.W. and M.S.; validation, P.Ś., J.S.-G., K.K., formal analysis P.K., D.K., R.O., A.W., P.Ś., J.S.-G., K.K. resources, P.K., D.K., K.K.; data curation, K.K., M.S. and A.W.; writing—original draft preparation, P.K., R.O. and D.K.; writing—review and editing, D.K., R.O. and P.K.; visualization P.K., D.K., R.O.; project administration P.K., D.K., R.O.; supervision, P.K., D.K.; funding acquisition K.K. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by a grant from the Medical University of Białystok SUB/2/DN/22/001/2201 and by National Science Center, Poland project OPUS No. 2019/33/B/ST4/01118.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

On request of those interested.

Acknowledgments

The authors thank Jolanta Łukasiewicz from the Ludwik Hirszfeld Institute of Immunology and Experimental Therapy (Polish Academy of Sciences) for providing the strains of E. coli.

Conflicts of Interest

The authors declare no conflict of interest.

Abbreviations

MICminimum inhibitory concentration
MBCminimum bactericidal concentration
Ocopen circle
Ccccovalently closed circle
BERbase excision repair
FpgDNA-formamidopyrimidine glycosylase

References

  1. Albrecht, L.; Albrecht, A.; Krawczyk, H.; Jorgensen, K.A. Organocatalytic Asymmetric Synthesis of Organophosphorus Compounds. Chem. Eur. J. 2010, 16, 28–48. [Google Scholar] [CrossRef] [PubMed]
  2. Zhao, D.; Wang, R. Recent developments in metal catalyzed asymmetric addition of phosphorus nucleophiles. Chem. Soc. Rev. 2012, 41, 2095–2108. [Google Scholar] [CrossRef] [PubMed]
  3. Hiratake, J.; Oda, J. Aminophosphonic and Aminoboronic Acids as Key Elements of a Transition State Analogue Inhibitor of Enzymes. Biosci. Biotechnol. Biochem. 1997, 61, 211–218. [Google Scholar] [CrossRef] [Green Version]
  4. Kafarski, P.; Lejczak, B. Biological activity of aminophosphonic acids. Phosphorus Sulfur Silicon Relat. Elem. 1991, 63, 193–215. [Google Scholar] [CrossRef]
  5. Nassan, M.A.; Aldhahrani, A.; Amer, H.H.; Elhenawy, A.; Swelum, A.A.; Ali, O.M.; Zaki, Y.H. Investigation of the Anticancer Effect of α-Aminophosphonates and Arylidine Derivatives of 3-Acetyl-1-aminoquinolin-2(1H)-one on the DMBA Model of Breast Cancer in Albino Rats with In Silico Prediction of Their Thymidylate Synthase Inhibitory Effect. Molecules 2022, 27, 756. [Google Scholar] [CrossRef]
  6. Rezaei, Z.; Khabnadideh, S.; Zomorodian, K.; Pakshir, K.; Nadali, S.; Mohtashami, N.; Mirzaei, E.F. Design, Synthesis, and Antifungal Activity of New α-Aminophosphonates, Design, Synthesis, and Antifungal Activity of New α-Aminophosphonates. Int. J. Med. Chem. 2011, 2011, 678101. [Google Scholar] [CrossRef] [Green Version]
  7. Varga, P.R.; Keglevich, G. Synthesis of α-Aminophosphonates and Related Derivatives; The Last Decade of the Kabachnik–Fields Reaction. Molecules 2021, 26, 2511. [Google Scholar] [CrossRef]
  8. Iwanejko, J.; Samadaei, M.; Pinter, M.; Senfter, D.; Madlener, S.; Kochel, A.; Rohr-Udilova, N.; Wojaczyńska, E. Cytotoxic Activity of Piperazin-2-One-Based Structures: Cyclic Imines, Lactams, Aminophosphonates, and Their Derivatives. Materials 2021, 14, 2138. [Google Scholar] [CrossRef]
  9. Packialakshmi, P.; Gobinath, P.; Ali, D.; Alarifi, S.; Alsaiari, N.S.; Idhayadhulla, A.; Surendrakumar, R. Synthesis and Characterization of Aminophosphonate Containing Chitosan Polymer Derivatives: Investigations of Cytotoxic Activity and in Silico Study of SARS-CoV-19. Polymers 2021, 13, 1046. [Google Scholar] [CrossRef]
  10. López-Francés, A.; del Corte, X.; de Marigorta, E.M.; Palacios, F.; Vicario, J. Ugi Reaction on α-Phosphorated Ketimines for the Synthesis of Tetrasubstituted α-Aminophosphonates and Their Applications as Antiproliferative Agents. Molecules 2021, 26, 1654. [Google Scholar] [CrossRef]
  11. Xu, Y.; Yan, K.; Song, B.; Xu, G.; Yang, S.; Xue, W.; Hu, D.; Lu, P.; Ouyang, G.; Jin, L.; et al. Synthesis and Antiviral Bioactivities of α-Aminophosphonates Containing Alkoxyethyl Moieties. Molecules 2006, 11, 666–676. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  12. Song, B.-A.; Wu, Y.-L.; Yang, S.; Hu, D.-Y.; He, X.-Q.; Jin, L.-H. Synthesis and Bioactivity of a-Aminophosphonates Containing Fluorine. Molecules 2003, 8, 186–192. [Google Scholar] [CrossRef]
  13. Onita, N.; Şisu, I.; Penescu, M.; Purcarea, V.L.; Kurunczi, L. Synthesis Characterization and Biological activity of some α-aminophoshonates. Farmcia 2010, 58, 531–545. [Google Scholar]
  14. Bhagat, S.; Shah, P.; Garg, S.K.; Mishra, S.; Kaur, P.K.; Singh, S.; Chakraborti, A.K. α-Aminophosphonates as novel anti-leishmanialchemotypes: Synthesis, biological evaluation, and CoMFA studies. Med. Chem. Commun. 2014, 5, 665–669. [Google Scholar] [CrossRef]
  15. Lewkowski, J.; Malinowski, Z.; Matusiak, A.; Morawska, M.; Rogacz, D.; Rychter, P. The Effect of New Thiophene-Derived Aminophosphonic Derivatives on Growth of Terrestrial Plants: A Seedling Emergence and Growth Test. Molecules 2016, 21, 694. [Google Scholar] [CrossRef] [Green Version]
  16. Kim, I.-H.; Park, Y.-K.; Nishiwaki, H.; Hammock, B.D.; Nishi, K. Structure–activity relationships of amide–phosphonate derivatives as inhibitors of the human soluble epoxide hydrolase. Bioorg. Med. Chem. 2015, 23, 7199–7210. [Google Scholar] [CrossRef]
  17. Klimczak, A.A.; Matusiak, A.; Lewkowski, J.; Bitner, J.; Szemraj, J.; Kontek, R. Dimethyl (2-Furyl)-N-(2-Methoxyphenyl)Aminomethylphosphonate Induces Apoptosis in Esophageal Squamous Cancer Cells. Structure Versus Activity of its Selected Analogs. Phosphorus Sulfur Silicon Relat. Elem. 2015, 190, 1088–1099. [Google Scholar] [CrossRef]
  18. Klimczak, A.A.; Kuropatwa, A.; Lewkowski, J.; Szemraj, J. Synthesis of new N-arylamino(2-furyl)methylphosphonic acid diesters, and in vitro evaluation of their cytotoxicity against esophageal cancer cells. Med. Chem. Res. 2012, 22, 852–860. [Google Scholar] [CrossRef]
  19. Kafarski, P.; Lejczak, B. Aminophosphonic acids of potential medical importance. Curr. Med. Chem. Anti-Cancer Agents 2001, 1, 301–312. [Google Scholar] [CrossRef]
  20. Li, C.; Song, B.; Yan, K.; Xu, G.; Hu, D.; Yang, S.; Jin, L.; Xue, W.; Lu, P. One Pot Synthesis of α-Aminophosphonates Containing Bromo and 3,4,5-Trimethoxybenzyl Groups under Solvent-free Conditions. Molecules 2007, 12, 163–172. [Google Scholar] [CrossRef] [Green Version]
  21. Sonar, S.S.; Sadaphal, S.A.; Labade, V.B.; Shingate, B.B.; Shingare, M.S. An Efficient Synthesis and Antibacterial Screening of Novel Oxazepine α-Aminophosphonates by Ultrasound Approach. Phosphorus Sulfur Silicon Relat. Elem. 2009, 185, 65–73. [Google Scholar] [CrossRef]
  22. Sayed, I.E.T.E.; Fathy, G.; Ahmed, A.A.S. Synthesis and Antibacterial Activity of Novel Cyclic α-Aminophsophonates. Biomed. J. Sci. Tech. Res. 2019, 23, 17609–17614. [Google Scholar] [CrossRef]
  23. Abdel-Megeed, M.F.; Badr, B.E.; Azaam, M.M.; El–Hiti, G.A. Antimicrobial Activities of a Series of Diphenyl (4′-(Aryldiazenyl)Biphenyl-4-Ylamino)(Pyridin-3-YL)Methylphosphonates. Phosphorus Sulfur Silicon Relat. Elem. 2012, 187, 1202–1207. [Google Scholar] [CrossRef]
  24. Kudzin, Z.H.; Kudzin, M.H.; Drabowicz, J.; Stevens, C.V. Aminophosphonic Acids—Phosphorus Analogues of Natural Amino Acids. Part 1: Syntheses of α-Aminophosphonic Acids. Curr. Org. Synth. 2011, 15, 2015–2071. [Google Scholar] [CrossRef]
  25. Allen, J.G.; Atherton, F.R.; Hall, M.J.; Hassall, C.H.; Holmes, S.W.; Lambert, R.W.; Nisbet, L.J.P.; Ringrose, S. Phosphonopeptides, a new class of synthetic antibacterial agents. Nature 1978, 272, 56–58. [Google Scholar] [CrossRef]
  26. Shaik, M.S.; Nadiveedhi, M.R.; Gundluru, M.; Katike, U.; Obulam, V.S.R.; Cirandur, S.R. Efficient catalyst free green synthesis and in vitro antimicrobial, antioxidant and molecular docking studies of α-substituted aromatic/heteroaromatic aminomethylene bisphosphonates. Synth. Commun. 2021, 51, 747–764. [Google Scholar] [CrossRef]
  27. Mulla, S.A.R.; Pathan, M.Y.; Chavan, S.S.; Gample, S.P.; Sarkar, D. Highly efficient one-pot multi-component synthesis of α-aminophosphonates and bis-α-aminophosphonates catalyzed by heterogeneous reusable silica supported dodecatungstophosphoric acid (DTP/SiO2) at ambient temperature and their antitubercular evaluation against Mycobactrium Tuberculosis. RSC Adv. 2014, 4, 7666–7672. [Google Scholar] [CrossRef]
  28. Ali, N.S.; Zakir, S.; Patel, M.; Farooqui, M. Synthesis of new α aminophosphonate system bearing Indazole moiety and their biological activity. Eur. J. Med. Chem. 2012, 50, 39–43. [Google Scholar] [CrossRef]
  29. Sampath, C.; Vani, K.V.; Kotaiah, Y.; Krishna, N.H.; Raju, C.N.; Rao, C.V. A facile and efficient One-pot Three Component Reaction (Kabachinik-Fields Reaction) for the Synthesis of Novel α-Aminophosphonates by 1, 4-Dimethylpiperazine as a new catalyst. J. Chem. Pharm. Res. 2012, 4, 1375–1382. [Google Scholar]
  30. Maruyama, H.B.; Arisawa, M.; Sawada, T. Alafosfalin, a new inhibitor of cell wall biosynthesis: In vitro activity against urinary isolates in Japan and potentiation with beta-lactams. Antimicrob. Agents Chemother. 1979, 16, 444–451. [Google Scholar] [CrossRef] [Green Version]
  31. Kasthuraiah, M.; Kumar, K.A.; Reddy, C.S.; Reddy, C.D. Syntheses, spectral property, and antimicrobial activities of 6-α-amino dibenzo [d,f][1,3,2]dioxaphosphepin 6-oxides. Heteroat. Chem. 2007, 18, 2–8. [Google Scholar] [CrossRef]
  32. Kasthuraiah, M.; Balakrishna, A.; Reddy, K.R.K.K.; Kumar, B.S.; Reddy, C.S.; Nagaraju, C. Synthesis and Antimicrobial Activity of New 2,10-Dichloro-6-phenylaminobenzyl-dibenzo[d,g] [1,3,6,2]dioxathiaphosphocin 6-Oxides. Heterocycl. Chem. 2008, 45, 103–107. [Google Scholar] [CrossRef]
  33. Tajtihttps, Á.; Bálinthttps, E.; Keglevich, G. Microwave-assisted synthesis of α-aminophosphonates and related derivatives by the Kabachnik-Fields reaction. Phosphorus Sulfur Silicon Relat. Elem. 2019, 194, 379–381. [Google Scholar] [CrossRef]
  34. Maestro, A.; del Corte, X.; López-Francés, A.; de Marigorta, E.M.; Palacios, F.; Vicario, J. Asymmetric Synthesis of Tetrasubstituted α-Aminophosphonic Acid Derivatives. Molecules 2021, 26, 3202. [Google Scholar] [CrossRef] [PubMed]
  35. Viveros-Ceballos, J.L.; Ordóñez, M.; Sayago, F.J.; Cativiela, C. Stereoselective Synthesis of α-Amino-C-phosphinic Acids and Derivatives. Molecules 2016, 21, 1141. [Google Scholar] [CrossRef] [Green Version]
  36. Bálint, E.; Tajti, Á.; Kalocsai, D.; Mátravölgyi, B.; Karaghiosoff, K.; Czugler, M.; Keglevich, G. Synthesis and utilization of optically active α-aminophosphonate derivatives by Kabachnik-Fields reaction. Tetrahedron 2017, 73, 5659–5667. [Google Scholar] [CrossRef]
  37. Tajti, Á.; Bálint, E.; Keglevich, G. Synthesis of Ethyl Octyl-α-Aminophosphonate Derivatives. Curr. Org. Synth. 2016, 13, 638–645. [Google Scholar] [CrossRef] [Green Version]
  38. Philippe, N.; Denivet, F.; Vasse, J.; Santos, J.; Levacher, V.; Dupas, G. Highly stereoselective Friedel-Crafts type cyclization. Facile access to enantiopure 1,4-dihydro-4-phenyl isoquinolinones. Tetrahedron 2003, 59, 8049–8056. [Google Scholar] [CrossRef]
  39. Bálint, E.; Tajti, Á.; Drahos, L.; Ilia, G.; Keglevich, G. Alcoholysis of Dialkyl Phosphites Under Microwave Conditions. Curr. Org. Chem. 2013, 17, 555–562. [Google Scholar] [CrossRef]
  40. Gao, Y.; Huang, Z.; Zhuang, R.; Xu, J.; Zhang, P.; Tang, G.; Zhao, Y. Direct Transformation of Amides into α-Amino Phosphonates via a Reductive Phosphination Process. Org. Lett. 2013, 15, 4214–4217. [Google Scholar] [CrossRef]
  41. Maier, L.; Diel, P.J. Organic Phosphorus compounds 941 preparation, physical and biological properties of amino-arylmethylphosphonic and phoshonous acids. Phosphorus Sulfur Silicon Relat. Elem. 1991, 57, 57–64. [Google Scholar] [CrossRef]
  42. Bhagat, S.; Chakraborti, A.K. Zirconium(IV) Compounds As Efficient Catalysts for Synthesis of α-Aminophosphonates. J. Org. Chem. 2008, 73, 6029–6032. [Google Scholar] [CrossRef] [PubMed]
  43. Bedolla-Medrano, M.; Hernández-Fernández, E.; Ordóñez, M. Phenylphosphonic Acid as Efficient and Recyclable Catalyst in the Synthesis of α-Aminophosphonates under Solvent-Free Conditions. Synlett 2014, 25, 1145–1149. [Google Scholar] [CrossRef]
  44. Tibhe, G.D.; Bedolla-Medrano, M.; Cativiela, C.; Ordóñez, M. Phenylboronic Acid as Efficient and Eco-Friendly Catalyst for the One-Pot, Three-Component Synthesis of α-Aminophosphonates under Solvent-Free Conditions. Synlett 2012, 23, 1931–1936. [Google Scholar] [CrossRef]
  45. Liu, Q.; Yu, S.; Hu, L.; Hussain, M.I.; Zhang, X.; Xiong, Y. Cross-dehydrogenative coupling strategy for phosphonation and cyanation of secondary N-alkyl anilines by employing 2,3-dichloro-5,6-dicyanobenzoquinone. Tetrahedron 2018, 74, 7209–7217. [Google Scholar] [CrossRef]
  46. Lee, O.-Y.; Law, K.-L.; Yang, D. Secondary Amine Formation from Reductive Amination of Carbonyl Compounds Promoted by Lewis Acid Using the InCl3/Et3SiH System. Org. Lett. 2009, 11, 3302–3305. [Google Scholar] [CrossRef]
  47. Hall, R.G. The design and synthesis of biologically active organophosphorus compounds--the role of a central research laboratory. Chimia 2010, 64, 34–36. [Google Scholar] [CrossRef]
  48. Orsini, F.; Sello, G.; Sisti, M. Aminophosphonic Acids and Derivatives. Synthesis and Biological Applications. Curr. Med. Chem. 2010, 17, 264–289. [Google Scholar] [CrossRef]
  49. Ahmadi, F.; Assadi, Y.; Hosseini, S.M.R.M.; Rezaee, M. Determination of organophosphorus pesticides in water samples by single drop microextraction and gas chromatography-flame photometric detector. J. Chromatogr. A 2006, 1101, 307–312. [Google Scholar] [CrossRef]
  50. Amira, A.; Aouf, Z.; K’tir, H.; Chemam, Y.; Ghodbane, R.; Zerrouki, R.; Aouf, N.-E. Recent Advances in the Synthesis of α-Aminophosphonates: A Review. ChemistrySelect 2021, 6, 6137–6149. [Google Scholar] [CrossRef]
  51. Shastri, R.A. Review on the Synthesis of α-Aminophosphonate Derivatives. Chem. Sci. Trans. 2019, 8, 359–367. [Google Scholar] [CrossRef] [Green Version]
  52. Basha, M.H.; Subramanyam, C.; Rao, K.P. Ultrasound-promoted solvent-free synthesis of some new α-aminophosphonates as potential antioxidants. Main Group Met. Chem. 2020, 43, 147–153. [Google Scholar] [CrossRef]
  53. Eyckensa, D.J.; Henderson, L.C. Synthesis of α-aminophosphonates using solvate ionic liquids. RSC Adv. 2017, 7, 27900–27904. [Google Scholar] [CrossRef] [Green Version]
  54. Babak, K. Surface-Mediated Solid Phase Reactions: A Simple and New Method for the Synthesis of α-Aminophosphonates under Solvent-Free Conditions. Chem. Lett. 2001, 30, 880–881. [Google Scholar] [CrossRef]
  55. Basha, S.K.T.; Kalla, R.M.N.; Varalakshmi, M.; Sudhamani, H.; Appa, R.M.; Hong, S.C.; Raju, C.N. Heterogeneous catalyst SiO2–LaCl3·7H2O: Characterization and microwave-assisted green synthesis of α-aminophosphonates and their antimicrobial activity. Mol. Divers 2022. Online ahead of print. [Google Scholar] [CrossRef] [PubMed]
  56. Kowalczyk, D.; Albrecht, Ł. An organocatalytic biomimetic approach to α-aminophosphonates. Chem. Commun. 2015, 51, 3981–3984. [Google Scholar] [CrossRef]
  57. Chen, Y.; Li, Z.; Liu, Y.; Lin, T.; Sun, H.; Yang, D.; Jiang, C. Identification of novel and selective non-peptide inhibitors targeting the polo-box domain of polo-like kinase 1. Bioorg. Chem. 2018, 81, 278–288. [Google Scholar] [CrossRef]
  58. Sravya, G.; Suresh, G.; Zyryanov, G.V.; Balakrishna, A.; Reddy, N.B. K2CO3/Al2O3: An Efficient and Recyclable Catalyst for One-Pot, Three Components Synthesis of α-Aminophosphonates and Bioactivity Evaluation. Asian J. Org. Chem. 2019, 31, 2383–2388. [Google Scholar] [CrossRef]
  59. Chandrasekhar, S.; Prakash, S.J.; Jagadeshwar, V.; Narsihmula, C. Three component coupling catalyzed by TaCl5–SiO2: Synthesis of α-amino phosphonates. Tetrahedron Lett. 2001, 42, 5561–5563. [Google Scholar] [CrossRef]
  60. Yadav, J.S.; Reddy, B.V.S.; Raj, K.S.; Reddy, K.B.; Prasad, A.R. Zr4+-Catalyzed Efficient Synthesis of α-Aminophosphonates. Synthesis 2001, 15, 2277–2280. [Google Scholar] [CrossRef]
  61. Manjula, A.; Rao, B.V.; Neelakantan, P. One-Pot Synthesis of α-Aminophosphonates: An Inexpensive Approach. Synth. Comun. 2003, 33, 2963–2969. [Google Scholar] [CrossRef]
  62. Milen, M.; Ábrányi-Balogh, P.; Dancsó, A.; Frigyes, D.; Pongó, L.; Keglevich, G. T3P®-promoted Kabachnik–Fields reaction: An efficient synthesis of α-aminophosphonates. Tetrahedron Lett. 2013, 54, 5430–5433. [Google Scholar] [CrossRef]
  63. Lee, S.G.; Lee, J.K.; Song, C.-E.; Kim, D.C. Microwave-assisted Kabachnik-Fields Reaction in Ionic Liquid. Bull. Korean Chem. Soc. 2002, 23, 667–668. [Google Scholar] [CrossRef] [Green Version]
  64. Janicki, I.; Łyżwa, P.; Kiełbasiński, P. The first enzyme-promoted addition of nitromethane to imines (aza-Henry reaction). Bioorg. Chem. 2020, 94, 103377. [Google Scholar] [CrossRef]
  65. Cai, J.-F.; Guan, Z.; He, Y.-Y. The lipase-catalyzed asymmetric C-C Michael addition. J. Mol. Catal. B Enzym. 2011, 68, 240–244. [Google Scholar] [CrossRef]
  66. Li, C.; Feng, X.-W.; Wang, N.; Zhou, Y.-J.; Yu, X.-Q. Biocatalytic promiscuity: The first lipase-catalysed asymmetric aldol reaction. Green Chem. 2008, 10, 616–618. [Google Scholar] [CrossRef]
  67. Albuquerque, T.B.; da Silva, C.D.G.; de Oliveira, A.R.; Santos, B.F.d.; da Silva, B.A.L.; Katla, R.; Rochaa, M.P.D.; Domingues, N.L.C. Lipase catalyzed 1,2-addition of thiols to imines under mild conditions. New J. Chem. 2018, 42, 1642–1645. [Google Scholar] [CrossRef]
  68. Reetz, M.T.; Mondière, R.; Carballeira, J.D. Enzyme promiscuity: First protein-catalyzed Morita–Baylis–Hillman reaction. Tetrahedron Lett. 2007, 48, 1679–1681. [Google Scholar] [CrossRef]
  69. Kapoor, M.; Gupta, M.N. Lipase promiscuity and its biochemical applications. Proc. Biochem. 2012, 47, 555–569. [Google Scholar] [CrossRef]
  70. Dwivedee, B.P.; Soni, S.; Sharma, M.; Bhaumik, J.; Laha, J.K.; Banerjee, U.C. Promiscuity of Lipase-Catalyzed Reactions for Organic Synthesis: A Recent Update. ChemistrySelect 2018, 3, 2441–2466. [Google Scholar] [CrossRef]
  71. Guezane-Lakoud, S.; Toffano, M.; Aribi-Zouioueche, L. Promiscuous lipase catalyzed a new P–C bond formation: Green and efficient protocol for one-pot synthesis of α-aminophosphonates. Heteroat. Chem. 2017, 28, e21408. [Google Scholar] [CrossRef] [Green Version]
  72. Aissa, R.; Guezane-Lakoud, S.; Kolodziej, E.; Toffano, M.; Aribi-Zouioueche, L. Diastereoselective synthesis of bis(α-aminophosphonates) by lipase catalytic promiscuity. New J. Chem. 2019, 43, 8153–8159. [Google Scholar] [CrossRef]
  73. Chavan, A.S.; Kharat, A.S.; Bhosle, R.; Dhumal, S.T.; Mane, R.A. CAL-B accelerated novel synthetic protocols for 3,3’-arylidenebis-4-hydroxycoumarins and dimethyl ((substituted phenyl) (phenylamino)methyl) phosphonates. Res. Chem. Intermed. 2021, 47, 4497–4512. [Google Scholar] [CrossRef]
  74. Kowalczyk, P.; Koszelewski, D.; Gawdzik, B.; Samsonowicz-Górski, J.; Kramkowski, K.; Wypych, A.; Lizut, R.; Ostaszewski, R. Promiscuous Lipase-Catalyzed Markovnikov Addition of H-Phosphites to Vinyl Esters for the Synthesis of Cytotoxic α-Acyloxy Phosphonate Derivatives. Materials 2022, 15, 1975. [Google Scholar] [CrossRef] [PubMed]
  75. Koszelewski, D.; Brodzka, A.; Madej, A.; Trzepizur, D.; Ostaszewski, R. Evaluation of gem-Diacetates as Alternative Reagents for Enzymatic Regio- and Stereoselective Acylation of Alcohols. J. Org. Chem. 2021, 86, 6331–6342. [Google Scholar] [CrossRef]
  76. Koszelewski, D.; Ostaszewski, R.; Śmigielski, P.; Hrunyk, A.; Kramkowski, K.; Laskowski, Ł.; Laskowska, M.; Lizut, R.; Szymczak, M.; Michalski, J.; et al. Pyridine Derivatives—A New Class of Compounds That Are Toxic to E. coli K12, R2–R4 Strains. Materials 2021, 14, 5401. [Google Scholar] [CrossRef] [PubMed]
  77. Koszelewski, D.; Ostaszewski, R. The studies on chemoselective promiscuous activity of hydrolases on acylals transformations. Bioorg. Chem. 2019, 93, 102825. [Google Scholar] [CrossRef]
  78. Koszelewski, D.; Ostaszewski, R. Enzyme promiscuity as a remedy for the common problems with Knoevenagel condensation. Chem. Eur. J. 2019, 25, 10156–10164. [Google Scholar] [CrossRef]
  79. Koszelewski, D.; Ostaszewski, R. Biocatalytic Promiscuity of Lipases in Carbon-Phosphorus Bond Formation. ChemCatChem 2019, 11, 2554–2558. [Google Scholar] [CrossRef]
  80. Albanese, D.C.M.; Gaggero, N. Albumin as a promiscuous biocatalyst in organic synthesis. RSC Adv. 2015, 5, 10588–10598. [Google Scholar] [CrossRef] [Green Version]
  81. Fang, H.; Xie, X.; Hong, B.; Zhao, Y.; Fang, M. Copper (I) Iodide-Catalyzed Solvent-Free Synthesis of α-Aminophosphonates. Phosphorus Sulfur Silicon Relat. Elem. 2011, 186, 2145–2155. [Google Scholar] [CrossRef]
  82. Kandula, M.K.R.; Gundluru, M.; Nemallapudi, B.R.; Gundala, S.; Kotha, P.; Zyryanov, G.V.; Chadive, S.; Cirandur, S.R. Synthesis, antioxidant activity, and α-glucosidase enzyme inhibition of α-aminophosphonate derivatives bearing piperazine-1,2,3-triazole moiety. J. Heterocycl. Chem. 2021, 58, 172–181. [Google Scholar] [CrossRef]
  83. Azaam, M.M.; Kenawy, E.R.; El-din, A.S.B.; Khamis, A.A.; El-Magd, M.A. Antioxidant and anticancer activities of α-aminophosphonates containing thiadiazole moiety. J. Saudi. Chem. Soc. 2018, 22, 34–41. [Google Scholar] [CrossRef]
  84. Bollinger, A.; Molitor, R.; Thies, S.; Koch, R.; Coscolín, C.; Ferrer, M.; Jaeger, K.-E. Organic-Solvent-Tolerant Carboxylic Ester Hydrolases for Organic Synthesis. Appl. Environ. Microbiol. 2020, 86, e00106-20. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  85. Kowalczyk, P.; Wilk, M.; Parul, P.; Szymczak, M.; Kramkowski, K.; Raj, S.; Skiba, G.; Sulejczak, D.; Kleczkowska, P.; Ostaszewski, R. The Synthesis and Evaluation of Aminocoumarin Peptidomimetics as Cytotoxic Agents on Model Bacterial E. coli Strains. Materials 2021, 14, 5725. [Google Scholar] [CrossRef] [PubMed]
  86. Samsonowicz-Górski, J.; Kowalczyk, P.; Koszelewski, D.; Brodzka, A.; Szymczak, M.; Kramkowski, K.; Ostaszewski, R. The Synthesis and Evaluation of Amidoximes as Cytotoxic Agents on Model Bacterial E. coli Strains. Materials 2021, 14, 7577. [Google Scholar] [CrossRef]
  87. Kowalczyk, P.; Trzepizur, D.; Szymczak, M.; Skiba, G.; Kramkowski, K.; Ostaszewski, R. 1,2-Diarylethanols—A New Class of Compounds That Are Toxic to E. coli K12, R2–R4 Strains. Materials 2021, 14, 1025. [Google Scholar] [CrossRef]
  88. Kowalczyk, P.; Madej, A.; Szymczak, M.; Ostaszewski, R. α-Amidoamids as New Replacements of Antibiotics—Research on the Chosen K12, R2–R4 E. coli Strains. Materials 2020, 13, 5169. [Google Scholar] [CrossRef]
  89. Kowalczyk, P.; Borkowski, A.; Czerwonka, G.; Cłapa, T.; Cieśla, J.; Misiewicz, A.; Borowiec, M.; Szala, M. The microbial toxicity of quaternary ammonium ionic liquids is dependent on the type of lipopolysaccharide. J. Mol. Liq. 2018, 266, 540–547. [Google Scholar] [CrossRef]
  90. Borkowski, A.; Kowalczyk, P.; Czerwonka, G.; Cieśla, J.; Cłapa, T.; Misiewicz, A.; Szala, M.; Drabik, M. Interaction of quaternary ammonium ionic liquids with bacterial membranes—Studies with Escherichia coli R1–R4-type lipopolysaccharides. J. Mol. Liq. 2017, 246, 282–289. [Google Scholar] [CrossRef]
  91. Kowalczyk, P.; Gawdzik, B.; Trzepizur, D.; Szymczak, M.; Skiba, G.; Raj, S.; Kramkowski, K.; Lizut, R.; Ostaszewski, R. δ-Lactones—A New Class of Compounds That Are Toxic to E. coli K12 and R2–R4 Strains. Materials 2021, 14, 2956. [Google Scholar] [CrossRef] [PubMed]
  92. Maciejewska, A.; Kaszowska, M.; Jachymek, W.; Lugowski, C.; Lukasiewicz, J. Lipopolysaccharide-linked Enterobacterial Common Antigen (ECALPS) Occurs in Rough Strains of Escherichia coli R1, R2, and R4. Int. J. Mol. Sci. 2020, 21, 6038. [Google Scholar] [CrossRef] [PubMed]
  93. Prost, M.E.; Prost, R. Basic parameters of evaluation of the effectiveness of antibiotic therapy. OphthaTherapy 2017, 4, 233–236. [Google Scholar] [CrossRef]
  94. Kabachnik, M.I.; Medved, T.Y. Нoвый метoд синтеза сс-аминoфoсфинoвых кислoт [A new method for the synthesis of α-amino phosphoric acids]. Doklady Akademii Nauk SSSR 1952, 83, 689ff. [Google Scholar]
  95. Fields, E.K. The synthesis of esters of substituted amino phosphonic acids. J. Am. Chem. Soc. 1952, 74, 1528–1531. [Google Scholar] [CrossRef]
Figure 1. Biologically active antimicrobial α-aminophosphonate derivatives.
Figure 1. Biologically active antimicrobial α-aminophosphonate derivatives.
Materials 15 03846 g001
Figure 2. α-Aminophosphonates 116 obtained via an enzyme-catalyzed Kabachnik–Fields reaction. Yields in brackets provided for isolated products 116.
Figure 2. α-Aminophosphonates 116 obtained via an enzyme-catalyzed Kabachnik–Fields reaction. Yields in brackets provided for isolated products 116.
Materials 15 03846 g002
Scheme 1. Enzyme-catalyzed synthesis of α-aminophosphonates 116.
Scheme 1. Enzyme-catalyzed synthesis of α-aminophosphonates 116.
Materials 15 03846 sch001
Scheme 2. Plausible mechanism of the porcine pancreas lipase-catalyzed Kabachnik−Fields reaction.
Scheme 2. Plausible mechanism of the porcine pancreas lipase-catalyzed Kabachnik−Fields reaction.
Materials 15 03846 sch002
Figure 3. Minimum inhibitory concentration (MIC) of the phosphonate derivatives in model bacterial strains. The x-axis features compounds 116 used sequentially. The y-axis shows the MIC value in µg/mL−1. Investigated strains of E. coli K12 as the control (blue), R2 strains (orange), R3 strains (grey), and R4 strains (yellow). The order in which the compounds were applied to the plate is shown in Supplementary Materials Figure S1.
Figure 3. Minimum inhibitory concentration (MIC) of the phosphonate derivatives in model bacterial strains. The x-axis features compounds 116 used sequentially. The y-axis shows the MIC value in µg/mL−1. Investigated strains of E. coli K12 as the control (blue), R2 strains (orange), R3 strains (grey), and R4 strains (yellow). The order in which the compounds were applied to the plate is shown in Supplementary Materials Figure S1.
Materials 15 03846 g003
Figure 4. Minimum bactericidal concentration (MBC) of the phosphonate derivatives. The x-axis features compounds 116 used sequentially. The y-axis shows the MIC value in µg/mL−1. Investigated strains of E. coli K12 as control (blue), R2 strains (orange), R3 strains (grey), and R4 strains (yellow). The order in which the compounds were applied to the plate is shown in Supplementary Materials Figure S1.
Figure 4. Minimum bactericidal concentration (MBC) of the phosphonate derivatives. The x-axis features compounds 116 used sequentially. The y-axis shows the MIC value in µg/mL−1. Investigated strains of E. coli K12 as control (blue), R2 strains (orange), R3 strains (grey), and R4 strains (yellow). The order in which the compounds were applied to the plate is shown in Supplementary Materials Figure S1.
Materials 15 03846 g004
Figure 5. The ratio of MBC/MIC of the phosphonate derivatives. The x-axis features compounds 116 used sequentially. The y-axis shows the MIC value in µg/mL−1. Investigated strains of E. coli K12 as control (blue), R2 strains (orange), R3 strains (grey), and R4 strains (yellow). The order in which the compounds were applied to the plate is shown in Supplementary Materials Figure S1.
Figure 5. The ratio of MBC/MIC of the phosphonate derivatives. The x-axis features compounds 116 used sequentially. The y-axis shows the MIC value in µg/mL−1. Investigated strains of E. coli K12 as control (blue), R2 strains (orange), R3 strains (grey), and R4 strains (yellow). The order in which the compounds were applied to the plate is shown in Supplementary Materials Figure S1.
Materials 15 03846 g005
Figure 8. Percentage of bacterial DNA recognized by the Fpg enzymes in model bacterial strains after ciprofloxacin, bleomycin, and cloxacillin treatment. The compounds were statistically significant at p < 0.05.
Figure 8. Percentage of bacterial DNA recognized by the Fpg enzymes in model bacterial strains after ciprofloxacin, bleomycin, and cloxacillin treatment. The compounds were statistically significant at p < 0.05.
Materials 15 03846 g008
Table 1. Model Kabachnik–Fields reaction catalyzed by enzymes. [a] Optimization studies.
Table 1. Model Kabachnik–Fields reaction catalyzed by enzymes. [a] Optimization studies.
EntryCatalystT (°C)SolventYield [%] f
1None25neat<5
2Porcine pancreas lipase (PpL)25neat73
3Porcine pancreas lipase (PpL)25Toluene64
4Porcine pancreas lipase (PpL)25EtOAc18
5Porcine pancreas lipase (PpL)25THF52
6Porcine pancreas lipase (PpL)252-Me THF55
7Porcine pancreas lipase (PpL)25TBME88
8Porcine pancreas lipase (PpL)30TBME91
9Porcine pancreas lipase (PpL)40TBME87
10Porcine pancreas lipase (PpL) b30TBME93
11Wheat germ lipase20neat29
12Pseudomonas cepacia lipase (PfL)20neat44
13Candida cylindracea lipase (CcL)20neat57
14Candida rugosa lipase (CrL)20neat35
15Novozym 43520neat67
16Bovine serum albumin (BSA)30TBME9
17Bovine liver acetone powder (BLAP) c20neat43
18Denatured PpL d30TBME<1
19CuI e25neat39
20Cu2O e25neat24
21Cu(OAc)2 e25neat33
22PhB(OH)2 e25neat14
a Reaction conditions: benzaldehyde (1 mmol), benzylamine (1 mmol), dimethyl phosphite (1 mmol), and enzyme (50 mg) in a solvent (2 mL) for 24 h, 200 rpm; b PpL (80 mg); c domestically prepared; d thermally deactivated at 100 °C for 24 h; e 10 mol%; f yield of the isolated product 1 after chromatography on silica gel.
Table 2. Statistical analysis of all analyzed compounds by MIC, MBC, and MBC/MIC; <0.05 *, <0.01 **, <0.001 ***.
Table 2. Statistical analysis of all analyzed compounds by MIC, MBC, and MBC/MIC; <0.05 *, <0.01 **, <0.001 ***.
No. of Samples131516Type of Test
K12*********MIC
R2*********MIC
R3*********MIC
R4*********MIC
K12*****MBC
R2*****MBC
R3*****MBC
R4*****MBC
K12*******MBC/MIC
R2*******MBC/MIC
R3*******MBC/MIC
R4*******MBC/MIC
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Koszelewski, D.; Kowalczyk, P.; Śmigielski, P.; Samsonowicz-Górski, J.; Kramkowski, K.; Wypych, A.; Szymczak, M.; Ostaszewski, R. Relationship between Structure and Antibacterial Activity of α-Aminophosphonate Derivatives Obtained via Lipase-Catalyzed Kabachnik−Fields Reaction. Materials 2022, 15, 3846. https://doi.org/10.3390/ma15113846

AMA Style

Koszelewski D, Kowalczyk P, Śmigielski P, Samsonowicz-Górski J, Kramkowski K, Wypych A, Szymczak M, Ostaszewski R. Relationship between Structure and Antibacterial Activity of α-Aminophosphonate Derivatives Obtained via Lipase-Catalyzed Kabachnik−Fields Reaction. Materials. 2022; 15(11):3846. https://doi.org/10.3390/ma15113846

Chicago/Turabian Style

Koszelewski, Dominik, Paweł Kowalczyk, Paweł Śmigielski, Jan Samsonowicz-Górski, Karol Kramkowski, Aleksandra Wypych, Mateusz Szymczak, and Ryszard Ostaszewski. 2022. "Relationship between Structure and Antibacterial Activity of α-Aminophosphonate Derivatives Obtained via Lipase-Catalyzed Kabachnik−Fields Reaction" Materials 15, no. 11: 3846. https://doi.org/10.3390/ma15113846

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

Article Metrics

Back to TopTop