Share The Article

Hey there! If you're enjoying the article you're reading, why not share it with your friends and spread the knowledge? Let's make sure everyone gets a chance to benefit from this great read!

You can also tag us on social media and we would be happy to re-post it. Here are our social media accounts:

Instagram: @etflin
Twitter: @Etflin1
Facebook: Etflin

Cite The Article

Export the citation:




Citation
ACS Style

Sangma, C.D., Chetia, D., Borthakur, M.S., Patowary, L., Tayeng, D. In-silico design and screening of cephalosporin derivatives for their inhibitory potential against Haemophilus influenza. Sciences of Phytochemistry 2022, 1(2), 1-10.

AMA Style

Sangma, CD, Chetia, D, Borthakur, MS, Patowary, L, Tayeng, D. In-silico design and screening of cephalosporin derivatives for their inhibitory potential against Haemophilus influenza. Sciences of Phytochemistry. 2022; 1(2):1-10.

Chicago Style

Chikame Dawa Sangma, Dipak Chetia, Malita Sarma Borthakur, Lima Patowary, Dubom Tayeng. 2022. "In-silico design and screening of cephalosporin derivatives for their inhibitory potential against Haemophilus influenza" Sciences of Phytochemistry 1, no. 2:1-10.

The Article's Metrics

AI Dimensions Metrics


PlumX Metrics by Elsevier

In-silico design and screening of cephalosporin derivatives for their inhibitory potential against Haemophilus influenza

Article Access

Views: 993
Downloads: 30

Corresponding Author

Affiliation

Contribution

ORCID


Check the author works here


Reference



Check the reference here


Article's Figures
Latest Articles from Sciences of Phytochemistry

Table of Contents

(clickable & vertically scrollable)

Home / Sciences of Phytochemistry / Volume 1 Issue 2 / 10.58920/sciphy01020001

Research Article

In-silico design and screening of cephalosporin derivatives for their inhibitory potential against Haemophilus influenza

by Chikame Dawa Sangma, Dipak Chetia, Malita Sarma Borthakur, Lima Patowary, Dubom Tayeng

Academic editor: James H. Zothantluanga
Sciences of Phytochemistry 1(2): 1-10 (2022); https://doi.org/10.58920/sciphy01020001
This article is licensed under the Creative Commons Attribution (CC BY) 4.0 International License.


Received
23 Aug 2022
Revised
12 Oct 2022
Accepted
12 Oct 2022
Published
12 Oct 2022

Abstract: Antibiotics kill bacteria by blocking essential metabolic processes which prevent them from reproducing thereby allowing the immune system to fight bacterial infections. However, the emergence and the quick spread of bacterial resistance against clinically approved antibiotics have become alarming. This necessitates the development of novel treatment options and alternative antimicrobial therapies in the fight against bacterial infections. In this study, we aim to virtually design and carry out in-silico studies to identify a cephalosporin derivative with inhibitory potential against Haemophilus influenza. Data Warrior software, Discovery studio software, PyRx tool, Swiss ADME web tool, and ProTox-II web tool were used to screen the cephalosporin derivatives. Initially, 17 cephalosporin derivatives were preliminarily screened for their toxicity followed by in-silico ADME studies. Among the cephalosporin derivatives, C1, C6, and C12 were found to be the potential drug-like molecules with binding energies of -7.4 kcal/mol, -7.1 kcal/mol, and -7.1 kcal/mol, respectively. In particular, C1 was predicted to have a moderate biological activity with a high bioavailability score. Based on the ADME profile, toxicity, binding energy, drug-likeness, and drug score, we conclude C1 (‘F’ at the 3rd position) as the potential lead molecule to inhibit H. influenza.

Keywords: In-silicoCephalosporinsAntibioticsDrug resistanceHaemophilus influenza


1. Introduction

Antibiotics are substances that kill or inhibit the growth of living organisms. Antibiotics operate by killing bacteria, blocking essential bacterial processes, preventing them from proliferating, and allowing the immune system to fight bacterial infection [1]. Antibiotics have been used in the health care system for many years and recently, it was found that some bacterial infections were resistant to the current antibiotics. Antibiotic resistance is a major global problem as it is associated with high morbidity and mortality rates [2]. Resistance of bacterial pathogens to multiple antimicrobial therapies is increasing at an alarming rate and this is known as multidrug resistance. Antibiotic resistance rises rapidly with poor infection control practices and these resistant bacteria spread easily to other patients [3]. The current shortage of effective drugs, lack of successful prevention measures, and only a few new antibiotics in the clinical pipeline have necessitated the need to develop novel treatment options to fight drug-resistant bacteria [3].

Cephalosporins (Figure 1) belonged to the class of broad-spectrum beta-lactam antibiotics. They have been in clinical use since their discovery during the 1960s [4]. In comparison to other drugs, cephalosporins have a low rate of drug-related adverse effects and their pharmacokinetic characteristics are favorable [4]. Cephalosporins are commonly utilized as the first-line treatment for pneumonia, meningitis, gonorrhea, and other microbial infections [4]. The basic structure of cephalosporins (C15H21N3O7S) consists of a four-member beta-lactam ring. In cephalosporin, the ring is condensed with a six-member sulfur ring (the dihydrothiazine ring) [5]. The substituent position 7 determines stability while the group at position 3 determines metabolic stability and pharmacokinetic characteristics [6].


Figure 1 Basic structure of Cephalosporin


Cephalosporins work by interfering with the synthesis of peptidoglycan which is a major structural component of the bacterial cell wall [4]. Carboxypeptidases, endopeptidases, and transpeptidases cross the cytoplasmic membrane and insert themselves into the cross-linked peptidoglycans. These enzymes are known as penicillin-binding proteins (PBPs) and are the targets of beta-lactam medicines. The cephalosporin amide group is identical to the peptidoglycan pentapeptide. Cephalosporin attaches covalently to the PBPs, renders them inert, significantly compromises the structural integrity of the peptidoglycan, and kills the bacteria [4]. In this study, we will virtually design a few derivatives of cephalosporins and use in-silico techniques to evaluate their inhibitory potential against the PBP 4 complexed with the novel beta-lactam (FMZ) of H. influenza.

2.    Materials and methods

2.1         Hardware

The device used for the study was a personal laptop Device name: LAPTOP-KM8A50JS, Processor: AMD 3020e with Radeon Graphics, System type: 64-bit operating system x64-based processor.

2.2         Selection of target protein

The protein selected for the study bears a PDB ID: 3A3F. It is a crystal structure of PBP-4 from Haemophilus influenza, complexed with the novel beta-lactam (FMZ). The protein has a resolution of 2.10 Ã… with two chains i.e., chain A and chain B. There are 453 amino acids (total sequence length) and the organism is H. influenza.

2.3         Preparation of protein

The crystal structure of the PBP-4 from H. influenza complexed with novel beta-lactam (FMZ) was downloaded in the PDB file format from the RSCB website (https://www.rcsb.org/). The Biovia Discovery Studio software was used to prepare the protein. Water molecules, hetero atoms, and chain B were removed from the protein. Polar hydrogens were added to the protein. The prepared protein was saved in the PDB file format. The 2D crystal structure of the prepared protein is shown in Figure 2.


Figure 2 The 2D crystal structure of the prepared protein

2.4         Identification of the active binding site coordinates

The original target protein complexed with FMZ was loaded into the PyRx virtual screening platform. First of all, chain B was removed from the scene. The prepared protein was also loaded onto the virtual screening platform. The amino acid residues which made up the original protein were revealed by expanding the protein. The position of the native ligand was identified and the 3D binding affinity grid box was adjusted to cover the entire active site residues. Following this, the original protein was removed from the scene.

2.5         Molecular docking simulation studies

The compounds were loaded and their energy was minimized with the PyRx virtual tool. As we had already identified the active binding site, docking was executed. The results were saved as comma-separated values (CSV) files.

2.6         Bioavailability studies

SWISS ADME was used to study the Lipinski rule of 5 along with gastrointestinal (GI) absorption and blood-brain barrier permeant. The SMILE CODES of all the derivatives were uploaded and all the information displayed was copied to an MS Excel file. The purpose of this assessment was to find out whether the compounds follow the Lipinski rule of 5 (Molecular weight less than 500 Daltons, hydrogen bond donors less than 5, hydrogen bond acceptors no more than 10, Octanol-water partition coefficient/log P not greater than 5).

2.7         Toxicity study

Toxicity analysis was done to assess the safety of a particular compound. It helps us to determine whether a compound or molecule, negatively impacts the normal biological function of an organism. OSIRIS Data warrior v5.5.0 was used for this process. The SMILES CODE of all the derivatives prepared was uploaded into the Data Warrior software, and each derivative was checked for mutagenicity, tumorigenicity, reproductive effective, and skin irritation. The toxic compounds were eliminated.

2.8         Study of interactions between the cephalosporin analogs and the protein

BIOVIA Discovery Studios Visualizer v21.1.0.20298 was opened and the prepared protein was loaded. The protein was defined as the receptor and the cephalosporin derivatives were defined as the ligand. The molecular interaction was observed in the form of a 2D diagram. The process was repeated for all the other derivatives.

3.    Results

3.1 Target protein and cephalosporin derivatives

The crystal structure of chain A of PBP protein was retrieved from the RCSB-PDB website, shown in Figure 3. Cephalosporins, like other beta-lactam medicines, work by interfering with the synthesis of peptidoglycan, which is a major structural component of the bacterial cell wall. Modifications in the molecule were made at the R3 position. A total of 17 Cephalosporin derivatives were prepared. The SMILES ID of all the compounds was generated with the Chem Draw Professionals 16.0 software. Their 2D chemical structures along with their respective compound code are listed in Table 1.


Figure 3 Chain A of the penicillin-binding protein with co-crystal ligand


Table 1 List of Cephalosporin Derivatives

Compound code

Compound Structure

C1 

C2

C3

C4

C5

C6

C7

C8

C9

C10

C11

C12

C13

C14

C15

C16

C17

3.2          Molecular docking simulation study

Molecular docking is an efficient tool for in-silico drug screening. To rate the binding potential of ligands to a protein, the PyRx tool can produce the binding affinity values (kcal/mol) for each ligand, and the binding potential of test ligands toward a protein can be ranked respectively by using the binding affinity of the native ligand as the benchmark. The binding affinity of the compounds is shown in Table 2.


Table 2 The binding energy of compound derivatives

Compounds

Binding Affinity (kcal/mol)

Cephalosporin

-7.4

Native ligand (FMZ)

-7.1

C1

-7.4

C2

-6.8

C3

-6.2

C4

-6.2

C5

-6.9

C6

-7.1

C7

-6.4

C8

-6.3

C9

-6.3

C10

-6.8

C11

-5.8

C12

-7.1

C13

-6.3

C14

-6.5

C15

-5.8

C16

-6.7

C17

-6.3

3.3          Preliminary ADME studies with Lipinski’s rule

After determining the binding affinity of the cephalosporin derivatives, the ADME properties of the compounds were determined. Compounds that are active under in-vitro settings can show lower activity under in-vivo conditions due to poor ADME properties. Therefore, the bioavailability of a compound needs to be studied [9, 10]. Lipinski’s rule of five also known as Pfizer’s rule of five or rule of five is a rule of thumb to evaluate drug-likeness or to determine if the chemical compound with certain pharmacological or biological activity has chemical properties and physical properties that would make it a likely orally active drug in human.

Lipinski’s rule of 5 is:

1)  Not more than 5 hydrogen bond donors

2)  Not more than 10 hydrogen bond acceptors

3)  Molecular mass less than 500 Daltons

4)  Log P does not exceed 5

The ADME properties of the cephalosporin derivatives are given in Table 3. All the compounds corroborate to specified parameters of Lipinski’s rule of 5. It is assumed that they will most likely be bioavailable. Since there were no Lipinski violations, therefore, the cephalosporin derivatives that were free from bioavailability issues were subjected to further studies.

Table 3 Swiss ADME against Lipinski’s rule of five

Compounds

MW

#H-bond acceptors

#H-bond donors

GI absorption

BBB permeant

Lipinski #violations

Leadlikeness #violations

C1

260.24

5

2

High

No

0

0

C2

276.7

4

2

High

No

0

0

C3

321.15

4

2

High

No

0

0

C4

368.15

4

2

High

No

0

1

C5

287.25

6

2

Low

No

0

0

C6

286.26

4

2

High

No

0

0

C7

267.26

4

2

High

No

0

0

C8

270.26

4

2

High

No

0

0

C9

257.27

5

2

High

No

0

0

C10

271.29

5

2

High

No

0

0

C11

285.32

4

2

High

No

0

0

C12

258.25

6

3

Low

No

0

0

C 13

272.28

4

3

Low

No

0

0

C14

256.28

4

3

High

No

0

0

C 15

270.3

4

2

High

No

0

0

C16

268.29

5

2

Low

No

0

0

C17

266.27

4

2

High

No

0

0

3.4          Toxicity analysis

Toxicity often leads to the withdrawal of drugs from clinical use [7]. Data Warrior is a reliable software used by many researchers to predict the toxicity of compounds [8, 9]. So, for the next step in designing the cephalosporin derivatives, we carried out an in-silico toxicity study to determine the toxicity of the compounds. Out of 17 compounds, C2, C3, C4, C5, and C6 showed no signs of toxicity. The results of the toxicity analysis of the Cephalosporin derivatives are given in Table 4.

Table 4 Toxicity Analysis of 17 compounds

Compounds

Mutagenicity

Carcinogenicity

Hepatotoxicity

Immunotoxicity

Cytotoxicity

C1

Inactive

Inactive

Active

Active

Inactive

C2

Inactive

Inactive

Inactive

Inactive

Inactive

C3

Inactive

Inactive

Inactive

Inactive

Inactive

C4

Inactive

Inactive

Inactive

Inactive

Inactive

C5

Inactive

Inactive

Inactive

Inactive

Inactive

C6

Inactive

Inactive

Inactive

Inactive

Inactive

C7

Inactive

Inactive

Active

Active

Inactive

C8

Inactive

Inactive

Active

Active

Inactive

C9

Inactive

Inactive

Active

Active

Inactive

C10

Inactive

Inactive

Active

Active

Inactive

C11

Inactive

Inactive

Active

Active

Inactive

C12

Inactive

Inactive

Active

Active

Inactive

C13

Inactive

Inactive

Active

Active

Inactive

C14

Inactive

Inactive

Active

Active

Inactive

C15

Inactive

Inactive

Active

Active

Inactive

C16

Inactive

Inactive

Active

Active

Inactive

C17

Inactive

Inactive

Active

Active

Inactive

3.5   Binding energy of selected compounds

After comparing the binding energy of the original cephalosporin and the native ligand with the Compounds, it was found that C1, C6, and C12 compound derivatives have a better binding affinity than cephalosporin and the native ligand. The binding energy comparison between the original cephalosporin, native ligand, C1, C6, and C12 is shown in Table 5.

Table 5 Binding energy comparison between Cephalosporin, Native Ligand, and C1, C6, and C12

Code

Binding energy (kcal/mol)

Cephalosporin

-7.4

Native Ligand (FMZ)

-7.1

C1

-7.4

C6

-7.1

C12

-7.1

3.6   Interaction of compounds with target

Visualization of the 2D ligand interactions was done with the Discovery Studio Visualizer software. The summary of the ligand interactions of each compound with the amino acids of the proteins is shown in Table 6. The images of the 2D ligand interactions of the Compounds with protein are given in Figure 3.

Table 6 Summary of ligand interaction with the active sites of penicillin-binding protein

Compound

Conventional Hydrogen bond

Other Interactive sites

Cephalosporin

ILE159 (1.84 Ã…, 2.85 Ã…), SER423 (3.06 Ã…), LEU362 (2.20 Ã…)

ASN312 (3.79Ã…), SER69 (3.60Ã…)

FMZ

ASP162 (2.41 Ã…, 2.48 Ã…), LEU362 (2.95 Ã…), LYS425 (2.00 Ã…)

PHE167 (5.07Ã…), LEU424 (5.37Ã…), ASP162 (3.41Ã…)

C1

ILE159 (2.10Ã…, 2.34Ã…), ASN168 (2.78Ã…)

ILE59 (4.93Ã…), LEU367 (3.44Ã…), LEU424 (4.33Ã…)

C6

ILE159 (2.15Ã…), ASN168 (2.78)

ILE159 (5.06Ã…), LEU362 (5.50Ã…), LEU424 (4.23Ã…), LEU424 (3.44Ã…)

C12

ASN168 (2.51Ã…), ARG364 (4.08), LYS425 (1.81Ã…, 2.73Ã…)

PHE167 (4.89Ã…), LEU362 (4.80Ã…)

Figure 4 Visualization of 2D interaction of (A) Cephalosporin, (B) Native ligand and (C) Compound 1, (D) Compound 6, and (E) Compound 12

4.    Discussion

Molecular docking is a computational technique to search for an appropriate ligand that fits both energetically and geometrically at the binding site of a protein. A more negative binding affinity value suggests a better binding between a compound and a protein. A low binding affinity value also indicates the low energy requirement for protein-ligand binding [11]. Since the initial position has the highest binding affinity and the last pose has the lowest binding affinity to the target protein, the first pose is always regarded as the optimal pose. Out of all the cephalosporin derivatives, C1, C6, and C12 have shown a better binding affinity towards Chain A of the protein as compared to the binding affinity of the native ligand. C1 was found to have more binding affinity compared to the other 2 derivatives. In a toxicity study of the compounds, C1 was found to have toxicity for immunotoxicity and hepatoxicity, and C6 and C12 passed the toxicity. But still, we consider C1 for further study as we believe the benefit will outweigh the risks.

Nowadays, researchers use innovative research techniques that deviate from the conventional methods used to study synthetic drugs. To find promising synthetic compounds for the treatment of various diseases, in-silico methods like molecular docking and molecular dynamics simulations have been used more and more in drug discovery research [9, 12, 13–16]. Some drugs, however, have a low oral bioavailability. Many researchers have chosen innovative drug delivery systems as the solution to the bioavailability problems related to drugs. Drug discovery and development processes have also used artificial intelligence, unsupervised machine learning, and supervised machine learning [11]. Pharmaceutical researchers are utilizing novel methods in drug discovery programs as a result of scientific advancements. To identify a potent cephalosporin derivative against Haemophilus influenza, we have used the in-silico methods that are affordable, safe, and sustainable.

5.    Conclusions

The rate at which antibiotic resistance develops among bacterial species continues to increase due to the continued improper use of antibiotics and gene transfer among different species. To discover and develop new cephalosporins similar to the original cephalosporin, we designed and optimized various cephalosporins analogs using virtual screening and molecular docking analysis and compared them against cephalosporin and FMZ as the standard drugs. We identified some analogs that were good candidates and displayed better in-silico activity against H. influenza. After analyzing all the parameters, including ADME properties, toxicity data, binding energy, drug-likeness, and drug score, cephalosporin with fluorine substitution at the 3rd position was identified as the best analog as this showed a good binding affinity with the target protein. In the future, the analog can be synthesized and evaluated for the in-vitro activity to confirm the in-silico antibacterial potency displayed by the compound.


Declarations

Ethics Statement

Not applicable.

Data Availability

The unpublished data is available upon request to the corresponding author.

Funding Information

Not applicable.

Conflict of Interest

The authors declare no conflicting interest.

Reference

  1. Jeśman C, Młudzik A, Cybulska M (2011) History of antibiotics and sulphonamides discoveries. Polski merkuriusz lekarski: organ Polskiego Towarzystwa Lekarskiego. 30(179):320–322.
  2. Akova M (2016) Epidemiology of antimicrobial resistance in bloodstream infections. Virulence 7(3):252–266.
  3. Mühlen S, Dersch P (2016) Anti-virulence Strategies to Target Bacterial Infections. Current topics in microbiology and immunology 398:147–183.
  4. Marshall WF, Blair JE (1999) The cephalosporins. Mayo Clinic Proceedings 74(2):187–195.
  5. Rodriguez R, Mayorga C, Torres MJ, Blanca M (2005) Cephalosporin chemical reactivity and its immunological implications. Current opinion in allergy and clinical immunology 5(4):323–330.
  6. Elks J (1987) Structural formulae and nomenclature of the cephalosporin antibiotics. Drugs 34:240–246.
  7. Zothantluanga, JH, Aswin SK, Rudrapal M, Chetia D (2022) Antimalarial Flavonoid-Glycoside from Acacia pennata with Inhibitory Potential Against PfDHFR-TS: An In-silico Study. Biointerface Research in Applied Chemistry 12(4):48714887.
  8. Patowary L, Sarma M, Zothantluanga JH, Chetia D (2022) Repurposing of FDA Approved Drugs Having Structural Similarity to Artemisinin Against PfDHFR-TS Through Molecular Docking and Molecular Dynamics Simulation Studies. Current Trends in Pharmaceutical Research 8(2):1434.
  9. Zothantluanga JH (2021) Molecular docking simulation studies, toxicity study, bioactivity prediction, and structure-activity relationship reveal rutin as a potential inhibitor of SARS-CoV-2 3CL pro. Journal of Scientific Research 65: 96104.
  10. Paul S, El Bethel Lalthavel Hmar JH, Zothantluanga HKS (2020) Essential oils: a review on their salient biological activities and major delivery strategies. Science Vision 20(2):5471.
  11. Patowary L, Borthakur MS (2022) Computational studies of Bridelia retusa phytochemicals for the identification of promising molecules with inhibitory potential against the spike protein and papain-like protease of SARS-CoV-2. Sciences of Phytochemistry 1(1):2941.
  12. Umar A, Zothantluanga JH, Aswin K, Maulana S, Sulaiman Zubair M, Lalhlenmawia H, Chetia D (2022) Antiviral phytocompounds “ellagic acid” and “(+)-sesamin” of Bridelia retusa identified as potential inhibitors of SARS-CoV-2 3CL pro using extensive molecular docking, molecular dynamics simulation studies, binding free energy calculations, and bioactivity prediction. Structural Chemistry 33(3):121.
  13. Zothantluanga JH, Gogoi N, Shakya A, Chetia D, Lalthanzara H (2021) Computational guided identification of potential leads from Acacia pennata (L.) Willd. as inhibitors for cellular entry and viral replication of SARS-CoV-2. Future Journal of Pharmaceutical Sciences 7(1):118.
  14. Khan J, Asoom LIA, Khan M, Chakrabartty I, Dandoti S, Rudrapal M, Zothantluanga JH (2021) Evolution of RNA viruses from SARS to SARS-CoV-2 and diagnostic techniques for COVID-19: A review. Beni-Suef University Journal of Basic and Applied Sciences 10(1):114.
  15. Pasala PK, Shaik RA, Rudrapal M, Khan J, Alaidarous M A, Khairnar SJ, Walode SG (2022) Cerebroprotective effect of aloe emodin: in silico and in vivo studies. Saudi Journal of Biological Sciences 29(2):9981005.
  16. Umar AK, Zothantluanga JH (2021) Structure-Based Virtual Screening and Molecular Dynamics of Quercetin and Its Natural Derivatives as Potent Oxidative Stress Modulators in ROS-induced Cancer. Indonesian Journal of Pharmaceutics 3(2):6071.

Citation
ACS Style

Sangma, C.D., Chetia, D., Borthakur, M.S., Patowary, L., Tayeng, D. In-silico design and screening of cephalosporin derivatives for their inhibitory potential against Haemophilus influenza. Sciences of Phytochemistry 2022, 1(2), 1-10.

AMA Style

Sangma, CD, Chetia, D, Borthakur, MS, Patowary, L, Tayeng, D. In-silico design and screening of cephalosporin derivatives for their inhibitory potential against Haemophilus influenza. Sciences of Phytochemistry. 2022; 1(2):1-10.

Chicago Style

Chikame Dawa Sangma, Dipak Chetia, Malita Sarma Borthakur, Lima Patowary, Dubom Tayeng. 2022. "In-silico design and screening of cephalosporin derivatives for their inhibitory potential against Haemophilus influenza" Sciences of Phytochemistry 1, no. 2:1-10.

We Computerize Sciences, We Publish Sciences, We Are Scientist

ETFLIN

Become our peer-reviewer

Join us in shaping the future of scholarly research and making a meaningful contribution to academia.

Newsletter

Receive any update from us

Connect with us

Please reach us on our social media below.
ETFLIN Social ETFLIN Social ETFLIN Social ETFLIN Social ETFLIN Social ETFLIN Social
© 2015 - 2024 ETFLIN (Palu, Indonesia)