QSAR, Pharmacophore and Docking Studies on Human Phaspholipase a2 Inhibitors

104
1 GVK Biosciences Private Limited # phase-1, technocrats industrial estates,Balanagar, Hyderabad-500037, India. by IRFAN N [email protected] QSAR, PHARMACOPHORE AND DOCKING STUDIES ON HUMAN PLA2 INHIBITORS

Transcript of QSAR, Pharmacophore and Docking Studies on Human Phaspholipase a2 Inhibitors

Page 1: QSAR, Pharmacophore and Docking Studies on Human Phaspholipase a2 Inhibitors

1

GVK Biosciences Private Limited

# phase-1, technocrats industrial

estates,Balanagar,

Hyderabad-500037,

India.

by

IRFAN N

[email protected]

QSAR, PHARMACOPHORE AND

DOCKING STUDIES ON HUMAN PLA2

INHIBITORS

Page 2: QSAR, Pharmacophore and Docking Studies on Human Phaspholipase a2 Inhibitors

2

Protein Modeling and Rational Drug Designing

by

IRFAN N

In bioCampus Centre of Excellence

GVK Biosciences Private Limited

# phase-1, technocrats industrial estates,

Balanagar,

Hyderabad-500037,

There are no sources in the current document.

There are no sources in the current document.

QSAR, PHARMACOPHORE AND DOCKING STUDIES ON

HUMAN PLA2 INHIBITORS

Page 3: QSAR, Pharmacophore and Docking Studies on Human Phaspholipase a2 Inhibitors

3

S.NO DESCRIPTION PAGE NO

1.

2.

3.

4

5

6

7

Abstract

Legends

Introduction

3.1 Drug discovery

3.2 introduction to protein

3.3 Software

Material and Methods

4.1 Analogue based drug designing

4.1.1 Quantitative structure activity relationships(Qsar)

4.1.2 Pharmacophore

4.2 structure based drug designing

4.2.1 Structure based pharmacophore generation

4.2.2 Docking studies

4.2.2a Ligand Fit

4.2.2b C –Docker

4.2.2c Lib Dock

4.2.2d Ludi

Result and Discussions

5.1 Qsar

5.2 Common feature pharmacophore generation

5.3 3D Qsar pharmacophore generation

5.4 structure based pharmacophore generation

5.5 Ligand fit

5.6 C – Docker

5.7 Lib Dock

5.8 Ludi

Conclusion

Reference

3

4

7

9

19

28

51

60

70

71

72

73

74

78

88

90

97

100

102

112

117

125

127

Page 4: QSAR, Pharmacophore and Docking Studies on Human Phaspholipase a2 Inhibitors

4

1. ABSTRACT

Phospholipase A2 is an enzyme which hydrolyzes the sn-2 position of certain

cellular phospholipids. The liberated lysophospholipid and arachidonic acid are precursors in the

biosynthesis of various biologically active products. As human no pancreatic sPLA2 is present in

high level in the blood of patients in several pathological conditions like septic shock,

pancreatities, trauma, bronchial asthma, gout and other diseases. The potent PLA2 inhibitors have

been suggested to be useful drugs. In this qsar, pharmacophore and docking studies on human

PLA2 inhibitors useful to find new and potent active compounds against several pathological

condition. As per this studies the compound 28v having high dock score and it formed hydrogen

bond interaction with gly29, his27, his47, lys62 amino acids. Novel drug 5-(1-methoxy-4-

methylpentan-3-yl)[1]benzothieno[3,2-b]furan Found through the ludi have the C-dock energy of

-21.094 and it formed hydrogen bond interaction with active site amino acids gly 22, gly 29 and

his 47. Analogue based studies were performed using qsar and pharmacophore generation on

sPLA2 inhibitors. Qsar model having the r² value is 0.968. This study provides the insight in to

binding interaction between receptor and ligands and useful in designing of novel and potent

inhibitors against inflammatory conditions.

Page 5: QSAR, Pharmacophore and Docking Studies on Human Phaspholipase a2 Inhibitors

5

Legends

PLA2 Phospholipase A2

GLY Glycine

HIS Histidine

LYS Lysine

CADD Computer Aided drug design

NSAIDS Nonsteroidal Anti-inflammatory drugs

CNS Central Nervous system

HDL High density lipids

ASP Aspartic acid

PHE Phenylalanine

LEU Leucine

TYR Tyrosine

LF Ligand fit

CHARMM Chemistry at Harvard macromolecular mechanics

QM Quantum mechanics

HYPO Hypothesis

MD Molecular dynamics

SD FILE Structural data file

µM Micro molar

NM Nano molar

% Percent

IC50 Half maximal inhibitory concentration

R² Regression co-efficient

Page 6: QSAR, Pharmacophore and Docking Studies on Human Phaspholipase a2 Inhibitors

6

XVR2 Cross validated regression co-efficient

PRESS Predicted residual error sum squares

LOF Lake of fit

CSD Cambridge structure data base

MLR Multiple linear regression

HBD Hydrogen bond donor

HBA Hydrogen bond acceptor

HY Hydrophobic

PDB Protein data bank

SBDD Structure based drug designing

ABGD Analog based drug designing

RMS Root mean square

HTS High throughput screening

DNA Deoxyribonucleic acid

NMR Nuclear magnetic resonance

QSAR Quantitative structure activity relationship

SAR Structure activity relationship

ADMET Adsorption distribution metabolism excretion toxicity

Page 7: QSAR, Pharmacophore and Docking Studies on Human Phaspholipase a2 Inhibitors

7

Page 8: QSAR, Pharmacophore and Docking Studies on Human Phaspholipase a2 Inhibitors

8

3.1 Drug Discovery:

Drug discovery is the process by which drugs are discovered and/or designed. In the

past most drugs have been discovered either by identifying the active ingredient from traditional

remedies or by serendipitous discovery. A new approach has been to understand how disease and

infection are controlled at the molecular and physiological level and to target specific entities

based on this knowledge. The process of drug discovery involves the identification of candidates,

synthesis, characterization, screening, and assays for therapeutic efficacy. Once a compound has

shown its value in these tests, it will begin the process of drug development prior to clinical trials.

Figure 1. Drug Discovery and development.

Page 9: QSAR, Pharmacophore and Docking Studies on Human Phaspholipase a2 Inhibitors

9

Problem in drug discovery:

Estimates of time and cost of currently bringing a new drug to market vary, but 7–12

years and $ 1.2 billion are often cited. Furthermore, five out of 40,000 compounds tested in

animals reach human testing and only one of five compounds reaching clinical studies is

approved. This represents an enormous investment in terms of time, money and human and other

resources. It includes chemical synthesis, purchase, curation, and biological screening of hundreds

of thousands of compounds to identify hits followed by their optimization to generate leads which

requiring further synthesis.

In addition, predictability of animal studies in terms of both efficacy and toxicity is

frequently suboptimal. Therefore, new approaches are needed to facilitate, expedite and streamline

drug discovery and development, save time, money and resources, and as per pharma mantra “fail

fast, fail early”. It is estimated that computer modeling and simulations account for ~ 10% of

pharmaceutical R&D expenditure and that they will rise to 20% by 2016

Role of computer aided drug designing:

Both computational and experimental techniques have important roles in drug

discovery and development and represent complementary approaches. CADD entails:

Use of computing power to streamline drug discovery and development process

Leverage of chemical and biological information about ligands and/or targets to identify

and optimize new drugs

Design of in silico filters to eliminate compounds with undesirable properties (poor

activity and/or poor Absorption, Distribution, Metabolism, Excretion and Toxicity,

ADMET) and select the most promising candidates

Figure 2.Role of computer aided drug designing

Page 10: QSAR, Pharmacophore and Docking Studies on Human Phaspholipase a2 Inhibitors

10

Benefits of CADD

CADD methods and bioinformatics tools offer significant benefits for drug discovery programs.

1. Cost Savings. The Tufts Report suggests that the cost of drug discovery and development

has reached $800 million for each drug successfully brought to market. Many

biopharmaceutical companies now use computational methods and bioinformatics tools to

reduce this cost burden. Virtual screening, lead optimization and predictions of

bioavailability and bioactivity can help guide experimental research. Only the most

promising experimental lines of inquiry can be followed and experimental dead-ends can

be avoided early based on the results of CADD simulations.

2. Time-to-Market. The predictive power of CADD can help drug research programs choose

only the most promising drug candidates. By focusing drug research on specific lead

candidates and avoiding potential “dead-end” compounds, biopharmaceutical companies

can get drugs to market more quickly.

3. Insight. One of the non-quantifiable benefits of CADD and the use of bioinformatics tools

is the deep insight that researchers acquire about drug-receptor interactions. Molecular

models of drug compounds can reveal intricate, atomic scale binding properties that are

difficult to envision in any other way. When we show researchers new molecular models

of their putative drug compounds, their protein targets and how the two bind together, they

often come up with new ideas on how to modify the drug compounds for improved fit.

This is an intangible benefit that can help design research programs.

CADD and bioinformatics together are a powerful combination in drug research and development.

An important challenge for us going forward is finding skilled, experienced people to manage all

the bioinformatics tools available to us, which will be a topic for a future article.

3.2 Introduction to target protein:

The Inflammatory Response

The inflammatory response is a major part of the non-specific defense system, and is activated by

any damage caused to the tissues of the body, whether caused by a pathogen (such as damage

caused by an infectious microorganism) or even physical injury such as that caused by a scratch or

an insect bite. The affected area becomes red and swollen, or inflamed.

Figure3. Shows the steps of the inflammatory response. In the example shown, a pin pierces

through the skin surface, and infects the tissue with bacteria. The steps shown are:

As soon as the tissue is ruptured, the damaged cells release chemicals such as histamine,

which serve as alarm signals.

The chemicals released activate numerous defense mechanisms in the body. For example,

histamine forces nearby blood vessels to dilate and to allow more diffusion by becoming

leakier. Due to this, blood flow to the affected area increases, and the plasma of the blood

seeps into the interstitial fluid of the damaged tissues. Other chemicals that are released

attract phagocytes and other leukocytes to the affected area. These leukocytes squeeze out

of the blood vessels into the interstitial fluid and tissue spaces. This increase in blood

flow, blood plasma, and white blood cells causes the redness, heat, and swelling that are

normally found in inflammation.

Page 11: QSAR, Pharmacophore and Docking Studies on Human Phaspholipase a2 Inhibitors

11

The leukocytes that have been attracted to the area engulf the bacteria, and any dead body

cells damaged by the pathogens or by the injury. This may result in the death of the

leukocytes, as well, and their remains are also digested. Pus found at the site of infection

consists mainly of white blood cells and blood plasma.

figure3. The Inflammatory Response against pathogens.

The inflammatory response has two major purposes: to disinfect and to clean injured tissues. In

addition to this, the inflammatory system also helps halt the spread of pathogens to tissues not

already infected. Clotting proteins that are present in the blood plasma also leak into the interstitial

fluid when the blood vessels dilate and become leakier. With platelets, thromboplastin,

prothrombin, fibrinogen, and calcium ions, localized clots can be formed, and healing can be

underway, while the pathogens are also restricted to one area, making it easier for them to be

engulfed by phagocytes.

Although the inflammatory response may be localized, as shown, it may also be widespread and in

effect throughout the body. If there are numerous pathogens, or pathogens have traveled through

the bloodstream and come to reside all over the body, the body will react with a widespread

inflammatory response that has other effects in addition to the ones experienced in localized

responses. The number of leukocytes in the blood may increase. The body may also experience

abnormally high body temperatures, or fever, which may be caused by either toxins released by

pathogens, or due to compounds released by specific leukocytes. Although an extremely high

fever is dangerous to the body, a less extreme temperature may aid the body by stimulating

phagocytosis and inhibiting the reproduction and growth of pathogens.

The classical signs inflammation:

Pain (dolor),

Heat (calor),

Redness (rubor),

Swelling (tumor), and

Loss of function (functio laesa).

Responsible mediator for inflammation:

Phospholipase A2(PLA2)

Lipooxygenase(LOX)

Cyclooxigenase(COX),

Page 12: QSAR, Pharmacophore and Docking Studies on Human Phaspholipase a2 Inhibitors

12

Figure4. Inflammatory process

Page 13: QSAR, Pharmacophore and Docking Studies on Human Phaspholipase a2 Inhibitors

13

Current drug against inflammation:

NSAIDS (Non Steroidal Anti-inflammatory drugs)

o Aspirin

o Indomethacin

o Ibuprofen

o Diclofenac

o Piroxicam

Corticosteroids.

o Prednisolon

o Cortisone

o Betamethasone

o Fludrocortisone

WHY NEW DRUGS ARE NEEDED:

Despite decades of research, corticosteroids and NSAIDs remain the main

pharmacological weapons to control inflammation in the clinic. Unfortunately, these drugs have

significant side effects, especially when used chronically. Consequently, there is tremendous

interest in the development of novel, safer, and more effective anti-inflammatory drugs.

Side effects of NSAIDS:

GASTEROINTESTIONAL:

Gastric irritation, erosions, peptic ulceration, gastric bleeding , esophagitis.

RENAL:

Na+ and water retention, chronic renal failure, interstitial nephritis.

CNS

Headache, mental confusion, behavioral disturbances, seizure precipition.

OTHERS:

Asthma exacerbation, nasal polyposis, pruritus, angioedema

Side effects CARTICOSTEROIDES:

Cushing‟s habitués

Hyperglycemia

Muscular weakness

Susceptibility to infection

Delayed healing

Peptic ulceration

Osteoporosis

Glaucoma

Fetal abnormalities

mental confusion

Phospholipase a2 (PLA2): The secretary PLA2 (sPLA2) family, in which 10 isozymes have been identified,

consists of low molecular weight, Ca2+-requiring secretory enzymes that have been implicated in

a number of biological processes, such as modification of eicosanoid generation, inflammation,

and host defense.

Page 14: QSAR, Pharmacophore and Docking Studies on Human Phaspholipase a2 Inhibitors

14

This enzyme has been proposed to hydrolyze phosphatidylcholine (PC) in lipoproteins to liberate

lyso- PC and free fatty acids in the arterial wall, thereby facilitating the accumulation of bioactive

lipids and modified lipoproteins in atherosclerotic foci.

In mice, sPLA2 expression significantly influences HDL particle size and composition

and demonstrate that an induction of sPLA2 is required for the decrease in plasma HDL

cholesterol in response to inflammatory stimuli. Instillation of bacteria into the bronchi was

associated with surfactant degradation and a decrease in large: small ratio of surfactant aggregates

in rats.

sPLA2-IIA can exert beneficial action in the context of infectious diseases since recent

studies have shown that this enzyme exhibits potent bactericidal effects. Induction of the synthesis

of sPLA2-IIA is generally initiated by endotoxin and a limited number of cytokines via paracrine

and/or autocrine processes.

Figure5 Biosynthesis of Arachidonic acid

Page 15: QSAR, Pharmacophore and Docking Studies on Human Phaspholipase a2 Inhibitors

15

Role of phospholipase A2:

Phospholipase A2 (PLA2) catalyzes the hydrolysis of the sn-2 position of membrane

glycerophospholipids to liberate Arachidonic Acid (AA), a precursor of eicosanoids including

prostaglandins and leukotrienes. The same reaction also produces lysophosholipids, which

represent another class of lipid mediators.

Figure6 Role of phospholipase A2

Mechanism

Close-up rendering of PLA2 active site with phosphate enzyme inhibitor. Calcium

ion (pink) coordinates with phosphate (light blue). Phosphate mimics tetrahedral intermediate

blocking substrate access to active site. His-48, Asp-99, and 2 water molecules.

The suggested catalytic mechanism of pancreatic sPLA2 is initiated by a His-48/Asp-99/calcium

complex within the active site. The calcium ion polarizes the sn-2 carbonyl oxygen while also

coordinating with a catalytic water molecule, w5. His-48 improves the nucleophilicity of the

catalytic water via a bridging second water molecule, w6. It has been suggested that two water

molecules are necessary to traverse the distance between the catalytic histidine and the ester. The

basicity of His-48 is thought to be enhanced through hydrogen bonding with Asp-99. An

asparagines substitution for His-48 maintains wild-type activity, as the amide functional group on

asparagines can also function to lower the pKa, or acid dissociation constant, of the bridging water

molecule. The rate limiting state is characterized as the degradation of the tetrahedral intermediate

composed of a calcium coordinated oxyanion. The role of calcium can also be duplicated by other

relatively small cations like cobalt and nickel.

Page 16: QSAR, Pharmacophore and Docking Studies on Human Phaspholipase a2 Inhibitors

16

Figure7 Mechanism of PLA2

PLA2 can also be characterized as having a channel featuring a hydrophobic wall

in which hydrophobic amino acid residues such as Phe, Leu, and Tyr serve to bind the substrate.

Another component of PLA2 is the seven disulfide bridges which are influential in regulation and

stable protein folding.

Why phospholipase a2 inhibitores are needed: Activation of PLA2 leads to the release of fatty acids and lysophospholipid, which

are than converted to mediators of inflammation and allergy, such as prostaglandins, leukotrienes,

and platelet activating factor .therefore, blocked of phospholipase pla2 can result in the suppersion

of three important classes of lipid mediators and offers an attractive therapeutic approach to

inflammation

Inhibition of phosphlipase A2:

sPLA2 inhibitor can be a therapeutically useful drug in the treatment of

1. septic shock

2. acute respiratory

3. distress syndrome,

4. pancreatitis,

5. trauma,

6. bronchial asthma,

7. allergic rhinitis,

Page 17: QSAR, Pharmacophore and Docking Studies on Human Phaspholipase a2 Inhibitors

17

8. rheumatoid arthritis,

9. gout, and

10. Other diseases.

REPORT HIGHLIGHTS 1. The market for anti-inflammatory drugs to treat the diseases covered in this report was

approximately $21.9 billion in 2005 and is projected to increase to $35.5 billion in 2010.

2. The fastest growing disease category for anti-inflammatory treatment is psoriasis, which

saw the first introductions of expensive monoclonal antibody products in the last two

years.

3. The largest market by far in 2005 is that for the treatment of asthma and chronic

obstructive pulmonary disease, which accounted for approximately 36% of the total market

in 2005. The asthma/COPD market will remain the largest in 2010, but will decline to

31.4% of the total of market by the end of the forecast period.

Figure8 Report ID: PHM048A, Published: March 2006, Analyst: Lynn Gray

Pipeline drugs against PLA2:

Page 18: QSAR, Pharmacophore and Docking Studies on Human Phaspholipase a2 Inhibitors

18

Target protein

PDB id : 1DB4

Name : Hydrolase/hydrolase inhibitor

Title : Human s-pla2 in complex with indole 8

Structure : Phospholipase a2. Chain: a. Synonym: hnp-spla2

Source : Homo sapiens.

Biological unit : Dimer

Enzyme class : E.C.3.1.1.4

Reaction : Phosphatidylcholine + H2O = 1-acylglycerophosphocholine + a

Carboxylate

Cofactor : Calcium

Resolution : 2.00Å

R-factor : 0.226

R-free : 0.256

Amino acid length : 124 AA

Authors : N.Y.Chirgadze, R.W.Schevitz, and and J.-P.Wery

Page 19: QSAR, Pharmacophore and Docking Studies on Human Phaspholipase a2 Inhibitors

19

Figure 9 crystal structure of secretory phospholipase a2 (1DB4)

Page 20: QSAR, Pharmacophore and Docking Studies on Human Phaspholipase a2 Inhibitors

20

3.3 software

Presnt experimental studies carried out using the tools

Accelrys software

Discovery studio

Discovery studio is a complete modeling and simulations environment for life science

researchers. Discovery Studio is a single, easy-to-use, graphical interface for powerful drug design

and protein modeling research. Discovery Studio 2.1 combines established gold-standard

applications such as Catalyst, Modeler, and CHARMm that have years of proven results and

utilizes cutting-edge science to address the drug discovery challenges of today. Discovery Studio

2.1 is built on the Pipeline Pilot open operating platform to seamlessly integrate protein modeling,

pharmacophore analysis, virtual screening, and third-party applications. It offers

Figure 10: feature of discovery studio

o Interactive, visual and integrated software.

o Consistent, contemporary user interface for added ease-of-use

Page 21: QSAR, Pharmacophore and Docking Studies on Human Phaspholipase a2 Inhibitors

21

o Tools for visualization, protein modeling, simulation, docking, pharmacophore

analysis, qsar and library design

o Access computational servers and tools, share data, monitor jobs, and prepare and

communicate their project progress.

Page 22: QSAR, Pharmacophore and Docking Studies on Human Phaspholipase a2 Inhibitors

22

4. Materials and methods

In the last few years the role of computational methods in both pharmaceutical

and academic research has developed dramatically. The emphasis being placed on high throughput

methods in the pharmaceutical industry, which has increased the number of compounds in the

discovery pipeline. Characterizing the position and orientation of small molecules bound to a

protein surface can be an important step in drug design. Computational methods developed rapidly

as groups seek high throughput, low cost approaches in accelerating the drug discovery process.

Such approaches will be necessary as scientists attempt to characterize the large number of drugs

currently being generated. Structural information of biological macro molecules and their

importance with ligand is increasingly being used in modern medicinal chemistry. There is a

pressing used for novel computational methods that can evaluate the structural information about

ligand receptor complexes in a more quantitative way , both to improve existing leads and to

design de novo compounds with accurately predicted binding affinities . The following

experimental methods categorically divided into two parts.

4.1 Analogue based drug designing

4.1.1 Quantitative structure activity relationships (Qsar)

4.1.2 Common feature pharmacophore (hip hop)

4.1.3 3D Qsar pharmacophore (hypogen)

4.2 structure based drug designing

4.2.1 Structure based pharmacophore generation

4.2.2 Docking studies

4.2.2a Ligand Fit

4.2.2b C –Docker

4.2.2c Lib Dock

4.2.2d Ludi

Page 23: QSAR, Pharmacophore and Docking Studies on Human Phaspholipase a2 Inhibitors

23

Preparation of molecular system:

Macro molecule (protein 1db4) preparation:

Load the protein and Apply the force field

For this qsar, pharmacophore and docking

studies, the protein 1DB4 load from RCSB protein data bank

(www.rcsb.org/pdb/) and apply the force field .Force field refers to

the functional form parameter sets which are used to find out potential energy of a system. It

includes parameter which is obtained through experimental works and quantum mechanics

calculations. All molecules in a molecule in a mechanical system are made up of a number of

components. Covalently bonded atoms takes into consideration several parameters such as bond

length , bond angle , dihedral angles etc., similarly there exists non bonded interactions such as

vanderwaals interactions , electrostatic interactions . Thus the total potential energy of the system

is calculated as follows

E1= [E bond + E angle + E torsion + E vandervaals + E electronic]

This summation when given is an explicit form, represents force field, evaluating the potential of a

system.

minimization :

The minimizer uses algorithm to identify the geometrics of the molecule

corresponding to the minimum points on the potential surface energy. The minimum reduced the

unwanted forces which are present in the molecule and lower the energy level of the molecule.

There are many algorithms available in the minimization process. Some of the minimization

methods used in the smart minimizer is steepest decent method, conjugate gradient method,

Newton raphson method and quasi Newton method. From the DS protocols select the

minimization and run .the following figure shows the minimized protein with fixed constraint

.than sve the minimized protein for further studies.

Page 24: QSAR, Pharmacophore and Docking Studies on Human Phaspholipase a2 Inhibitors

24

Figure 11 Minimized protein with fixed constraint

Page 25: QSAR, Pharmacophore and Docking Studies on Human Phaspholipase a2 Inhibitors

25

Preparation of bio active molecules:

The 111 bio active compounds are collected from the journals

with the activity range 0.005 to >50 µM.

Journal of medicinal chemistry 1996, vol 39, page no 3636-33658 with the title potent

inhibitors of secretory phospholipase A2: synthesis and inhibitory activities of indolizine

and indene derivatives.

Journal of medicinal chemistry 2005, vol 48, page no 893-896 with the title carbocyclic

[g]indole inhibitors of human nonpancreatic sPLA2

Journal of medicinal chemistry 2008, vol 51, 4708-4714 wit the title highly specific and

broadly potent inhibitors of mammalian secreted phospholipase A2.

1 One molecule was drawn with basic scaffold and the other molecules were constructed

with one drawn earlier as the reference model.

2. Drawn compounds are typed with charmm force field.

3. The typed molecule are subjected to the energy minimization using smart minimizer.

Minimizes a series of ligand poses using CHARMm

4. Minimized molecule is saved with .sd and .mol2 extension for further study.

Following table shows the 2d structure of the molecule and activity

Page 26: QSAR, Pharmacophore and Docking Studies on Human Phaspholipase a2 Inhibitors

26

4.1 Analogue Based Drug Design

The unknown 3D structural target knowledge is applied to rationally design a drug; this is

referred to as Analogue Based Drug Design. This refers to the application of the knowledge of the

ligand structure ant their activity when the 3D structure of the target is having a very less

information or is completely not known .It is required to design the binding site based on the

known structure of the ligands.

4.1.1 Qsar:

The fundamental quantitative structure activity relationship studies reveals

that the structures can be easily be compared, overlayed and displayed. The Quantitative structure

of activity relationship is obtained by providing more parameters to optimize a series of bioactive

molecules. The quantitative structure activity relationship based on physio chemical properties

describes a drugs structural, electronic and physiochemical characteristics. Data sets are produced

using all available descriptors.

Apply knowledge of the three-dimensional (3D) structure of the target

(receptor/enzyme/DNA) to rationally design drug molecules to bind to the target for the following

reasons are:-

1. Understand atomic details of drug binding strength and specificity (drug-receptor interactions).

2. Develop novel drugs (unique chemical structures) for a selected target via de novo drug design

or database searching techniques.

3. Optimize the therapeutic index of an already available drug or lead compound concerning

structural requirements for activity from a minimum number of compounds are tested.

figure12: concept of QSAR

Page 27: QSAR, Pharmacophore and Docking Studies on Human Phaspholipase a2 Inhibitors

27

A QSAR equation numerically defines the chemical properties, Biological activity

form physiochemical properties. Biological activity is defined as pharmacological response

usually expressed in millions such as the effective dose in 50% of the subjects (ED50). The lethal

dose is 50% of the subjects (LD50) or the minimum inhibitory concentration IC50. It is common to

express the biological activity as a reciprocal QSAR equation is similar to the equation for a

straight line:-

Y = mx + c

Log biological activity = a (physiochemical property) + c

A = regression coefficient of slope of the straight line.

C = intercept on y-axis (when the physiochemical property equals zero)

Biological activity expressed as a reciprocal to produce a positives lope and also due

to the inverse relationship between physiochemical chemical property and biological potency.

There is a positive relationship between the reciprocal of the biological activity(I/BA) and

physiochemical property, because (I/BA) increases as the studies are based on the descriptors and

biological activity relationship the biological activity data must be minimal .and the choice of the

descriptors of the descriptors must be accurate and appropriate .

OBJECTIVE OF QSAR

1. Drug transport/ mechanism

2. Prediction of activity.

3. Classification of molecules as highly active, moderately active and inactive.

4. Optimization of activity by steric, electrostatic and hydrophobicity

5. Refinement of synthetic targets.

6. Reduction and replacement of animals for the action of drugs

BASIC REQUIREMENT IN QSAR STUDIES

1. All analogue belong congeneric series

2. All analogues exert same mechanisms of actions.

3. All analogue bind in a comparable manner.

4. Effect of isosteric replacement can be predicted

5. Binding affinity correlated to interaction energies

6. Biological activities correlated to binding activity

QSAR STUDIES INVOLVE THE FOLLOWING STEPS

A. CSD data base.

C. Choice of descriptors.

D. Statistical methods to evaluate to evolve QSAR equation.

E. Validation.

Page 28: QSAR, Pharmacophore and Docking Studies on Human Phaspholipase a2 Inhibitors

28

A. CSD DATABASE

Experimental information about the structures of molecules can often be extremely useful

for forming theories of conformational analysis and hoping to predict the structures of molecules

for which no experimental information is available. The most important technique currently

available for determining the three dimensional structure of molecules is x-ray crystallography

community has distributed in electronic form two practically important databases for molecular

modeler are the Cambridge structural database CSD which contains crystal structures of organic

and organ metallic molecules and the protein data bank (PDB) which contain structures of proteins

and some DNA fragments.

A data base of little use without software tools to search extract and manipulate the data. A

simple use of a database is for extracting information about a particular molecule or group of

molecules .the data may also be identified by creating a two dimensional representation of

molecule and using a substructure search program to search the database. Crystallographic

database have also been used to develop an understanding of the factors that influence the

conformations of the molecules, and of the ways in which molecules interact with each other. For

example, the CSD has comprehensively analyzed to characterize how the lengths of chemical

bonded depend upon the atomic numbers, hybridization and the environment of the atoms

involved. Analyzing of intermolecular hydrogen bonding have revealed distinct distance and

angular preferences a major use of the CSD is substructure searching for molecules which contain

a particular fragment, in order to investigate the conformation that the fragment adopts.

A crystallographic database can only provide information about the crystal state of

matter and that the possible influence of crystal packing forces should always be taken into

account. This is less of concern for protein than for small molecules as protein crystals contain a

large amount of water and indeed NMR studies are established that protein have approximately,

the same structure in solution as in the crystal.

A second, more stable subtle, bias is that crystallographic databases only contain

molecules that can be crystallized and indeed only those molecules whose X-ray structures were

considered enough to be published. The structures in a crystallographic database may therefore not

be a wholly representative set.

C. MOLECULAR DESCRIPTORS

The study of steric requirements for interaction between ligands and corresponding

biological acceptor sites is often of decisive importance in understanding the role played by the

structural features in promoting activity in its most general form drug receptor theory requires that

a ligand exerts its biological action as a consequence of binding or otherwise interacting with a

specific biological acceptor site such as membrane protein , an enzyme etc., which may be

generally termed the receptor the concept is the basis for modern drug receptor theory involves

the old principle that a ligand fits its receptor much as a key fits a lock. This concept, although

some what arbitrary since a high degree of flexibility is present in biomacromolecules, structure,

governs the principle of molecular recognition and molecular discrimination. Although

stereochemistry often plays a major role in drug bioactive, care must be taken when considering

structure activity relationship to explore whether other differences in physiochemical properties

exists before one makes significant correlations with the steric properties of the structure under

study.

In early studies organic chemists defined a number of steric parameters in order to

explain steric effects of substituents on the reaction centers of organic molecules. The same type

of steric effects observe in studies of variation of physical properties and the chemical reactivity

with structure may be assumed to be involved in biological activity studies which at least as a first

Page 29: QSAR, Pharmacophore and Docking Studies on Human Phaspholipase a2 Inhibitors

29

approximation may be treated in similar fashion in the past 35 years owing to the development of

drug design and Hansch Approach many other parameters and methods have been developed

which have the permit of trying to avoid a simple empirical correlation with given ligand

properties and also trying to propose the possible geometric features of the receptor. Steric descriptors are classified into following groups:

1. Topological indices based on characterization of the chemical structures of the graph theory.

2. Geometric descriptors resulting from the view of organic molecules as three dimensional

objects from which standard dimensions can be calculated.

3. Chemical descriptors derived from steric influence upon a standard reaction.

4. Physical descriptors derived when an organic molecule is considered as three dimensional

object with size determined physical properties and different descriptors which result when an

organic molecule is considered as a three dimensional object from reference structure.

I. FRAGMENT CONSTANT DESCRIPTORS

Fragment constant descriptors are constants that relate the effect of substituents on a reaction

center one type of process to other. The basic idea is that similar changes in structure are likely to

produce similar changes in reactivity, ionization or binding. There are different constants

corresponding to different effects. These are typically used to parameterize the Hammet equation

some series of analogs.

Log kx= pσ +log kh

Where Kx and kh are reaction rate constants for the substituents x and h , respectively ;0 is an

electronic constant by an ionization constant and p is fit to set etc at different properties

(electronic , steric )etc at different R group positions are used . In this way measurements of

ionization constants can be used to predict rate constants once a sealing factor (p) is determined

effects for the rate of constant.The default database currently contains the following types of

constants. These come from table VI –I of hansch expect for the sterimol constant which is

calculated.

Sm, Sp

Electronic effect sigma Meta and sigma para respectively. Positive values correspond in

electronic withdrawal, negative ones with electronic release. Sigma is generally not appropriate for

ortho substituents because of steric interaction with reaction center.

F and R

Decomposition of sigma Para constant into an inductive polar part F and a resonance part R for

the case when the substituent is conjugated with the reaction center producing through resonance

effects.

Pi

Hydrophobic character Pi for substituent x is given by the difference of its log P from the

log P for hydrogen.

HA hydrogen bond acceptor

HB hydrogen bond acceptor

MR molar refractivity is given by p

MR= (n2-1/n2+1)*(MW/d)

Where n is the refractive index .MW is the molecular weight and d is the compound density

sterimol L.

Sterimol-L

Steric length parameter, measured long the substitution point bond axis.

Sterimol –B 1 through B4

Page 30: QSAR, Pharmacophore and Docking Studies on Human Phaspholipase a2 Inhibitors

30

Steric distance s perpendicular to bond axis, these define a bounding box for the substituent and

are numbered in ascending size axis.

Sterimol –BS

The overall maximum steric distance is perpendicular to the bond axis.

II. CONFORMATIONAL DESCRIPTORS

Energy descriptor energy is the energy of the currently selected conformation in the study table.

Low energy Low energy is the energy of the most stable conformation in the set of conformations

belonging to each molecular model.

E penalty E penalty is the difference between energy and low energy

III. ELECTRONIC DESCRIPTORS

The following table lists the electronic descriptors available in QSAR are as follows:

Table 3: Electronic descriptors

SYMBOL DESCRIPTION

Charge sum of partial charges

F charge sum of formal charges

Apol sum of atomic polarizabilities

Dipole dipole moment

HOMO highest occupied molecular orbital

LUMO lowest occupied molecular orbital

SR super delocalizability

IV. MOLECULAR SHAPE ANALYSIS (MSA) DESCRIPTORS

The following table lists the MSA descriptors available in QSAR are as follows:

Table4: Molecular shape analysis descriptors

SYMBOL

DESCRIPTORS

DIFF difference volume

Fo common overlap volume (ratio)

NCOSV non common overlap steric volume

Shape RMS RMS to shape reference

COSV common overlap steric volume

SR vol volume of shape reference compound

Page 31: QSAR, Pharmacophore and Docking Studies on Human Phaspholipase a2 Inhibitors

31

V) STRUCTURAL DESCRIPTORS

The following table lists the structural descriptors available in QSAR are as follows:

Table 5: Structural descriptors

SYMBOL DESCRIPTORS

Mw molecular weight

Rot bonds number of rotatable bonds

H bond acceptors number of hydrogen acceptors

H bond donors number of hydrogen bond donor

VI. THERMODYNAMIC DESCRIPTORS

The following table lists the thermodynamic descriptors available in QSAR follows:

Table 6: Thermodynamics descriptors

SYMBOL DESCRIPTORS

AlogP log of partition coefficient

FH2o desolvation free energy of water

Foct desolvation free energy of octanol

HF heat of formation

Molref molar refractivity

VII. RECEPTOR DESCRIPTORS

Quantitative values such as the interaction energy calculated in receptor for a generated

receptor model are available to use in QSAR. By using receptor data to develop a QSAR model,

you can evaluate the goodness of fit between a candidate‟s structure and postulated pseudo

receptor. When you have generated a receptor model ad have aligned the models you want to

study, you can proceed to build a QSAR using data from the receptor structure iterations.

The following table lists the receptor descriptors available to QSAR are as follows:

Table 7: Receptor Descriptors

SYMBOL DESCRIPTION

Intra energy molecular internal energy inside receptor

Inter Elec energy Non bonded electrostatic energy between molecule and receptor

InterVDW energy Non bonded vanderwaals energy between molecule and receptor

Inter energy total nonbonded energy between molecules and receptor

Min intra energy molecular internal energy minimized without receptor

Stain energy molecular strain energy within receptor

VIII. MOLECULAR FIELD ANALYSIS (MFA) DESCRIPTORS:

Molecular field analysis (MFA) evaluates the energy between a probe and molecular model at a

series of points defined by a rectangular or spherical grid. This energy may be added to the study

table to form new columns headed according to the probe type. The new columns may be used as

independent X variables in the generation of QSAR.

Page 32: QSAR, Pharmacophore and Docking Studies on Human Phaspholipase a2 Inhibitors

32

Molecular field analysis (MFA) MFA evaluates the interaction energy between a probe and a molecular model at a series of

points defined by a rectangular or spherical grid. This method quantifies the interaction energy

between a probe molecule and a set of aligned target molecules in QSAR. Six descriptors are

available in this family.

1. H+

probe: This selects proton “as a probe‟, having +1 charge and zero vanderwaals radius.

It has electrostatic interactions and non bonded interaction are not considered

2. CH3 probe: This probe with a vanderwaals radius of united CH3 group but with a zero

charge. The energy of interaction of this probe with a study molecule will include only non

bonded interactions.

3. Donor / acceptor probe: It is two atom probes consisting of oxygen bounded to hydrogen.

The vanderwaals radii of eth atoms are exactly how they are defined in the particular force

field loaded. The probe is neutral. Depending on the orientation of this probe. It is capable

of bleaching as a hydrogen bond donor or an acceptor.

4. CH3 probe: It is single atom probe with a vanderwaals radius of a united CH3 of -1. The

energy of interaction of this probe includes both non-bonded of interaction of this probe

includes both non bounded and electrostatic interactions.

5. Generic probe: There is a generic single atom probe with a user specified Vander radius

and charge.

6. Other probes: Any multi atom model may be employed as a probe specifying the Msi file

format.

D. STATISTICAL METHOD TO EVALUATE QSAR EQUATION

QSAR analysis uses statistical methods for studying the correlation of biological activity to

structural and physio chemical properties of candidate molecules. Here are different statistical

techniques used to fit the molecule under multivariate statistics, which include the following:-

1. PCA (PRINCIPAL COMPONENT ANALYSIS): It aims at representing large amount of

multidimensional data by transforming them into a more intuitive low dimensional representation.

This method does not create a model, but searches for relationship among the independent

variables. It then creates new variables (the principal components) which represent most of the

information contained in the independent variables.

2. CLUSTER ANALYSIS: The goal of cluster analysis is to partition (typically to representing

set of models in a molecular descriptor property space) into classes or categories consisting of

elements of comparable similarity. The algorithm assumes that models are represented by points

in multidimensional property space with Euclidian distance between points representing model

dissimilarity. The below mentioned are the types in this category

1. Jarivs – Patrick clustering

2. Variable-Length Jarnis Patrick clustering

3. Relocation Clustering

4. Hierarchical Clustering Analysis (HCA)

3. SIMPLE LINEAR REGRESSION: It performs a standard linear regression calculation to

generate a set of QSAR equations that includes one equation for each independent variable. It is

good for exploring simple relations between structure and activity

4. LINEAR (MULTIPLE LINEAR REGRESSIONS): This method calculates QSAR equation

by performing standard multi variable regression calculations using multiple variables in a single

equation. In this method variables are independent correlated).

Page 33: QSAR, Pharmacophore and Docking Studies on Human Phaspholipase a2 Inhibitors

33

5. STEPWISE MULTIPLE LINEAR REGRESSION: It calculates QSAR equation s by adding

one variable data time and testing each addition for significance and such variables are sued in

QSAR equation. It is useful when the number of variables is large and when the key descriptors

are not known. If the number of variables exceeds number of structures this method should not be

used.

6. PLS (PARTIAL LEAST SQUARES): This method carries out regression using latent

variables. From the independent and dependent data that are along their axes of greatest variation

and are most highly correlated. It can be used with more than one dependent variable. It is

typically applied when the independent variables are correlated or the number of independent

variables exceeds the number of observations (rows).

7. GFA (GENETIC FUNCTION APPROXIMATION): It is an alternative to the standard

regression analysis for constructing QSAR equations. The method provides multiple models that

are created by evolving random initial models using a genetic algorithm GFA can build linear and

higher- order polynomials, splines and other non–linear equations. In this method, models are

collected that have a randomly chosen proper subset of the independent variables and then the

collected models are evolved A generation is eth set of models resulting from performing multiple

linear regression on each model, a selection of the best one becomes the next generation .crossover

operations are performed on this equation.

8. G/PLS: (GENETIC PARTIAL LEAST SQUARES): It is a method derived from GFA and

PLS that are valuable analytical tools for datasets that have more descriptors than samples. The

following three statistical methods are useful in combi chem. and analog builder.

10. FA (FACTOR ANALYSIS): It addresses one of the main problems found in PCA that is not

simple to relate the principal component to molecular properties. All the common factors have a

close relation ship to real molecular properties.

11. RP (RECURSIVE PARTITION): It identifies the internal representation of classes used by

classification structure activity relations hip (CSAR) for deriving recursive portioning models.

E. VALIDATION METHODS

Once a regression equation is obtained it is important to determine its reliability and its

significance. Internal validation uses the data set for which the model is derived and checks for

internal consistency. The procedure derives a new model and is used to predict the activities of the

molecules that were not included in the new model set. This is repeated until all compounds have

been deleted and predicted once. Internal validation is less rigorous than external validation.

External validation evaluates how well the equation generalization. The original data are divided

into two groups, the training set and the test set. The training set is used to derive a model, and the

model is used to predict the activities of the test set numbers. The following procedures are used to

check that the size of the model is appropriate for the quantity of data availability as well as

provides some estimate of how well the model can predict activity for new models are as

follows:-

1. CROSS VALIDATION: This process repeats the regression may times on subsets of the data.

Usually each molecule is left out intern and r2 is computed using the predicted values of the

missing molecules (r2)

2. RANDOMIZATION TEST: Even with large number of observations and a small number of

terms, an equation can still have a very poor predictive power. This can come about it the

observation are not sufficiently independent of each other.

Page 34: QSAR, Pharmacophore and Docking Studies on Human Phaspholipase a2 Inhibitors

34

F. INTERPRETING QSAR EQUATION

QSAR is used for predicting the activities of as yet untested and possibly not yet synthesized)

molecules. The predictive ability of a QSAR is generally more accurate for interpolative (for

compound that have parameters with in the range of those considered in the data set) than for the

extra polative predictions (compounds that are outside the range)

A QSAR equation provides insights into the mechanism of the process being studies.

1. SQUARE OF CORRELATION COEFFICIENT (R2): If x (independent) and y (dependent)

variables are highly correlated, there is considerable information in x and y that is redundant. The

degree of correlation is measured by the correlation coefficient (r2)

2. CROSS VALIDATED R2 (TERMED AS Q2 OR XVR2): r

2can be computed using cross

validation methods (XVr2) o r boost strap methods (BSr

2). It is also the fraction of the variance

explained by the model. Cross validated r2 is always some what lower and often much lower than

the r2.

3. PRESS (PREDICTIVE ERROR SUM OF SQUARES): The sum of overall compares of the

squared differences between the actual and the predicted values for independent variables [1/i y]2.

The intensity of the cross validated process is controlled by selecting the number of groups or

number of times the cross validation step is to be carried out while predicting all rows (at each

stage of model development).

PROCEDURE:

Figure13: Flowchart of qsar procedure

Page 35: QSAR, Pharmacophore and Docking Studies on Human Phaspholipase a2 Inhibitors

35

Calculate molecular properties:

The Calculate Molecular Properties protocol will calculate many properties or perform basic

statistical and correlation analysis of the numeric properties as requested.

To set up a Calculate Molecular Properties protocol:

1. Load the QSAR and apply the force field on molecules and Calculate Molecular

Properties protocol from the Protocols Explorer. The parameters display in the Parameters

Explorer.

2. On the Parameters Explorer, click in the cell for the Input Ligands parameter and click

the button to specify the ligand source on the Specify Ligands dialog. On the dialog, select

all ligands from a Table Browser, a 3D Window, or a file.

3. Select the properties to calculate by clicking the button in a cell for the Molecular

Properties, Semi empirical QM descriptors, or Density Functional QM descriptors, and

follow the instructions in the popup dialog window.

The Create genetic function approximation can build a Create genetic function approximation

model for a dependent property using the selected molecular descriptors.

To set up a Create genetic function approximation Model protocol

1. Load the QSAR | Create genetic function approximation Model protocol from the

Protocols Explorer. The parameters display in the Parameters Explorer.

2. On the Parameters Explorer, click in the cell for the Input Ligands parameter and click

the button to specify the ligand source on the Specify Ligands dialog. On the dialog, select

all ligands from a Table Browser, a 3D Window, or a file.

3. Set the desired model name using the Model Name parameter. Once created, this model

will appear under the other category of the Molecular Properties parameter in the Calculate

Molecular Properties protocol and can be used to compute the property for future ligands.

4. Set the initial equation length and remaining parameters as desired. Parameters

presented in red are required.

4.1.2. PHARMACOPHORE:

“A pharmacophoreis the ensemble of stericand electronic features that is necessary to

ensure the optimalsupramolecular interactions with a specific biological target and to trigger (or

block) its biological response.” Perceiving a pharmacophore is the most important first step

towards understanding the interaction between a receptor and ligand. In the early 1900‟s Paul

Ehrlich offered the first definition for a pharmacophore. A pharmacophore was first defined by

Paul Ehrlich in 1909 as "a molecular framework that carries (phoros) the essential features

responsible for a drug‟s (=pharmacon's) biological activity" . Catalyst provides the tools for

selecting potential ligand compounds prior to synthesis. The aim of this software is to reduce the

time and cost of screening, synthesis and biological testing. It accelerates the drug discovery

process by identifying lead candidates faster.

Pharmacophore or hypothesis describes the generalized molecular features involved in the

binding of ligand to activate site of proteins molecular features including 1D which represents the

physical and biological properties, 2D represents the sub structures and 3D represents the chemical

features such as acceptors, donors, positive, negative, ionizable, hydrophobic (aromatic &

aliphatic) and ring compounds features. In catalyst each hypothesis can be defined in four parts.

The first one is chemical features, second is location and orientation in 3D dimensional space,

Page 36: QSAR, Pharmacophore and Docking Studies on Human Phaspholipase a2 Inhibitors

36

tolerance and weight. Weight represents the relative importance of each chemical function in

conferring activity

A pharmacophore model or hypothesis consists of a three-dimensional configuration of

chemical functions surrounded by tolerance spheres. A tolerance sphere defines that area in space

that should be occupied by a specific type of chemical functionality. Pharmacophore models are

routinely used in lead identification and optimization in the areas of library focusing, evaluation

and prioritization of virtual high throughput screening (VHTS) results, de novo design, and

scaffold hopping. Pharmacophore models can be constructed using analog-based (using known

active ligands) or receptor-based techniques (using receptor active site information). In the

absence of crystallographic structure data of a protein for which the active site for receptor binding

is clearly identified, a chemist must rely on the structure activity data for a given set of ligands. If

these ligands are known to bind to the same receptor, then one can attempt to define the

commonality between them. Accelrys Catalyst program can generate two types of automated

pharmacophore models, Hypo Gen and Hip Hop, depending on whether or not activity data is

used. In the presence of protein crystal structure data, active site pharmacophore models can be

used as a pre-filter for docking large libraries. Generation of a pharmacophore model using the

active site residue information is key to the success of any pharmacophore-based docking

algorithm. In the absence of X-Ray bound ligand information; it is a challenge to select a single

pharmacophore model that represents the binding characteristics. A methodology is proposed in

this case study that can be used to analyze and visualize multiple pharmacophore models. This

methodology can be applied to different types of Catalyst pharmacophore models (qualitative,

quantitative, receptor-based, etc.) as it only considers feature types and coordinates.

This methodology can be applied successfully to the following applications:

1. VHTS screening

2. multiple binding mode identification

3. classification of proteins based on binding characteristics

4. visualization of pharmacophore model space

To build a better pharmacophore, the following steps were employed:

1. Building a set of molecules

2. Conformer generation

3. Hypothesis Generation

4. Database Search

5. Compare/Fit to estimate Activity

The Feature Dictionary list contains the generalized chemical functions in Catalyst.

Definitions of these functions are:

1. HB ACCEPTOR (vector): Matches these types of atoms or groups of atoms with surface

accessibility:

sp or sp2 nitrogen‟s that have a lone pair and charge less than or equal to zero

sp3 oxygen‟s or sulfurs that have a lone pair and charge less than or equal to zero

non-basic amines that have a lone pair

Does not match: basic, primary, secondary, and tertiary amines that are protonated at physiological

pH. There is no exclusion of electron-deficient pyridines and imidazoles.

2. HB ACCEPTOR lipid (vector): Matches these types of atoms or groups of atoms: nitrogen‟s,

oxygens, or sulfurs (except hypervalent) that have a lone pair and charge less than or equal to zero.

This function is the same as HB ACCEPTOR except that it includes basic nitrogen. There is no

exclusion of electron-deficient pyridines and imidazoles.

Page 37: QSAR, Pharmacophore and Docking Studies on Human Phaspholipase a2 Inhibitors

37

3. HB DONOR (vector): Matches these types of atoms or groups of atoms:

Non-acidic hydroxyls

Thiols

Acetylenic hydrogens

NHs (except tetrazoles and trifluoromethyl sulfonamide hydrogens)

Does not match: electron-rich pyridines and imidazoles that would be protonated or nitrogen‟s that

would be protonated due to their high basicity

4. HYDROPHOBIC (point): Matches these types of groups of atoms:

A contiguous set of atoms that is not adjacent to any concentrations of charge (charged atoms or

electronegative atoms) in a conformer such that the atoms have surface accessibility such as

phenyl, cycloalkyl, isopropyl, and methyl.

5. HYDROPHOBIC ALIPHATIC (point): Matches these types of groups of atoms:

A contiguous set of atoms that are not adjacent to any concentrations of charge (charged atoms or

electronegative atoms) in a conformer such that the atoms have surface accessibility is cycloalkyl,

isopropyl, and methyl

6. HYDROPHOBIC AROMATIC (point): Matches these types of groups of atoms:

A contiguous set of atoms that is not adjacent to any concentrations of charge (charged atoms or

electronegative atoms) in a conformer such that the atoms have surface accessibility such as

phenyl and indole.

7. NEG CHARGE (atom): Matches negative charges not adjacent to a positive charge.

8. NEG IONIZABLE (point): Matches atoms or groups of atoms that are likely to be

deprotonated at physiological pH, such as:

Trifluoromethyl sulfonamide hydrogens

Sulfonic acids (centroid of the three oxygens)

Phosphoric acids (centroid of the three oxygen‟s)

Sulfinic, carboxylic, or phosphinic acids (centroid of the two oxygen‟s)

Tetrazoles

Negative charges not adjacent to a positive charge

9. POS CHARGE (atom): Matches positive charges not adjacent to a negative charge.

10. POS IONIZABLE (point): Matches atoms or groups of atoms that are likely to be protonated

at physiological pH, such as:

Basic amines

Basic secondary amidines (iminyl nitrogen)

Basic primary amidines, except guanidine‟s (centroid of the two nitrogen‟s)

Basic guanidine‟s (centroid of the three nitrogen‟s)

Positive charges adjacent to a negative charge do not match weakly basic aromatic nitrogen‟s such

as pyridine and imidazole.

11. RING AROMATIC (vector and plane): Matches 5- and 6-membered aromatic rings. The

feature defines 2 points, the ring centroid and a projected point normal to the ring plane. The

projected point can map both above and below the ring.

STEPS TO BE FOLLOWED IN DS

1. Construct or import the molecules.

2. Perform conformational search

3. Examine the each conformer for the presence of chemical features.

4. Determine the set of features that correlate with activity

Page 38: QSAR, Pharmacophore and Docking Studies on Human Phaspholipase a2 Inhibitors

38

STEPS AND APPLICATION OF PARAMETERS WHICH ARE USED IN HYPOTHESIS

GENERATION

Import the molecules in view compound work bench and cleaning the constructed

molecules.

Apply catalyst force field , then do the 3D minimize

Conformation search: the aim of the conformation search is to obtain the diversified

conformations .Conformations generation methods are classified into two types. One is best

method and the other is fast method. Both the methods emphasize broad coverage to cover the

conformational space. Fast conformer generation is used to cover the conformational space of

molecules. It uses systematic or random search depending on the size of the molecules. Systematic

search is useful for small molecules and random search is used for macromolecules. In the case of

macro molecules the conformers are minimized by poling algorithm.

CONFORMATIONAL ANALYSIS STOPS WHEN ONE OF TEST THREE CONDITIONS

IS MET

After maximum number of conformers have generated.

Energy of the newly generated conformer is too high to the predefined energy rest hold.

If there is no possible new conformer generation after certain number of trials.

PHARMACOPHORE HYPOTHESIS

Catalysts confirm hip hop and hypogen are application that provides tools to generate

pharmacophore hypothesis. The hypothesis are created by generating conformation for a set of

study molecules, then using the conformation to find and align chemically important functional

groups common to the molecules in the study set.. Chemically important functional groups

common to the molecules in the study set. Each hypothesis can also incorporate data on the

biological activities of the study molecules.

STEPS INVOLVED GENERATING A PHARMACOPHORE HYPOTHESIS

I. GENERATE CONFORMATIONS

The interface to confirm is used to generate conformations for a single molecule or a set of

molecules. The number of conformation needed to produce a good representation of a compound

conformational space depends on the molecules. Both conformations generating algorithms

available in confirm (best and fast) are adjusted to produce a diverse set of conformations ,

avoiding repetition groups of conformations all representing local minima.

The conformations all representing local minima.

The conformations generated by confirm can be used as input into hip-hop and hypo to

align common molecular features and generate a hypothesis.

Align common features to generate a hypothesis.

The following procedure involves

Aligning common molecular features.

Setting preferences using control panel

Incorporating activity data into a hypothesis

Using aligned structures to generate receptor models.

Hip hop and hypo use conformations generated in confirm to align chemically important

functional groups common in the molecules in the study set. A pharmacophore hypothesis can

then be generated from these aligned structures. Incorporated biological activity data into a

hypothesis

Page 39: QSAR, Pharmacophore and Docking Studies on Human Phaspholipase a2 Inhibitors

39

The hip hop is also used to incorporate biological activity data into the hypothesis

generating process. Each hypothesis is tested by regression techniques to compare estimated

activity with actual activity data. The software uses the data from these tests to select the

hypothesis that do the best job predicting activity for the set of study molecules. This capability is

provided by catalyst / hypo.

4.2.2a HIP HOP THEORY

Pharamcophore based on multiple common features alignment generate receptor models

using hip hop. The objective is to identify and enumerate all possible pharamacophore

configurations that are common to the training set. The aligned structures the model receptor menu

card is included in the hypothesis models card deck so that you can use structures that have been

aligned in hip hop to generate a receptor surface model. Since structures used in hip hop are

aligned by common chemical features, the receptor surface model that is generated for them can

be significantly different from a receptor surface model generated from template aligned

structures.

The ideal hip hop training set area s follows:-

2-30 compounds ideally 6 molecules

Structurally diverse set of input molecules.

Feature rich compounds

Include the most active compounds

Spread sheet set up for hip hop

Molecules hypothesis generation work bench imported into a spread sheet principal

specific the reference molecules references configuration models are potential centers for

hypothesis

If (0) do not consider these molecules

If (1) consider configuration of the molecules.

If(2) use this compound as a reference molecules used only for hip hop hypothesis

generation

Maximum omit features how many feature for each compound may be omitted

If (0) all features must map to generate hypothesis

If (1) all but one feature must map to generate hypothesis

If(2) features need to map to generate hypothesis used only for hip hop hypothesis

generation

When compound data appear in the spreadsheet, you are ready to add values in the

Principal and MaxOmitFeat columns. Common-features hypothesis generation uses values in

these columns to determine which molecules should be considered when building hypothesis

space and which molecules should map to all or some of the features in the final hypotheses.

In the Principal column, a value of 2 means that all the chemical features in the compound

will be considered in building hypothesis space. A value of 1 means that features will be

considered when generating hypotheses and that at least one mapping for each generated

hypothesis will be found unless the Misses or Complete Misses options are used. A value of 0

means the compound will be ignored.

The Max Omit Feat column specifies how many hypothesis features must map to the

chemical features in each compound a 0 in this column forces mapping of all features, a 1 means

that all but one feature must map, and a 2 allows hypotheses to which no compound features map

Page 40: QSAR, Pharmacophore and Docking Studies on Human Phaspholipase a2 Inhibitors

40

4.2.2b HYPOGEN

Hypogen attempts to derive SAR models for a set of molecules for which activity value

(IC50 or Ki) on a given biological target are available. Hypogen optimizes hypothesis that are

present in the highly active compounds in the training set. But missing among the least active (or

inactive) ones. It attempts to construct the simplest hypothesis that best correlates that activity

(estimates vs. measured) the predicted models are created the predicted models are created in

three stages:

Constructive

Subtractive

Optimization

The constructive phase identifies hypothesis that are common to the most active set of

compounds.

The most active set is determined by the following equation of compounds. The most active set is

determined by the following equation

MA x UncA = (A/UncA)>0.0

Where MA is the activity of the most active compounds

Unc is the uncertainty in the measured activity and

A is the activity of the compound

The most active set of compounds is limited to a maximum of eight. Once the set is

determined hypogen enumerates all possible pharmacophore features for each of the

conformations for the two most active compounds. Furthermore, the hypothesis must fit a

minimum subset of features of the remaining most active compounds in order to be considered. At

the end of the constructive phase a database of every number of pharmacophore configurations is

generated. The objective of the substractive phase is to identify those pharmacophore

configurations is generated. The objective of the subtractive phase is to identify that

pharmacophore configuration developed in the constructive phase that is also present in the least

active set of molecules and remove them. The first step is the identification of the least active

compounds. This is accomplished by these of equations log (A) - log (MA).305 '' where the A is

the activity of the current compound and MA is the activity of the most active compound. in

simple terms, all compounds whose activity is 3.5 order of magnitude less than that or the most

active compound are considered to be in the set of least active molecules. The value 3.5 is user

adjustable parameter, if needed (i.e., if the activity range of the dataset does not span more than

3.5 orders of magnitude the subtractive phase identifies the hypothesis that are common to the

least active compounds the least active set is determined by the following equation,'' log (cmpdx)-

log (most active compounds)3.5''. It enumerates all possible pharmacophore configurations. Then

it checks for configuration with the most active compounds and eliminates if shred by more than

half of the least actives leading to feasible pharmacophores.

The optimization phase involves improvement of the hypothesis score. Small

perturbations are applied to those pharmacophore configurations that survived the subtractive

phase and that are scored based on errors I activity estimates from regression and complexity of

the hypothesis. The cost of a hypothesis is a quantitative extension of Occams razor (everything

else being equal, the simplest model is preferred;

Page 41: QSAR, Pharmacophore and Docking Studies on Human Phaspholipase a2 Inhibitors

41

Figure14 Hypogen process flow

A detail of the cost of each pharamcophore is computed by the sum of three costs: weight

error configuration. While the weight component increases with deviation of the feature weight

from the ideal value of 2.0, the error component increases with RMS difference between the

measured and estimated activities. The configuration cost is fixed and depends on the complexity

of the pharamcophore upon completion of this phase.

Hip hop and hypo use conformations generated in confirm to align chemically important

functional groups common to the molecules in a study set. Biological activity data can be

incorporated into this hypothesis so that the best hypothesis for predicting activity are generated

and selected. Additionally, you can use structures that have been aligned in these programs to

generate a receptor surface model.

HYPOGEN TRAINING AND TEST SET SELECTION

Selection of the training set molecules is one of the most important exercises the user

must purpose for the following reasons:

Catalyst derives the information used in subsequent analysis from those structures thus, the

garbage in garbage out” paradigm certainly applies.

The statistical procedures applied during analysis have limits in terms of over and under

fitting the data.

Data sets that are ideal for those analysis procedures and data sets from typical medicinal

chemistry structure activity series are often not the same thing.

The ideal training set

1. At least 16 compounds are necessary to assure statistical power.

2. Activities should span 4 orders of magnitude.

3. Each order of magnitude should be represented by at lest 3 compounds.

4. No redundant information.

5. No excluded volume problems.

Page 42: QSAR, Pharmacophore and Docking Studies on Human Phaspholipase a2 Inhibitors

42

METHODOLOGY

INTRODUCTION

To build a better pharamcophore the following steps were employed

1. Building set of molecules

2. Conformer generation

3. Hypothesis generation

4. Database generation

5. Database search

6. Compare / fit to estimate activity

Criteria to generate successful hypothesis are:

1. Cost factor: a dumping score that is the difference between fixed and null cost should be

greater than so hits i.e., larger difference gives better prediction.

2. Fixed cost represents the simplest method model that fits all data perfectly and the null cost

represents the highest cost of a pharmacophore with no features and which estimates

activity to be average of activity data of training set of molecules.

3. The configuration value which is a measure of magnitude of hypothesis space for a given

training set should be less than 18. If it is above, more degree s of freedom and the result

may not be useful.

4. The estimated and the actual activity data correlation value should be around 1.0

5. The RMS deviations, which should be as low as possible, nearly equal to 0, which

represents the quality of the correlation between the estimated and the actual activity data.

METHOD

BUILDING A SET OF MOLECULES

All molecules were built using catalyst view compound work bench. They were cleaned

using option 2D beautify and minimized using CHARMm like force field.

CONFORMER GENERATION

A conformer is a representation model of the possible conformational space of a ligand. It

is assumed that the biologically active conformation of a ligand (or a close approximation there of)

should be contained within this model. Conformers were generated for all molecules with cut off

energy range 20 Kcal /mol and up to a maximum of 255 conformers.

COST HYPOTHESIS

The lowest cost hypothesis is considered to be the best. However, hypothesis with costs

within 10-15 of the lowest cost hypothesis are also considered as good candidates. The units of

cost are binary bits. Hypothesis costs are calculated according to the number of bits required to

completely describe a hypothesis. Simplex hypothesis require bits for a complete description and

the assumption is made that simplex hypothesis are better.

HYPOTHESIS GENERATION / PHARAMCOPHORE SEARCH

A pharmacophore model consists of a collection of features necessary for the biological

activity of the ligand arranged in 3D space, the common ones being hydrogen bond acceptor,

hydrogen bond donor and hydrophobic features. Hydrogen bond donors are defined as vectors

from the donor atom of the ligand to the corresponding acceptor atom in the receptor. Hydrogen

bond acceptors are analogously defined. Hydrophobic features are located at the centroids of

hydrophobic atoms.

Conformation s for all molecular were generated in view compound work bench using

poling algorithm and the best quality conformer generation method. The best conformer

generation considers the arrangement of atoms. Best conformer generation accepts a maximum of

255 conformers for the set of molecules catalyst generated conformers that provided the most

Page 43: QSAR, Pharmacophore and Docking Studies on Human Phaspholipase a2 Inhibitors

43

comprehensive treatment of flexible ring systems. All the conformers are automatically saved and

the number of conformers generated for each molecule with lowest conformer energy in kcal/mol.

Conformers were selected that fell within 20 kcal/mol range above the lowest energy

conformation found.

HYPOTHESIS GENERATION

The pharmacophore hypothesis generated in generate hypothesis work bench. The

molecular were selected as training set based on order of magnitude. Hypothesis generation

carried out by employing following assumptions.

1. Highly active and most inactive molecule should represent in the training set.

2. At least 3 or more molecules from each order of magnitude should be selected for

pharmacophore generation.

3. A minimum of 15 or above molecules will constitute for a training set.

4. Molecules selected should represent diversity towards chemical features.

HYPOTHESIS CONSIDERATIONS

In order to achieve a better pharmacophore, the following limits or considerations should be

met by generated hypothesis.

Configuration value should be around 17.

RMS should be as low as possible, preferable nearer to zero.

Correlation should be around 1.0

Cost factor difference between fixed cost and Null cost should be between 40-80 bits.

FACTORS THAT DETERMINE THE QUALITY OF PHARMACOPHORE

The overall cost of a hypothesis is calculated by summing three cost factors, a weight cost,

an error cost and a configuration cost. These are qualitatively defined.

WEIGHT COST

A value that increases in a Gaussian form as the feature weight in model deviates from an

idealized value of 2.0. This cost factor is designed to favor hypothesis where the feature Weights

are close to 2.

ERROR COST

A value that increases at the RMS difference between estimated and measured activities for the

training set molecules increases. This cost factor is designed to favor models where the correlation

between estimated and measured activities is better.

CONFIGURATION COST

This is a fixed cost which depends on the complexity of the hypothesis space being optimized. It is

equal to the entropy of the hypothesis space.

Of the three, the error cost factor has the major effect in establishing hypothesis cost.

During the beginning phase of an automated hypothesis generation, Catalyst calculates the cost of

two theoretical hypothesis one in which the error cost is minimal (all compounds fall along a line

of slope=10, and one where the error cost is high (all compounds fall along a line of slope +O).

These models can be considered upper and lower bounds for the training set. The cost values for

them are useful guides for estimating the chances for a successful experiment and are available

within 15 minutes from the start of the run because these experiments can easily require days of

run time. The ideal hypothesis cost (fixed cost) is reported in the full file found in the hypothesis

Page 44: QSAR, Pharmacophore and Docking Studies on Human Phaspholipase a2 Inhibitors

44

generation directory. This value tends to be 70-100 bits. The null hypothesis cost is reported in the

log file found in the same directory and is usually higher than the fixed cost. What is important is

the difference between these two costs. The greater the difference, the higher is the probability for

finding useful model. In terms of hypothesis significance, what really matters is the magnitude of

the difference the cost of any returned hypothesis and the cost of the null hypothesis. In general, if

this difference is greater than 60 bits, there is an excellent chances the model represents a true

correlation. Since, most returned hypothesis will be higher in cost than fixed cost model, a

difference between fixed cost and null cost of 70 or more will be necessary in order to achieve the

60 bit difference. If a returned hypothesis has a cost that differs from the null hypothesis by 40-60

bits, there is a high probability it has a 75-90% chances of representing a true correlation in the

data. As the difference becomes less than 40 bits, likelihood of the hypothesis representing a true

correlation in the data rapidly drops below 50%%. Under these conditions, it may be difficult to

find a model that can be shown to be predictive. In the extreme situation where the fixed and null

cost differential is small (>20), there is little chance of succeeding and it is advisable to reconsider

the training set before proceeding. Another useful number is the entropy of hypothesis space. This

value is calculated early in the run and is in full near the value for fixed cost.

TRAINING SET

1. Training set should contain the most active compounds.

2. Each compound must posses some thing new to teach catalyst.

3. If two compounds have similar structures (collections of features), they must differ in

activity by an order of magnitude to be included, otherwise, pick only the more active of

the two.

4. If two compounds have similar activities (within one order of magnitude), they must be

structurally distinct (from a chemical feature point of view) in order to both be included,

other wise pick only the most active of the two.

The pharmacophore features are perceived from the hip hop data, the features present

in training set molecules are hydrogen bond acceptor, hydrophobic aliphatic, hydrophobic

aromatic, and 22 molecules are selected for the training set and activity values are loaded into a

spread sheet and all the preferences and uncertainty values are loaded. Then the hypogen

algorithm is used to generate the hypothesis are generated.

Page 45: QSAR, Pharmacophore and Docking Studies on Human Phaspholipase a2 Inhibitors

45

4.2 STRUCTURE OR TARGET BASED DRUG DESIGN

Structure based drug design, the three dimensional structure of drug target interacting with

small molecules (drug) is used to guide drug discovery. . Drug targets are typically key molecules

involved in a specific metabolic or cell signaling pathway that is known, or believed, to be related

to a particular disease state. Drug targets are most often proteins and enzymes in these pathways.

Drug compounds are designed to inhibit, restore or otherwise modify the structure and behavior of

disease-related proteins and enzymes.

SBDD uses the known 3D geometrical shape or structure of proteins to assist in the

development of new drug compounds. The 3D structure of protein targets is most often derived

from x-ray crystallography or nuclear magnetic resonance (NMR) techniques as they have the

resolution few angstroms (about 500,000 times smaller than the diameter of a human hair). At this

level of resolution, researchers can precisely examine the interactions between atoms in protein

targets and atoms in potential drug compounds that bind to the proteins. This ability to work at

high resolution with both proteins and drug compounds makes SBDD as one of the most powerful

methods in drug design

Once bound at the receptor site, drugs may act either to initiate a response (agonist action or

stimulant) or decrease the activity potential of that receptor (antagonist action or Depressant) by

blocking access to it by active molecules. Thus, any drug may have structural features that

contribute independently to the affinity for the receptor and to the efficiency with which the drug

receptor combination initiates the response (intrinsic activity or efficiency). The response is

related to the drug receptor complexes. The affinity of a drug may be estimated by comparison of

the dose required to produce a pharmacological response with the dose required by a reference

standard drug or the natural ligand for that receptor. The affinity of a drug may be estimated by

comparison of the dose required to produce a pharmacological response with the dose required by

a reference standard drug or the natural ligand for that receptor. Structure based drug design, the

three dimensional structure of drug target interacting with small molecules (drug) is used to guide

drug discovery. Structure based drug deigning is employed with the following parts:-

4.2.1 Structure based pharmacophore generation:

Structure based pharmacophore approach was find an out the essential feature of active

site which can contribute for ligand binding.

The interaction generation protocol takes an input receptor and a defined active site and

analyzes the active site for donors, acceptors, and hyderophobes. The result of the calculation is an

interaction map. The density of polar site parameter specifies the density of the vectors in the

interaction site for hydrogen bonds. The density of lipophilic sites parameter specifies the density

of points in the interaction site for lipophilic atoms.

Procedure:

1. Load the interaction generation protocol from the protocols explorer. The parameters

display in the parameter explorer

2. Ensure that the structure you want to define as the receptor is open in 3d window .use the

binding site tool panel to define the structure as the receptor.

3. Set the input site sphere parameter to define the active site. Select the ligand from the

receptor ligand complex and define the input site sphere

4. The radius of the site sphere can change by selecting the sphere and changing the radius in

the attributes dialog.

5. Select the receptor structure from the input receptor parameter list.

6. select the sphere as the input site sphere parameter

7. Set the remaining parameter as desired .an run the protocol.

Page 46: QSAR, Pharmacophore and Docking Studies on Human Phaspholipase a2 Inhibitors

46

4.2.2 Docking:

Molecular docking is the technique that is used to study molecular binding and how

molecules bind. The term “docking” is mostly related to protein molecule interactions. Following

chart shows the work flow of the docking process.

Figure 15 docking work flow

4.2.2a LIGAND FIT

Ligand fit is designed to search the binding site of a protein and dock a series of potential

ligands into the binding site. During docking the protein is rigid, in which the ligand remains

flexible allowing the conformations to be searched and docked with in the binding site. The three

dimensional structure of protein and ligand are required. There are three key steps in this process.

a. Site search

b. Conformational search

c. Ligand fitting

a. SITE SEARCH

The position and shape binding site of protein is defined to a grid. The active site shape is

defined based on the shape of the protein, from which all sites are detected. Docked ligand method

is used to define active site, in which unoccupied grid points with in a certain user definable

distance to ligand atoms are collected to form the site.

Page 47: QSAR, Pharmacophore and Docking Studies on Human Phaspholipase a2 Inhibitors

47

b. CONFORMATIONAL SEARCH

The Monte Carlo simulation is employed in the conformational search of the ligand. During

the search, bond lengths and bond angles are untouched only torsional angles (except those in a

ring) are randomized. Therefore, the ligand molecules should be energy minimized to ensure

correct bond lengths and bond angles before using ligand fit.

c. LIGAND FITTING

After a new conformer is generated, the ligand fitting is carried out in two steps. First the

non mass- weighted principle moment of inertia (PMI) of the binding site is compared with non

mass- weighted principle moment of inertia (PMI) of the ligand. If the value (Fitvalue) is above the

threshold or not better fitting results previously saved, no further docking process will be

performed. If the value (Fitvalue) is better than previously saved results the ligand is positioned

into the binding site according to the PMI. Because PMI is a scalar property, there are four

possible positions for the ligand to orient in the binding site. For each position, the corresponding

docking score is computed.

The docking score is negative value of the non-bonded inter molecular energy between

ligand and protein. After the docking score is calculated, for each orientation it is compared with

the results saved previously. If the new one is better, it is saved, and then the process of

conformational search and ligand fitting is iterated until number of trials is reached. Finally rigid

body minimization is applied to the saved conformations of the ligand to optimize their positions

and docking scores.

PROCEDURE

Steps followed for ligand fit

1. Potent inhibitor molecules which can inhibit the action of spla2 were taken.

2. Molecules with diversified similarities and pharmacophore features were selected from the

literature.

3. The molecules which are to be docked in a receptor site are created in a SD file so as all

molecules are processed for the docking score at a site.

4. The active site of a protein is identified by the find site from receptor cavities which is

processed by the flood flow algorithm.

5. The identification of the active site is located by the already docked ligand

6. The protein molecule is selected, the set of molecules in the SD file are chosen and docking

score is calculated.

7. Thus, the docking score for a set of molecules are calculated through ligand fit.

4.2.2b C-Docker:

C docker is a grid based molecular docking method that employs charm. It has been

employed in ds through the dock ligands (cdocker) protocol. In c docker, the receptor is held rigid

while the ligands are allowed to flex during the refinement. Random ligand conformations are

generated from the initial ligands structure through high temperature molecular dynamics followed

by random rotations. The random conformations are refined by grid based simulated annealing and

a final grid based or full force field minimization.

C-Docker steps:

1. Define the receptor and search for binding sites,

2. Prepare and run the dock ligands (c docker) protocol,

Procedure

1. open the receptor protein and apply the charmm force field

Page 48: QSAR, Pharmacophore and Docking Studies on Human Phaspholipase a2 Inhibitors

48

2. define the selected molecule as a receptor after that select the ligand define sphere from

selection

3. open the c docker protocol and set the parameters

4. run the protocol

4.2.2c Lip dock:

Lip dock uses protein site features referred to us hot spots. Hot spots consist of two types:

polar and pallor. a polar hot spot is preferred by a polar ligand atom (for example a hydrogen bond

donor are acceptor ) and an pallor hotspot is preferred by an pallor atom the receptor hot spot file

is calculated prior to the docking procedure.

However, if desired, a pre defined or user adjusted hotspot file can be used .the protocol allows the

user to specify several modes for generating ligand conformations for docking

The rigid ligand poses are placed into the active site and hot spots are matched as triplets. The

poses are pruned and a final optimization step is performed before the poses are scored. Lip dock

algorithm has four function aspects:

1. conformation generation of the ligands,

2. creating a binding site

3. matching the binding site image and the ligand

4. optimization stage and scoring

Procedure:

In the protocol explorer, expand ligand receptor interactions and then double click dock

ligands (lib dock). The lib dock ligands protocol opens in the parameter view with the

parameters for setting up the protocol.

Ensure that the structure you want to define as the receptor is open and selected in a 3d

window. The receptor should have hydrogen‟s added and all atom valances satisfied. open

the binding site tool pane and click define selected molecule as a receptor

On the parameter explorer, select the sphere as the input site sphere parameter. Before the

input site sphere parameter can be assigned, define the site sphere from the binding site

tool panel.

Click on the button for the input ligand parameter, this opens the specify ligands dialog.

On the dialog, select the legands from a table browser, a 3d window or .sd file.

The number of hot spots parameter determines how many hot spots to generate.

Set the remaining parameters as desired. Run the lip dock protocol.

Page 49: QSAR, Pharmacophore and Docking Studies on Human Phaspholipase a2 Inhibitors

49

4.2.2d DENOVO LIGAND DESIGN

LUDI

Ludi is method for the denovo design of ligand for protein (inhibitor)

It can be also suggest modification of known ligand that may enhance the target protein. The

following Chart shows the ludi work flow.

Figure 16: ludi work flow

Ligand design:

The design of new ligand for protein (enzyme inhibiter) for protein is carried out if the structure is

known. If the structure of one or more protein –inhibiter complex is known ,the design ma be

added by study that identifies essential ligand- protein interaction .there are two approach to find a

compound csn fit into active site

The known structure approach:

Searching through database such a Cambridge structure database identifies that structure that fit

the active site. The advantage of this approach is that the molecules retried from the database do

exit and the structure represents low energy confermation. This approach does not address the

issue conformation flexibility.

The fragment approach:

This approach use a library of fragment the idea is to position molecular fragment into the active

site, in such a way that hydrogen bond can be formed with the protein and hydrophobic pockets

filled with hydrophobic groups. The fragment is than connected by suitable apacer fragment to

form single molecules.

Page 50: QSAR, Pharmacophore and Docking Studies on Human Phaspholipase a2 Inhibitors

50

Ludi method:

Ludi is based on fragment approach method. It suggests how suitable and small fragment can be

positioned into cleft of protein structures. This positioning is the strength ludi because it

immediately provides with the ideas about how putative binding site on the protein can be

saturated by the fragment and those fragment might be linked together .ludi works in three steps

It calculate interaction site within the protein active site or from the active angles.

It searches libraries for fragments and fits than onto the interaction sites

To process an alignment or linked for the fragment.

Ludi distinguishes four types of interaction sites.

H-donor

H acceptor

Lipophilic aliphatic

Lipophlic aromatic.

The aromatic and aliphatic interactions are suitable sites for hydrophobic interactions

The H donor and H acceptor interaction sites are suitable for H bond formation. Ludi is capable

for fitting fragments on to the interaction sites and simultaneously a linking (i.e linking) them to

an existing ligand.

Method:

1. Identification of chemical nature of active site amino acids

2. Fragments identification and analysis of ludi score

3. Searching for link

4. Linking the fragments

5. Fusing the fragment and linking

6. Docking validation.

FRAGMENT FITTING

The next step is to fit fragments onto the interaction sites. Ludi searches the list of

interaction sites by distance criteria for suitable sets of two to sites to match the fragments.

Required interaction are specified are specified using targeted mode. In targeted mode fragments

are require to interact with the protein atom or atoms specified by the user. Any fragment fit that

does not interact with the entire set of specified target atoms is rejected.

To fit the fragment, Ludi performs a root mean squares (RMS) superimposition using

algorithm given by Kabasch (1978). A fragment fit is accepted if the RMS value is less than a user

defined threshold (typically 0.2A to 0.6A) , and no vanderwaals overlap of the fitted fragment with

the protein occurs , and if ,the electrostatic check parameter on the ludi runtimes parameters

control panel is checked , no unacceptable electrostatic repulsions are found. When the receptor

structure is not known, a fragment fit is rejected if the fragment extends outside the volume

defined by the set of active analogs.

LINK SITES: ALIGNING FRAGMENTS WITH PARTIALLY BUILT LIGANDS

Ludi is capable of fitting fragments onto the interaction sites and simultaneously aligning

(i.e. linking) them to an existing to a ligand. For this purpose, link sites are defined on the ligand.

A link site is a hydrogen atom that all the hydrogen atoms of the positioned ligand (within a user

specified cutoff radius) are link sites.

The ludi works as described above:

Page 51: QSAR, Pharmacophore and Docking Studies on Human Phaspholipase a2 Inhibitors

51

LUDI FRAGMENT LIBRARIES

The Ludi fragment library is divided into two parts. The de novo library is used when Ludi is run

in no-link mode. The link library is used when Ludi is run in link mode. The de novo library and

the link library each consist of two files, a file that specifies the fragment topologies and a file that

specifies the interaction types of fragment functional groups.

PROCEDURE

1. It calculates

interaction sites within the protein sPLA2 active site or from the active analogs.

2. It searches

libraries for fragments and fits them from onto the five interaction sites which are present

at the active site.

3. It proposes an

alignment or linking for the fragments and the new ligand is designed.

The highest activity with the best dock score is better fitted when compared to

other. A knowledge based approach is to suggest possible binding positions. The present

experimental studies carried out using ludi program. This program is studied to dock small

molecular fragments within protein binding sites using interactions between the donor hydrogen

and its acceptor is close to 1.8Å and the angle subtended at the hydrogen is rarely less than 1.20A.

Information about the preferred geometries of such interactions can be obtained from analysis of X

ray crystallographic database. Kelbe has performed a very careful analysis of non bonded contacts

observed in the CSD.

Page 52: QSAR, Pharmacophore and Docking Studies on Human Phaspholipase a2 Inhibitors

52

Page 53: QSAR, Pharmacophore and Docking Studies on Human Phaspholipase a2 Inhibitors

53

5.1. QSAR:

In the present study quantitative structure activity relationship studies were carried

out on phospholipasea2 inhibitors in order to design selective and potential inhibitors. QSAR

models were developed using1D and 2D-descriptors using discovery studio software. QSAR

attempts to model the activity of a series of compounds using measured or computed properties of

the compounds. In the equation the term „N‟ means the number of data points, r2

which is the

square of the correlation coefficient which describing the binding of the compounds to the QSAR

model. XV r2, a squared correlation coefficient generated during a validation procedure using the

equation

XV r2 = (SD PRESS)/SD

SD means the sum of squared deviations of the dependent variable values from their

mean the predicted sum of squares (PRESS), the sum of overall compounds of the squared

differences between the actual and the predicted values for the dependent variables. The PRESS

value is computed during a validation procedure for the entire training set. The larger the PRESS

value the more reliable is the equation. XV r2 is usually smaller than the overall r

2 for a QSAR

equation. It is used as a diagnostic tool to evaluate the predicted power of an equation generated

using the multiple leaner regression method.

GFA work by generating random populations of solution to a problem, scoring the

relative quality of the solution , and caring forward the most fit solutions or analogues(generated

through mutation and crossover)of other solutions to iteratively generated(and finally converge

on)new, more fit solution. In this study GFA analysis was done with following parameters.

Population size

Initial equation length

Final equation length

Number of generation

Boot strap r2 correlation coefficient calculated during the validation procedure. 79

compounds were included in the training set to generate the primitive QSAR model covering the

widest data range of IC50 values 0.005 to 50.01 µM. The predictive characters of QSAR were

further assessed using test molecules. To judge the predictive ability of the QSAR model for new

drug candidates the IC50 values for the test and training set were evaluated.

GFA parameters

Number of rows in model 79

Population 100

Maximum generation 50000

Initial terms per equation 20

Scoring function

Friedman

LOF

Mutation probability 0.1

Page 54: QSAR, Pharmacophore and Docking Studies on Human Phaspholipase a2 Inhibitors

54

The GFA method performs a search over the space of possible QSAR models using lack of fit

(LOF) scores to estimate the fitness of each model. These models lead to the discovery of

predictive QSAR equations.

1

GFA equation = 4.7849

+ 0.00716121 * −In−Situ Starting Energy

− 2.0176 * Activ

+ 0.10343 * Dipole_Mag_Propgen_VAMP

− 0.610585 * Local_polarity_Propgen_VAMP

+ 0.26681 * Mean_Polarizability_VAMP

+ 0.633171 * Num_H_Acceptors

− 0.149947 * Num_RotatableBonds

− 0.000507116 * Octupole_XYY_VAMP

+ 0.0647933 * RIJestateSumHal_Propgen_VAMP

+ 8.73998e−05 * −In−Situ Final Energy * −In−Situ Final Energy

+ 5.40146e−05 * ESP_point_count__3_Propgen_VAMP *

ESP_point_count__3_Propgen_VAMP

+ 17.3371 * Molecular_FractionalPolarSurfaceArea * Molecular_FractionalPolarSurfaceArea

− 7.29063e−07 * No._of_surface_points_with_−ve_ESP_Propgen_VAMP *

No._of_surface_points_with_−ve_ESP_Propgen_VAMP

+ 86.4313 * QsumHal_Propgen_VAMP * QsumHal_Propgen_VAMP

− 0.000148821 * Quadrupole_YY_VAMP * Quadrupole_YY_VAMP

− 2.65319e−07 * Total_Energy_VAMP * Total_Energy_VAMP

+ 13.6832 * Activ * Covalent_hydrogen_bond_acidity_Propgen_VAMP

− 0.000694297 * Activ * No._of_surface_points_with_+ve_ESP_Propgen_VAMP

+ 1.85589e−06 * DMol3_Mol_ID * Electronic_Energy_VAMP

+ 0.0165753 * Num_AromaticRings * RIJestateSumO_Propgen_VAMP

From the above equation, the positive values are the reference for the presence of specific group

at that point and increase the activity of molecule and the negative values indicate the presence of

ionic group which reduce the activity.

Table: The validation statistics for the model.

Fridman LOF 0.323

R-squared 0.968

adjusted R-squared 0.957

cross validated R-squared 0.941

lack of fit points 58

error for non significant LOF 0.176

significance of regression F value 89.789

Page 55: QSAR, Pharmacophore and Docking Studies on Human Phaspholipase a2 Inhibitors

55

Table 8: Experimental and predicted values of Training set compounds using GFA

MOLECULE NAME Activity GFA predicted value

1 7.89 7.829

10 6.97 7.091

11 8 7.983

12 7.14 7.103

13 8.05 7.953

14 6.88 6.489

16 6.09 6.336

17 7.09 7.383

18 6.97 7.373

19 7.89 7.743

20 8.15 8.314

21 6.96 6.866

23 7 6.813

24 7.96 7.684

25 6.83 7.062

26 7.96 7.82

12d 4.72 4.727

15b 7.85 7.813

15c 7.82 7.651

15e 7.89 7.624

15g 7.52 7.998

27ii 7.77 7.816

28i 8.1 7.984

28ii 8.22 7.958

28iv 8.05 8.121

Page 56: QSAR, Pharmacophore and Docking Studies on Human Phaspholipase a2 Inhibitors

56

28ix 7.68 7.834

28v 8.3 8.178

28viii 7.74 7.6

28x 7.72 7.556

28xi 7.64 7.831

28xii 7.21 7.563

28xiii 7.77 7.628

28xix 8 7.88

28xl 4.3 4.315

28xv 5.18 5.235

28xvi 7.29 7.545

28xviii 7.96 8.108

28xx 7.68 7.59

28xxi 7.44 7.481

28xxii 7.6 7.476

28xxiii 6.92 6.9

28xxix 7.85 7.752

28xxv 7.34 7.428

28xxvii 8 7.777

28xxviii 7.54 7.419

28xxx 7.37 7.42

28xxxi 7.46 7.434

28xxxii 7 7.179

28xxxiii 6.55 6.931

28xxxv 7.42 7.265

28xxxvii 7.64 7.668

28xxxviii 7.82 7.623

43a 6.7 6.906

43b 5.36 5.201

Page 57: QSAR, Pharmacophore and Docking Studies on Human Phaspholipase a2 Inhibitors

57

43c 6.85 6.722

43d 5.47 5.449

43e 5.85 5.888

43f 5.89 5.827

43g 6.62 6.907

48b 4.66 4.701

49b 8.15 8.25

49c 7.34 7.224

49d 7.08 7.158

49e 7.26 7.307

49g 7.1 6.768

49h 7.36 7.424

50b 7.52 7.535

51a 4.3 4.288

65a 6.04 6.206

65b 6.38 6.487

65c 6.8 6.757

65d 7 6.956

65e 6.4 6.667

65f 6.7 6.623

65g 7.17 7.066

65i 7.6 7.399

67a 4.82 4.823

67b 4.92 4.791

6b 5.96 6.009

Page 58: QSAR, Pharmacophore and Docking Studies on Human Phaspholipase a2 Inhibitors

58

Experimental activity

Graph 1: Showing correlation between experimental and predicted activities by

QSAR equation using GFA method

Page 59: QSAR, Pharmacophore and Docking Studies on Human Phaspholipase a2 Inhibitors

59

Test Set

The purpose of QSAR is not only to produce the biological activity of the training set but

also to predict the values of the test set molecules. From the above equation obtained for the

training set molecules of known activity are introduced to study table so as to predict the

biological activity. A series of molecules are introduced to study table which are known as test set

molecules. After the prediction of activities of test set molecules the activity of prediction crosses

over 80%.

Table 9: Experimental and predicted values of Test set compounds using GFA

Molecule Name Activity GFA predicted activity

11c 6.6 6.676

11d 5.79 5.641

11g 7 6.95

11h 5.79 5.494

12a 6.77 5.789

12b 5.79 5.005

12e 7.4 5.808

12f 5.79 5.057

14a 7.85 6.128

16a 5.79 5.35

16b 7.52 5.601

16c 7.46 6.055

1b 6.9 5.523

indoxam 7.22 5.856

15 8 7.114

22 8 7.584

15f 7.89 7.544

Page 60: QSAR, Pharmacophore and Docking Studies on Human Phaspholipase a2 Inhibitors

60

28iii 8.22 7.941

28vi 8.05 7.749

28vii 8.1 7.907

28xiv 7.77 8.006

28xli 4.3 -5.902

28xvii 8.22 7.841

28xxiv 8.1 8.062

28xxvi 7.96 7.915

28xxxiv 8.05 7.372

28xxxix 7.8 8.938

28xxxvi 7.29 7.543

44b 7.1 6.663

51b 4.3 -4.277

65h 6.62 7.418

65j 6.77 7.154

Page 61: QSAR, Pharmacophore and Docking Studies on Human Phaspholipase a2 Inhibitors

61

Graph 2: Showing correlation between experimental and predicted activities by

QSAR equation using GFA method for test set.

The result generated from QSAR equation using GFA method, the values observed for r2

and XV r2 are in specific range and there is a good correlation between experimental and GFA

predicted activity as listed. Good correlation is observed between the experimental IC50 and

computational predicted IC50 values. It has been suggested as since the predictive ability of

equations is good, they can be used to develop new analogs.

Page 62: QSAR, Pharmacophore and Docking Studies on Human Phaspholipase a2 Inhibitors

62

Page 63: QSAR, Pharmacophore and Docking Studies on Human Phaspholipase a2 Inhibitors

63

Pharmacpohore

The work in discovery studio shows how chemical features hydrogen acceptor, hydrogen

donor, hydrophobic aliphatic of set of compounds along with their activities ranging over several

orders of magnitude can be used to generate pharmacophore hypothesis, that can successfully

predict the activity. The models were not only predictive within the same series of compounds but

differences classes of diverse compounds also effectively mapped onto most of the features

important for activity. The pharmacopore generated can be used for diversified structures that can

be potentially inhibit lethal factor inhibitors discovery and to evaluate how well any newly

designed compound maps in the pharmacophore developed in this study, using inhibitors against

lethal factor showed distinct features that may be responsible for the activity of the inhibitors.

Analogue based pharmacophore generation:

5.2. Common Feature Pharmacophore Generation (HIP HOP):

The 10 most active molecules were used to derive common feature based alignments. All the

10 most active molecules were considered as reference molecules to get the best features. The best

features obtained from hip-hop run method are

1. Hydrogen bond acceptor, 2. Hydrogen bond acceptor lipid

3. Hydrogen bond donor 4. Hydrophobic

5. Ring aromatic

Table 10 Summary of feature definition hits by molecule

Molecule A H D Z Y X N P W R

28v 7.70 7.70 1.73 4.00 2.00 2.00 1.00 0.00 0.00 6.00

28ii 7.80 7.80 1.80 4.00 3.00 1.00 1.00 0.00 0.00 4.00

28xl 8.05 8.05 1.81 2.91 1.82 1.00 1.00 0.00 0.00 4.00

67a 5.63 5.63 1.43 3.77 1.77 2.00 0.00 0.00 0.00 4.00

43d 7.48 7.48 1.78 3.00 3.00 0.00 1.00 0.00 0.00 2.00

12d 6.59 6.59 5.57 4.93 3.93 1.00 0.00 0.00 0.00 4.00 A-hydrogen bond acceptor: H-hydrogen bond acceptor lipid: D-hydrogen bond donor: z-hydrophobic

Y-hydrophobic aliphatic: X-hydrophobic aromatic: N-negativeionizable: P-positive with Exclusions

W- PositiveIonizabl: R-ring aromatic.

Page 64: QSAR, Pharmacophore and Docking Studies on Human Phaspholipase a2 Inhibitors

64

Table 11: Common Feature Pharmacophore Generation Rank File:

Hypo.

no

Pharmacophore

feature

Rank score Direct hit Partial hit Max fit

1

ZDA 192.485 111111111 000000000 3

2

ZHA 191.559 111111111 000000000 3

3

ZHA 191.559 111111111 000000000 3

4

YZA 190.387 111111111 000000000 3

5

ZHA 190.012 111111111 000000000 3

6

ZHA 190.012 111111111 000000000 3

7

ZHA 190.012 111111111 000000000 3

8

ZHA 190.012 111111111 000000000 3

9

ZDA 189.761 111111111 000000000 3

10

ZDA 189.735 111111111 000000000 3

Page 65: QSAR, Pharmacophore and Docking Studies on Human Phaspholipase a2 Inhibitors

65

5.3. HYPOGEN (Training set):

Sets of 5 hypotheses were generated using the data from 22 training set compounds.

Different cost values correlation coefficient RMS deviations and pharmacophore features are

listed in table.

TABLE 12: The 5 pharmacophore models generated by the hip-hop algorithm

Note: Cost difference=null cost (128.34)-total cost

The best pharmacophore is taken as the hypothesis 1 which has the highest cost difference,

lowest error cost, lowest RMS difference and the best correlation coefficient has two hydrogen

bond acceptor, one hydrophobic and one hydrogen bond donor features. The best pharmacophore

(hypo1) has the highest cost difference of 35.867, the best correlation coefficient and RMS

difference.

For the highly active compound (28v) in training set, mapped all the features are perfectly

to the features of Hypo 1. In compound 28v, HBA1 feature mapped to the electron rich O atom of

Sulfur Dioxide group and HBA2 feature corresponded to the another O group of Sulfur Dioxide

group. The HBD feature mapped to the NH group attached with Sulfur Dioxide group. The

Hydrophobic group was mapped to the methyl attached to 3rd-position of the benzene ring of the

compound.

.

Hypo. no

Total

cost

Cost

difference

Error

cost RMS Correlation Feature

1 92.689 35.867 70.98 1.077 0.951 AADZ

2 98.9081 29.6479 77.6027 1.376 0.896 AADZ

3 104.54 24.016 86.435 1.296 0.898 AADZ

4 117.235 11.321 91.143 1.24 0.908 AAZR

5 122 6.556 94.289 1.36 0.893 AAZR

Page 66: QSAR, Pharmacophore and Docking Studies on Human Phaspholipase a2 Inhibitors

66

Figure18: Blank Pharmacophore feature of sPLA2 inhibitors

Page 67: QSAR, Pharmacophore and Docking Studies on Human Phaspholipase a2 Inhibitors

67

Figure 19: Showing the distances between pharmacophore features

Page 68: QSAR, Pharmacophore and Docking Studies on Human Phaspholipase a2 Inhibitors

68

Figure 20: Overlapping of highest active inhibitor molecules (28v) of

training set with the best pharamcophore (Hypo1).

Page 69: QSAR, Pharmacophore and Docking Studies on Human Phaspholipase a2 Inhibitors

69

Figure 21: Overlapping of lowest active inhibitor molecule 28xl of training set with

the best pharamcophore (Hypo1)

Page 70: QSAR, Pharmacophore and Docking Studies on Human Phaspholipase a2 Inhibitors

70

Table 13: Results of pharmacophore hypothesis generated using training set.

Name Activity ConfNumber Estimate Fit Value

28v 0.005 199 0.009 10.976

28iii 0.006 18 0.011 10.887

20 0.007 142 0.034 10.413

13 0.009 78 0.005 11.24

28iv 0.009 201 0.07 10.098

22 0.01 23 0.015 10.768

26 0.011 10 0.096 9.956

1 0.013 43 0.162 9.73

10 0.106 81 0.037 10.369

21 0.109 182 0.139 9.798

25 0.148 42 0.207 9.625

28xxxiii 0.28 153 0.126 9.838

16 0.806 119 0.55 9.2

43e 1.4 18 0.083 10.019

43d 3.4 139 3.222 8.432

43b 4.4 24 2.778 8.496

28xv 6.6 210 3.918 8.347

67b 12 88 16.157 7.732

67a 15 46 29.519 7.47

12d 19 191 6.771 8.109

48b 22 41 7.699 8.054

28xl 50.01 169 2.114 8.615

Page 71: QSAR, Pharmacophore and Docking Studies on Human Phaspholipase a2 Inhibitors

71

Discussion

Pharmacophore models of sPLA2 lethal factor inhibitors are generated in HypoGen

module in DS software. HypoGen attempts to construct the simplest hypotheses that best

correlates the activities (experimental vs. predicted).

The dataset was divided into training set (22 compounds) and test set (89 compounds,),

considering both structural diversity and wide coverage of the activity range. The compounds with

activity with < 1 μM were considered as highly actives (+++), compounds with an activity range

of 1-10 μM as moderate actives (++) and activity of >10 μM as least actives (+).At end of run,

HypoGen generated 5 pharmacophore models. The Null cost for ten hypotheses was 128.556, the

fixed cost of the run was 79.954 and the configuration cost was 18.83. A difference of 48.602 bits

obtained between fixed and null costs is a sign of highly predictive nature of hypotheses. All 5

hypotheses generated showed high correlation coefficient between experimental and predicted

IC50 values, in the range of 0.95 to 0.89 and moreover, these are having cost difference less than

45 bits between the cost of each hypothesis and the null cost. It indicates that all the hypotheses

are having true correlation between 80-95%. The cost values, correlation coefficients (r), RMSD,

and pharmacophore features are listed in Table12.The best pharmacophore (Hypo 1) consisted of

two H-bond acceptor (HBA), an H-bond donor (HBD), and a hydrophobic feature with a

correlation coefficient (r) of 0.95, total cost (92.689), and lowest RMSD value (0.89) was chosen

to further validate its predictive power by estimating the activity of test set.

The predictability of Hypogen one was evaluated by using diversifies test set compounds.

The generated pharmacophore model has predicted the activity of a diverse dataset of 89 test set

compounds with correlation value of 0.7987. Hence from this analysis, Hypo1 was able to

distinguish active compounds from the inactive compounds

Page 72: QSAR, Pharmacophore and Docking Studies on Human Phaspholipase a2 Inhibitors

72

5.4 Structure based pharmacophore:

Structure based pharmacophore approach was to find an out the essential feature of

active site which can contribute for ligand binding.

.

Interaction generation:

Enumerates pharmacophore features from a protein active site. The site finding algorithm

from Ludi to identify points in the active site that could interact with the receptor. Creates a

pharmacophore query containing Hydrogen bond acceptor, donor and hydrophobic features from

these points

After interaction generation run, it Found 329 features: minimizied1DB41

Found 98 lipophilic features

Found 131 H-acceptor features(

Found 100 H-donor features

Found 0 Link features

Figure 22: cluster feature of interaction generation.

Page 73: QSAR, Pharmacophore and Docking Studies on Human Phaspholipase a2 Inhibitors

73

Figure 23: center points of cluster feature

Page 74: QSAR, Pharmacophore and Docking Studies on Human Phaspholipase a2 Inhibitors

74

Figure24: Mapping of 28v molecule with structure based pharmacophore feature.

This structure based pharmacophore features are useful for virtual screening of large

database.

Page 75: QSAR, Pharmacophore and Docking Studies on Human Phaspholipase a2 Inhibitors

75

5.5 LIGAND FIT

Every molecule in the prepared bio active compound SD file will be docked

into the binding site chosen, the fits will be automatically processed according to the preferences

chosen and saved into the output SD file. The results containing RMS calculations perform by

comparing the RMS difference of every fit and the first conformer in the input SD file.

Minimization energies of the fits in the presence of the protein and ludi score according to the

references can be seen in the input SD file the option of performing ligand fit using flexible fit

method carried out initially in a random conformation. The docking score is the negative values of

the non-bonded inter molecular energy; if the ligand atom has partial charge on it, the electrostatic

grid is used to estimate electrostatic energy. If it is a hydrogen atom, the hydrogen grid is used for

vanderwaals energy. Otherwise carbon grid is used. The following table enlists the docking score

and the corresponding minimization energies obtained for the beast conformer for each molecule.

The activity of the each molecule may be contributed by the best lowest energy obtained in the

ligand fit with the corresponding dock scores in table14 are as follows:-

Table14: Docking scores of inhibitors molecules of LF obtained after subjecting to ligand fit.

s.no Compound

name

dock score Internal energy

Highest active compounds

1.

2.

28V

28II

75.456

73.954

-16.873

26.318

lowest acting compounds

3

4

28Xl

51a

81.063

69.885

-18.654

19.771

Intermediate active compounds

5

6

43d

11d

57.36

44.873

-2.282

-2.553

Page 76: QSAR, Pharmacophore and Docking Studies on Human Phaspholipase a2 Inhibitors

76

Figure25: conformation search of high active compound (28v) inside the protein

(1DB4) binding site.

Page 77: QSAR, Pharmacophore and Docking Studies on Human Phaspholipase a2 Inhibitors

77

Figure26: highest acting Molecule 28V which has been subjected to ligand fit

showing its interaction.

Page 78: QSAR, Pharmacophore and Docking Studies on Human Phaspholipase a2 Inhibitors

78

Figure27: Hydrogen bond interaction of high active compound 28v with active site

amino acids

Page 79: QSAR, Pharmacophore and Docking Studies on Human Phaspholipase a2 Inhibitors

79

Figure28: Hydrogen bond interaction of low active compound 11d with active site

amino acids

Page 80: QSAR, Pharmacophore and Docking Studies on Human Phaspholipase a2 Inhibitors

80

Discussion:

Docking studies shows that the compound 28v having the high dock score of 75.456. And

compound 11d has the low dock score of 44.873.the following table shows distance and active site

amino acids forming the hydrogen bond interactions with 28V and 11d.

Table15: Hydrogen bond distances and hydrogen bond forming amino acids with 28v and 11d

compound

Compound name Hydrogen bond

Forming amino acids

Hydrogen bond

Distance(Å)

28v

1.gly29

2.his27

3.his47

4.lys62

3.159

2.930

2.17

2.95

11d 1.gly29

2.his47

3.11

2.14

Page 81: QSAR, Pharmacophore and Docking Studies on Human Phaspholipase a2 Inhibitors

81

5.6 C docker:

Every molecule in the SD file will be docked into the binding site chosen and in these

docking Docks ligands into an active site using CHARMm.

Uses a CHARMm-based molecular dynamics (MD) scheme to dock ligands into a receptor

binding site. Random ligand conformations are generated using high-temperature MD. The

conformations are then translated into the binding site. Candidate poses are then created using

random rigid-body rotations followed by simulated annealing. A final minimization is then used to

refine the ligand poses. The following table enlists the docking energy and the corresponding

minimization energies obtained for the beast conformer for each molecule. The activity of the each

molecule may be contributed by the best lowest energy obtained in the c- docker with the

corresponding dock in energy in table 15 are as follows:-

Table15: C-Dock energy of inhibitors molecules of C-Dock obtained after subjecting to legend

fit.

s.no Compound

name

C docker energy C docker Interaction

energy

Highest active compounds

1.

2.

28V

28II

-29.754

-29.436

-57.136

-52.133

lowest acting compounds

3

4

28Xl

51a

-38.076

-36.065

-50.647

-47.674

Intermediate active compounds

5

6

43d

11d

-31.718

-29.952

-55.081

-55.592

Page 82: QSAR, Pharmacophore and Docking Studies on Human Phaspholipase a2 Inhibitors

82

Figure29: conformation search of high active compound (28v) inside the protein

(1DB4) binding site.

Page 83: QSAR, Pharmacophore and Docking Studies on Human Phaspholipase a2 Inhibitors

83

Figure30: highest acting Molecule 28V which has been subjected to C-Dock

showing its interaction.

Page 84: QSAR, Pharmacophore and Docking Studies on Human Phaspholipase a2 Inhibitors

84

Figure31: Hydrogen bond interaction of high active compound 28v with active site

amino acids

Page 85: QSAR, Pharmacophore and Docking Studies on Human Phaspholipase a2 Inhibitors

85

Figure32: Low acting Molecule 11d which has been subjected to C-Dock showing

its interaction.

Page 86: QSAR, Pharmacophore and Docking Studies on Human Phaspholipase a2 Inhibitors

86

Discussion:

C-Docking studies shows that the compound 28v having the high c-docker energy of

28.754. And compound 28xl having the low dock score of -38.076. The following table shows

distance and active site amino acids forming the hydrogen bond interactions with 28V and 28xl.

Table16: Hydrogen bond distances and hydrogen bond forming amino acids with 28v and 11d

compound

Compound

name

Hydrogen bond

Forming amino acids

Hydrogen bond

Distance(Å)

28v

1.gly29

2.his47

2.723

2.38

28xl 1.his47 2.29

Page 87: QSAR, Pharmacophore and Docking Studies on Human Phaspholipase a2 Inhibitors

87

5.7 Lib dock

Docks ligands into an active site using hotspots. Hotspots are polar and apolar interaction sites.

Ligand conformations can be recalculated or generated on the using DS.

Table17: Lip Docking scores of inhibitors molecules of obtained after subjecting to ligand fit.

Name Activity Lib Dock Score Hot Spots

High active

28v 0.005 161.275

58.71,27.73,46.89,A,73,2

55.71,31.12,44.29,P,36,24

58.11,25.93,42.29,P,13,27

28ii 0.006 160.269

58.71,27.73,46.89,A,73,16

62.91,33.92,48.49,A,94,23

56.71,32.53,44.89,P,44,24

intermediate

28xl 50.01 146.517

55.91,30.53,43.09,P,18,21

62.71,33.73,47.09,A,77,23

63.91,33.13,47.49,A,83,24

51a 50.01 151.647

64.11,33.73,48.09,A,90,19

57.31,26.32,45.89,A,56,23

58.91,32.73,41.49,A,4,29

Low active

43d 3.4 160.571

60.11,27.73,47.89,A,87,2

57.31,33.73,46.49,A,69,14

62.11,33.13,47.89,A,88,30

11d 1.61 147.918

60.11,34.53,47.09,A,78,11

59.31,28.73,44.49,A,30,36

56.71,32.53,44.89,P,44,38

Page 88: QSAR, Pharmacophore and Docking Studies on Human Phaspholipase a2 Inhibitors

88

Figure33: highest acting Molecule 28V which has been subjected to Lib dock

showing its interaction.

Page 89: QSAR, Pharmacophore and Docking Studies on Human Phaspholipase a2 Inhibitors

89

Figure34: Hydrogen bond interaction of high active compound 28v with active site

amino acids

Page 90: QSAR, Pharmacophore and Docking Studies on Human Phaspholipase a2 Inhibitors

90

Figure35: Low acting Molecule 11d which has been subjected to Lib dock showing

its interaction.

Page 91: QSAR, Pharmacophore and Docking Studies on Human Phaspholipase a2 Inhibitors

91

Figure36: Hydrogen bond interaction of high active compound 11d with active site

amino acids

Discussion:

The lib dock score of the above stated molecules are all positive values. Thus the molecules

can be used as the potential ligands for the inhibition of sPLA2. The molecule 28v and 28xl are

found to have a dock score 161.275and 146.516respectively.

Page 92: QSAR, Pharmacophore and Docking Studies on Human Phaspholipase a2 Inhibitors

92

5.8 Ludi

Ludi is method for the denovo design of ligand for protein (inhibitor) it can be also

suggest modification of known ligand that may enhance the target protein. In these studies the

denovo legand UA6 found by ludi. Following table shows the 2d structure of the ligand and there

molecular properties

S

O

CH3

CH3O

CH3

5-(1-methoxy-4-methylpentan-3-yl)[1]benzothieno[3,2-b]furan

Molecular Formula = C17

H20

O2S

Formula Weight = 288.4045Composition = C(70.80%) H(6.99%) O(11.10%) S(11.12%)

Molar Refractivity = 87.04 ± 0.3 cm3

Molar Volume = 252.7 ± 3.0 cm3

Parachor = 643.3 ± 4.0 cm3

Index of Refraction = 1.604 ± 0.02Surface Tension = 41.9 ± 3.0 dyne/cm

Density = 1.140 ± 0.06 g/cm3

Dielectric Constant = Not available

Polarizability = 34.50 ± 0.5 10-24cm3

Monoisotopic Mass = 288.1184 DaNominal Mass = 288 DaAverage Mass = 288.4045 Da

Uses Ludi to search a library of small fragments to find candidates that bind in an active

site. Fragments in the library that overlay with a calculated interaction map are found.

Page 93: QSAR, Pharmacophore and Docking Studies on Human Phaspholipase a2 Inhibitors

93

Figure37: Ludi molecule with interaction map

Page 94: QSAR, Pharmacophore and Docking Studies on Human Phaspholipase a2 Inhibitors

94

C–docking result for ludi ligand:

Table18: C-Dock energy of Ludi molecules obtained after subjecting to C dock.

ludi ligand c docker energy c docker internal energy

ua6-1 -21.094 35.271

ua6-2 -21.172 35.408

ua6-3 -21.221 35.25

ua6-4 -21.373 35.031

ua6-5 -21.768 35.159

ua6-6 -21.769 35.173

ua6-7 -21.845 34.848

ua6-8 -21.889 35.449

ua6-9 -21.98 34.728

ua6-10 -22.007 34.815

Page 95: QSAR, Pharmacophore and Docking Studies on Human Phaspholipase a2 Inhibitors

95

Figure38: Ludi Molecule UA6 which has been subjected to C dock showing its

interaction

Page 96: QSAR, Pharmacophore and Docking Studies on Human Phaspholipase a2 Inhibitors

96

Figure42: Hydrogen bond interaction of Ludi compound ua6 with active site amino acids

Page 97: QSAR, Pharmacophore and Docking Studies on Human Phaspholipase a2 Inhibitors

97

Pharmacophore mapping of ludi ligand:

Table 19: Estimated value of ludi ligand by using pharmacophore model

Name Estimate Mapped Atoms

Fit

value

UA6 1.901

4,0.231,-3.402,2.042,1.6,HBA 1.11

18,4.905,0.304,-1.932,1.6,HBA 2.11

19,2.548,5.188,1.714,1.6,Hydrophobic1

27,2.147,0.625,0.307,1.6,centroid1

3.35

Figure43: Feature mapping of compound UA6 with parmacophore model

Page 98: QSAR, Pharmacophore and Docking Studies on Human Phaspholipase a2 Inhibitors

98

Discussion

Docking shows that the new ligand molecule (UA6) has the c-dock energy -21.094

and UA6 compound forming the hydrogen bonding with active site amino acids gly 22, gly 27and

his 47. As per the pharmacophore feature mapping studies showed the new compound having

estimated value of 1.901 and fit value 3.35.

Page 99: QSAR, Pharmacophore and Docking Studies on Human Phaspholipase a2 Inhibitors

99

Page 100: QSAR, Pharmacophore and Docking Studies on Human Phaspholipase a2 Inhibitors

100

6. Conclusion

As far as Insilco studies considered for human phospholipase A2 (sPLA2) the

algorithms such as QSAR, Pharmacophore and docking were used. These algorithms showed good

results and further investigation for the drug collaboration can be done.

The 3D QSAR studies conducted for training set compound gave a good r2 score of 0.936

with four outliers with a GFA graph with a Fit line representing the good correlation of the

compounds with the activities. The pharmacophore studies gave the best quantitative

pharmacophore model in terms of predictive value consisted of three features like Hydrogen bond

acceptor, Hydrogen bond acceptor lipid, Hydrophobic, and Ring aromatic. Hypogen which is

further validated by using a set of sPLA2 inhibitors gave a correlation value of 0.968. The

Pharmacophore studies showed four regions which showed interactions i.e., hydrogen bond

acceptor, Hydrophobic, hydrogen bond acceptor lipid and ring aromatic. docking studies shows

that the compound 28v having the high dock score of 75.456and the compound 11d having less

dock score 44.87.

The Insilco modeling helped to guide the lead optimization and lead to the generation of a

highly potent series of sPLA2 inhibitors with good drug like properties and is subject of another

communication. However, the scope for fine tuning and optimizing this potent class of sPLA2

inhibitor could lead to the generation of new therapeutic agents.

The combined approach of analogue and structure based drug designing methods allowed

us to gain an insight into predicting the enhanced activity and exploring the docking interactions

between amino acid residues of lethal factor and the ligand. Good ligands may not act as good

drugs. Thus, the prime objective of this project to prove the authenticity of our techniques

obtained from the various journals is completed using computer aided drug designing. The results

obtained are used to develop new ligand molecules and find their activities Insilco and proving the

same in accordance to the experimental values. Thus, the results reported can successfully employ

in the rational drug designing of novel and potent lethal factor inhibitors.

Page 101: QSAR, Pharmacophore and Docking Studies on Human Phaspholipase a2 Inhibitors

101

Page 102: QSAR, Pharmacophore and Docking Studies on Human Phaspholipase a2 Inhibitors

102

Reference:

Humphrey P. Rang MB BS MA DPhil FMedSci FRS Drug Discovery and Development:

Technology In Transition (2006-01-12)

computer aided drug design by T. J. Perun1 edition (February 22, 1989) page 369, page 453,

and page 455

Essential of medical pharmacology by KD Tripathi page no 167-184 and 254-265

Goodman and gilman‟s The pharmacological basis of therapeutics 10th

edition

Henry,.D.R.; Ozkabak,A.G. “conformational flexibility in 3D structure searching,” in the

encyclopedia of computional chemistry,1998

6. Mayer, R. J.; Marshall, L. A. New Insights on Mammalian Phopspholipase A2(s); Comparison

of Arachidonoyl-Selective and Non Selective Enzymes. FASEB J. 1993, 7, 339-348. (b)

Bomalaski, J. S.; Clark, M. A. Phospholipase A2 and Arthritis. Arthritis Rheumatism 1993, 36,

190-198.

7. (A) Vadas, P.; Pruzanski, W.; Stefanski, E.; Ruse, J.; Farewell, V.; McLaughlin, J.; Bombardier,

C. Concordance of Endogeneous Cortisol and Phospholipase A2 Levels in Gram-Negative Septic

Shock: A Prospective Study. J. Lab. Clin. Med. 1988, 111, 584.

(b) Aufenanger, J.; Zimmer, W.; Kattermann, R. Characteristics and Clinical Application of a

Radiometric Escherichia Coli-Based Phospholipase A2 Assay Modified for Serum Analysis. Clin.

Chem. 1993, 39, 605-613.

8. Nevalainen, T. J. The role of phospholipase A in acute pancreatitis.Scand. J. Gastroenterol

1980, 15, 641-650.

9. (a) Vadas, P.; Pruzanski, W. Role of Secretory Phospholipase A2 in the Pathobiology of

Disease. Lab. Invest. 1986, 55, 391-404.

(b) Pruzanski, W.; Vadas, P. Secretory Synovial Fluid Phosphlipase A2 and its Role in the

Pathogenesis of Inflammation in Athritis. J. Rheumatol. 1988, 15, 1601-1603. (c) Pruzanski, W.,

Vadas, P. Phospholipase A2: A Mediator Between Proximal and Distal Effectors of Inflammation.

Immunol. Today 1991, 12, 143-146.

10. (a) Mobilio, D.; Marshall, L. A. Recent Advances in the Design and Evaluation of Inhibitors

of PLA2. Annu. Rep. Med. Chem. 1989, 24, 157-166. (b) Sofia, M. J.; Silbough, S. A. Novel

Approaches to Anti-Inflammatory Agents as Therapeutics for Pulmonary Disease. Annu. Rep.

Med. Chem. 1993, 28, 109-118.

Page 103: QSAR, Pharmacophore and Docking Studies on Human Phaspholipase a2 Inhibitors

103

(c) Wilkerson, W. W. Anti-Inflammatory Phospholipase A2 Inhibitors. Drugs Future 1990, 15,

139-148.

11. Schevitz, R. V.; Bach, N. J.; Carlson, D. G.; Chirgadze, N. Y.; Carlson, D. K.; Dillard, R. D.;

Draheim, S. E.; Hartley, L. W.;Jones, N. D.; Mihelich, E. D.; Olkowski, J. L.; Snyder, D. W.;

Sommers, C.; Wery, J.-P. Structure-Based Design of the First Potent and Selective Inhibitor of

Human non-Pancreatic Secretory Phospholipase A2. Nature Struct. Biol. 1995, 2, 458-465

12. Marison, L.; Cockburn, W. F. The synthesis of pseudoconhydrine. J. Am. Chem. Soc. 1949,

71, 3402-3404. Konakahara, T.; Takagi, Y. Convenient method for the preparation of 2-

phenacylpyridines. Heterocycles 1980, 14, 393-396.

13 (a) Uchida, T.; Matsumoto, K. Methods for the construction of the indolizine nucleus. Synthesis

1976, 209-236. (b) Casagrande, C.; Invernizzi, A.; Ferrini, R.; Miragoli, G. Indolizine analogues

of indomethacin. Farmaco. Ed. Sci. 1971, 26, 1059-1073.

14. (a) Desidiri, N.; Galli, A.; Seslili, I.; Stein, M. l. Synthesis and binding properties to GABA

receptors of 3-hydroxypyridyl- and 3-hydroxypiperidyl- analogues of Baclofen. Arch. Pharm.

1992, 325, 29-33. (b) Kimura. E.; Kotake, Y.; Koike, T.; Shionoya, M.; Shiro, M. A novel cyclam

appended with 3-hydroxypyridine. An ambient donor ligand comprising a pyridyl N and

apyridynolate O- donor. Inorg. Chem. 1990, 29, 4991-4996.

15 Lau, C. K.; Tardif, S.; Dufresne, C.; Scheigetz, J. Reductive deoxygenation of aryl aldehydes

and ketones by tert-butylamineborane and aluminum chloride. J. Org. Chem. 1989, 54, 491- 494.

16. Desideri, N.; Manna, F.; Stein, M. L.; Bile, G.; Filippelli, W.; Marmo, E. Eur. Synthesis of 3-

hydroxy-2-pyridineacetic acid and its evaluation on experimentallipaemia. J. Med. Chem. 1983,

18, 295-299.

17. Bellesia, F.; Ghelfi, F.; Grandi, R.; Pagnoni, U. M. Regioselective R-bromination of carbonyl

compounds with trimethylbromosilane- dimethylsulfoxide. J. Chem. Res. (s) 1986, 428-429.

Potent Inhibitors of Secretory Phospholipase A2 Journal of Medicinal Chemistry, 1996, Vol. 39,

No. 19 3657

18. (a) Rapport, H.; Volcheck jr., E. J. The synthesis of desoxycarpyrinic and carprinic acids. J.

Am. Chem. Soc. 1956, 78, 2451-2455. (b) Iorio, M. A.; Gatta, F.; Michalek, H. Synthesis

andconformational aspects of acetoxypiperidinium iodides related to acetylcholine. Eur. J. Med.

Chem. 1980, 15, 165-171.

19. Flitsh, W. Pyrroles with fused six-membered heterocyclic rings:

(i) a-fused. Comprehensive Heterocyclic Chemistry; Bird, C. W., Cheesman, G. W. M., Eds.;

Pergamon Press: London. 1984; Vol. 4, pp 443-495.

Page 104: QSAR, Pharmacophore and Docking Studies on Human Phaspholipase a2 Inhibitors

104

20. Kakehi, A.; Ito, S.; Yamada, N.; Yamaguchi, K. Preparation of new nitrogen-bridged

heterocycles. 21. A facile synthesis of 2-indolizinethiols using new protecting groups. Bull. Chem.

Soc. Jpn. 1990, 63, 829-834.

21. Gresham, T. L.; Jansen, J. E.; Shaver, F. W.; Bankart, R. A.; Beears, W. L.; Prendergast, M. G.

â-Propiolactone. VI. Reactions with phenols, thiophenols and their salts. J. Am. Chem. Soc. 1949,

71, 661-663.

22. Reynolds, L. J.; Hughes, L. L.; Dennis, E. A. Analysis of human synovial fluid phospholipase

A2 on short chain phosphatidylcholine- mixed micelles: development of a spectrometric assay

suitable for a microtiterplate reader. Anal. Biol. 1992, 204, 190-197.

23. Schadlich, H. R.; Buchler, M.; Berger, H. G. Improved method for the determination of

phospholipase A2 catalytic concentration in human serum and ascities. J. Clin. Chem. Clin.

Biochem. 1987, 25, 505-509.

24. Tojo, H.; Ono, T.; Okamoto M. Reverse-phase high performance liquid chromatographic assay

of phospholipases: application of spectrometric detection to rat phospholiase A2 isozymes J. Lipid

Res. 1993, 34, 837-844.

25. Hamanaka, Y.; Fukushima, S.; Hiyama, T. a convenient synthesis of substituted

heteroaromatic compounds via the palladium- catalyzed cross-coupling reaction of organosilicon

compounds. Heterocycles 1990, 30, 303-306.

26. Dole, V. P.; Meinertz, H. J. Microdetermination of long-chain fatty acids in plasma and

tissues. J. Biol. Chem. 1960, 235, 2595-2599.JM960395Q 3658 Journal of Medicinal Chemistry,

1996, Vol. 39, No. 19 Hagishita et al.