Sucheta Thesis Final - R2
Sucheta Thesis Final - R2
A Thesis
Submitted to
Banasthali University
For the award of degree of
DOCTOR OF PHILOSOPHY
in
(Pharmaceutical Chemistry)
Submitted By
Sucheta Das
Supervised By
Banasthali University
Foremost, I would like to express my deep respect and thank to the almighty for his
grace and blessings and for giving me the strength, will and wisdom to carry out my
project successfully.
I must offer my profoundest gratitude to Prof. Dharam Kishore, Dean and Secretary
Banasthali University, for providing me an opportunity to work and for his kind
cooperation and departmental facilities. I feel blessed in getting his kind guidance in
my life.
My thesis would not have been completed without continuous contribution and
support of my lab mates and friends. I find immense pleasure in stating my cordial
thanks to my colleagues for their love, motivation and all the possible help they
could extend during my research period. I wish to extend my special thanks to Ms.
Aarti Singh Ms. Mukta Sharma & Ms. Khushbu Verma for their consistent
encouragement, constant support and friendly advice.
I am also thankful to Dr. Anupama Mittal, Dr. Anubhuti Pandey, Mrs. Anuradha
Derashri, Mrs.Richa Arya Thakur, Neetika Tripathi & Monika for their kind co-
operation.
Some special words to special persons and close friends like Ms. Vandana Singh
whose dedication, support and love reposed confidence in me to take the load off my
shoulders. I am thankful to her for all those light and cheerful moments I shared
with her.
None of this would have been possible without the love and patience of my loving
parent Mr. Dev Brat Das & Mrs. Karuna Das. They are always with me both in
my dreams and actions. Their life and teachings have taught me that excellence is
continuous journey and has no destination. I hope they will be very happy to see
that the dreams which they nurtured and the developmental inputs which they
provided have started taking the desired shape. In fact, I am short of words for their
contributions in shaping my life as they are the real architects of my life.
No one can ever thrive in life without the blessings of their elders in the family. I
bow my head with great respect before my grandfathers Late Pashupati Das & Late
Ramesh Chandra Das; my uncles Late Mrinal Kanti Das & Late Sakti Ranjan
Karar; my loving aunty Late Gitanjali Das. Though they are not today with me but
they had always been inspiration.
My deepest gratitude goes to my uncle Dr. Shyam Sundar Poddar for his unflagging
support and faith throughout my research work
I can never count the contribution of a special person Mr. Abhijit Naskar for his
unstinted support, whose good wishes and inspiration greatly helped me to complete
out this study with determination.
And at last I would like to thank all who had helped me directly and indirectly for
completing this work.
(Sucheta Das)
Contents
Contents Page no.
List of Tables i-iii
List of Figures iv-vi
Chapter-1
Introduction
Diabetes Mellitus 1-41
Glucagon
Glucagon Receptor
Structure of the Human Glucagon Receptor (GCGR)
Glucagon Receptor Antagonists (GRA)
Peptide Antagonists
Non-peptide Antagonist
Monoclonal Antibodies (MAb)
Glucocorticoids
Cortisol: Major Role Player
11 Beta Hydroxysteroid Dehydogenase (11β HSD)
11 Beta Hydroxysteroid Dehydogenase type I (11β HSDI)
11β HSD1 Inhibitors
Drug Discovery
Drug Discovery Stages
Rational Drug Design
Computer Aided Drug Design(CADD)
Indirect Drug Design
QSAR
History of QSAR
QSAR Methodology
Major Parameters/Descriptors of 2D QSAR
Statistical Methods Used in QSAR Analysis
Evaluation of the QSAR Models
Ligand Based Pharmacophore (LBP) Modeling/ 3D QSAR
Pharmacophore Modeling
HypoGen
Cost Analysis in HypoGen
Validation of the model
Direct Drug Designing
Structure Based pharmacophore (SBP) modeling
Identification of Drug Target
Determination of Target Structure
Identification of Binding Site and Generation of
Pharmacophore
Molecular Docking
Search Algorithm
Scoring
Virtual Screening
Evaluation of Promising Lead Candidate
Chapter-2
Literature Review
Glucagon Receptor Antagonists (GRA) 42-69
11β HSD1 Inhibitors
Problem Envisaged
Proposed Plan of Work
Scheme of Work
Chapter-3
Global Descriptor Based QSAR
Descriptor Based QSAR Approach 70-122
Materials And Methods
Series Selection
Data Set Preparation
Defining Substituents
Descriptor Calculation
Data Reduction
Training and Test Set
Statistical Analysis
Multivariate Linear Regression (MLR) Analysis
Partial Least Square (PLS) Analysis
Forward Feed Neural Network (FFNN Analysis)
Validation of Model
Results and Discussions
Series 1: β alanine, isoserine and thiazole
Series 2: Pyrrolidine Carboxamide Inhibitors of 11β HSD1
Series 3: Bicyclo[2.2.2] Octyltriazole Inhibitors of 11β
HSD1
Chapter-4
Ligand Based Pharmacophore (LBP) Modeling
iii
List of Figures
iv
33 Figure 3.7 Effect of various descriptors on different substituents of GRA
Plot of actual activity versus estimated activity of training and test set
34 Figure 3.8
obtained from MLR analysis
Plot of actual activity versus estimated activity of training and test set
35 Figure 3.9
obtained from PLS analysis
Plot of actual activity versus estimated activity of training and test set
36 Figure 3.10
obtained from NN analysis
Dependency graph illustrating correlation between the Dipole moment X
37 Figure 3.11 component (whole molecule) used to train the neural network
architecture versus the actual activity data
Dependency graph illustrating correlation between the log P (whole
38 Figure 3.12 molecule) used to train the neural network architecture versus the actual
activity data
Effect of various descriptors on the whole molecule of 11β HSD1
39 Figure 3.13
inhibitory activity
Plot of actual activity versus estimated activity of training and test set
40 Figure 3.14
obtained from MLR analysis
Plot of actual activity versus estimated activity of training and test set
41 Figure 3.15
obtained from PLS analysis
Plot of actual activity versus estimated activity of training and test set
42 Figure 3.16
obtained from NN analysis
Dependency graph illustrating correlation between the Inertia moment1
43 Figure 3.17 length (Subst.2) used to train the neural network architecture versus the
actual activity data
Dependency graph illustrating correlation between the log P (whole
44 Figure 3.18 molecule) used to train the neural network architecture versus the actual
activity data
Dependency graph illustrating correlation between the VAMP
45 Figure 3.19 Polarization XX (whole molecule) used to train the neural network
architecture versus the actual activity data
Effect of various descriptors on the whole molecule of 11β HSD1
46 Figure 3.20
inhibitory activity
47 Figure 4.1 Pharmacophore modeling workflow
Plot of correlation coefficient between actual and estimated activity
48 Figure 4.2
value of training set
Pharmacophore features (Hypothesis 1) and pharmacophore mappings of
49 Figure 4.3
GRA
Plot of correlation coefficient between actual and estimated activity
50 Figure 4.4 value test set
v
55 Figure 4.9 Pharmacophore features (Hypothesis 1)
56 Figure 4.10 Pharmacophore mapping with active compounds
57 Figure 4.11 Pharmacophore mapping with inactive compound no. 6
Plot of correlation coefficient between actual and estimated activity
58 Figure 4.12
value of test set
59 Figure 4.13 Graph of 99% Fischer’s randomization test of hypothesis1
Plot of correlation coefficient between actual and estimated activity
60 Figure 4.14
value of external test set
61 Figure 4.15 Pharmacophore mapping of carbenoxolone
Plot of correlation coefficient between actual and estimated activity
62 Figure 4.16
value of training set
Pharmacophore features (Hypothesis 1) and pharmacophore mappings of
63 Figure 4.17
11β HSD1 inhibitor
Plot of correlation coefficient between actual and estimated activity
64 Figure 4.18
value of test set
65 Figure 4.19 Graph of 99% Fischer’s randomization test of hypothesis1
Plot of correlation coefficient between actual and estimated activity
66 Figure 4.20
value of external test set
67 Figure 4.21 Pharmacophore mapping of carbenoxolone
Three dimensional structure of glucagon receptor and 11β HSD1
68 Figure 5.1
enzyme
69 Figure 5.2 The inter-feature distances of the glucagon receptor pharmacophore
70 Figure 5.3 The inter-feature distances of the 11β HSD1 enzyme pharmacophore
71 Figure 6.1 Crystal structure of human glucagon receptor (4L6R)
72 Figure 6.2 Crystal structure of human glucagon receptor (2BEL)
73 Figure 6.3 Binding pattern of NCI compounds SKDGRA1
74 Figure 6.4 Binding pattern of NCI compounds SKDGRA2
75 Figure 6.5 Binding pattern of NCI compounds SKDGRA3
76 Figure 6.6 Binding pattern of NCI compounds SKDHSD1
77 Figure 6.7 Binding pattern of NCI compounds SKDHSD2
78 Figure 6.8 Binding pattern of NCI compounds SKDHSD3
Effect of SKDHSD2 and SKDHSD3 on blood glucose level of
79 Figure 6.9
previously treated STZ induced albino wistar mice
Effect of SKDHSD2 and SKDHSD3 on blood glucose level of STZ
80 Figure 6.10
induced albino wistar mice
vi
CHAPTER- 1
Introduction
CHAPTER-1
INTRODUCTION
Introduction
Diabetes Mellitus
Type II diabetes mellitus has been emerged as one of the most serious and costly
chronic metabolic disorder of the world.4 India in particular has become the leading
country in having the highest number of people suffering with type II diabetes
mellitus.5 The worldwide cases of diabetes are expected to increase by 366 million by
the end of 2030.6 Several studies have been reported which relates increase in type II
diabetes mellitus with lifestyle changes that have resulted in overweight, obesity, and
decreased physical activity levels. These changes in lifestyle affects genetic
predisposition and hence causes increase in insulin resistance and further progress to β
cell mass destruction, rise in glycemia in the non diabetic range.7
1
CHAPTER-1
INTRODUCTION
Glucagon
The production of glucagon is the result of tissue specific post translation of its 160
amino acid precursor, proglucagon (Figure 1.4). Cleavage of proglucagon into
glucagon takes place in the presence of the enzyme prohormone convertase 2 which
also results in the formation of glicentinrelated pancreatic peptide (proglucagon 1-30),
and a major proglucagon fragment (proglucagon 72-158).14
3
CHAPTER-1
INTRODUCTION
Glucagon level in a healthy individual varies with the glucose level in the plasma. It
reaches up to 40 pmol/l during fasting conditions when glucose concentration in the
plasma is reduced to 2-3 mmol/l and when the glucose level is elevated to 10-12
mmol/l, the glucagon level lowers down to 1-2pmol/l.15 Reduced level of glucose
generates an electric promising of Na+ and Ca2+ at pancreatic α- cells. This electrical
activity triggers Ca2+ signals to secrete glucagon. Increased glucose concentration
prevents this pathway.16 Apart from glucose concentration; there are certain amino
acids and some ANS activity which trigger the secretion of glucagon.17 The
glycogenolytic action of glucagon starts with binding of glucagon with glucagon
receptor.
Patient suffering with either type 1 diabetes or type II diabetes show increased plasma
glucagon concentration.18 This elevated plasma glucagon concentration is due to
increased basal hepatic glucose production.19 Thus, hyperglucagonemia is an
important factor of fasting hyperglycemia in diabetes. This effect is more pronounced
in the deficiency of insulin.20 Hence, diabetes is a result of insulin deficiency and
attenuated glucagon signaling.
Glucagon Receptor
Glucagon receptor is a class B GPCR. It is abundant in liver, adipose tissue, pancreas,
the gastrointestinal tract, heart, brain, and kidney.21 Abundance of glucagon receptor
in so many sites suggests its multiple roles beyond glucose homeostasis. Glucagon is
a 62-kDa polypeptide containing 4 N-linked oligosaccharide chains.22 Glucagon binds
to the glucagon receptor (GCGR) and stimulates adenyl cyclase pathway through
heterotrimeric Gs protein.23 The adenyl cyclase pathways raise circulating cAMP
levels and triggers PKA. Activated PKA in the liver is responsible of down streaming
4
CHAPTER-1
INTRODUCTION
Peptide Antagonists
des-His-glucagon, structural analogue of glucagon, lacking only His1 was found to
have in vitro GRA activity, in 1972.29 In 1980, a glucagon analogue identified as [l-N
6
CHAPTER-1
INTRODUCTION
Non-Peptide Antagonist
In 1998 Novo Nordisk developed NNC-92-1687 as the first non-peptide human GRA.
However, it exhibited poor IC 50 of 20 μM.33 Merck came with triarylpyrrole a
promising, orally bioavailable, non-peptide GRA with an IC50 of 3.7 nM.34 Further to
this, Merck formulated substituted imidazoles exhibiting a favorable potency, IC 50
of 53 nM and high selectivity to the glucagon receptor.35 Pfizer came up with a fungal
bisanthroquinone isolated from Talaromyces wortmanni, named as Skyrin as a small
molecule GRA. Skyrin inhibited glucagon activated cAMP formation and
glycogenolysis in hepatic cells.36 An alkylidene hydrazide and its derivative
comprising an alkoxyaryl substitution as antagonists to the human glucagon receptor
were jointly developed by Pfizer, Novo Nordisk, and Anadys Pharmaceuticals.37
Bayer introduced Bay 27-9955 as the first GRA effective to reduce glucagon induced
hepatic glucose output and plasma glucose level elevation in humans. Bay 27-9955 is
a non-peptidyl, orally active, competitive antagonist of the human glucagon receptor
with an IC 50 value of 110 nM.38 Currently, Eli Lilly is engaged in clinical
development of LY 2409021 as a novel orally active GRA.39 Thus, these studies
strengthen the need to develop orally active GRAs as a promising treatment approach
for diabetes.
7
CHAPTER-1
INTRODUCTION
Glucocorticoids
Glucocorticoids are stress hormones that regulate various biological actions involved
in metabolic, inflammatory, cardiovascular and behavioral processes. These are
secreted by the adrenal cortex under the regulation of hypothalamic-pituitary-adrenal
(HPA) axis. They impart their physiological actions in different target tissues by
binding with the glucocorticoid and the mineralcorticoid receptor. They are known as
the end-effector hormones of the hypothalamic-pituitary-adrenal axis.43
8
CHAPTER-1
INTRODUCTION
Glucocorticoids control the energy expenditure of the body by shifting the energy
balance from anabolism to catabolism.44 They stimulate metabolism of glucose in the
liver and facilitate lipolysis in adipose tissues and protein degradation in various
organs and tissues by the process of gluconeogenesis.45 Glucocorticoids also stimulate
appetite in the central nervous system, by facilitating nutrients uptake.46 Thus, chronic
excess of circulating glucocorticoids or increase in the glucocorticoid sensitivity to
their target tissues is connected with development of visceral obesity, T2DM, and
hyperlipidemia.47 A high concentration of glucose and fatty acids affects β cell
sensitivity by triggering stress on endoplasmic reticulum, reactive oxidative species,
degenerated insulin synthesis and abnormal insulin signaling pathways.48
metabolites into active glucocorticoids, i.e. cortisol in the presence of NADPH, while
11β HSD2 reverses the conversion in the presence of NAD (Figure 1.8).59 The
reversible inter conversion of cortisone into cortisol takes place in the endoplasmic
reticulum.60 11β HSD isoenzymes are genetically originated from different genes and
have distinct tissue distributions. 11β HSD1 is expressed in liver, adipose, kidney and
brain and 11β-HSD2 mainly in kidney and salivary glands.61
10
CHAPTER-1
INTRODUCTION
Functionally 11β HSD1 can be divided into four parts. The transmembrane domain,
which enables the enzyme to connect to the endoplasmic reticulum lumen, the
nucleotide binding domain, the catalytic site, and the C- terminal domain helps in the
oligomerisation of the enzyme. Ser170, Tyr183, Lys187 constitute the catalytic site,
and take part in the reaction.64 Increased 11β HSD1 based local cortisol production
raises hepatic glucose output and reduces glucose uptake in adipose tissue and skeletal
muscles. Apart from these, it triggers lipolysis, thus increases the free fatty acids, and
adverse glucocorticoids metabolic effects.65 Insulin inhibits 11β HSD1 expression
which shows dependency of isoenzymes activity on insulin resistance.66 11β HSD1
inhibitors block excess cortisol levels and thus results in less HGP.67
Exogenous natural product includes glycyrrhizic acid, a derivative of licorice, and its
hydrolytic product glycyrrhetinic acid 1 (GA). Licorice is a product of plant
Glycyrrhizia glabra. It is known for its medicinal properties for over a thousand years.
Based on 18β glycyrrhetinic acid, the hemisuccinate carbenoxolon 2 (CBX) was
prepared. Both compounds also promisingly inhibit 11β HSD2.69
Biovitrum was the first to report selective 11ß HSD1 inhibitors.70 Their
arylsulphonamidothiazole compounds efficiently inhibit 11ß HSD1 specifically both
in vitro and in vivo.71 BVT.2733 reduces in vivo plasma glucose and insulin, lowers
hepatic expression of PEPCK and glucose-6-phosphate mRNAs and thus reduces
hepatic glucose production.72
Merck’s compound 544 also found to be selective inhibitor of 11ß HSD1 activity. The
drug reduces body weight and appetite in diet-induced obesity. Analogous to previous
findings, compound 544 is effective in lowering fasting glucose, improving insulin
resistance, glucose tolerance and serum lipids in a type 2 diabetes mouse model.73
Table 1.1 summarizes selectivity and pharmacological effects of different 11β HSD1
inhibitors.
Table 1.1: Selectivity and pharmacological effects of different 11β HSD1 inhibitors
11β HSD1
Selectivity Carbohydrate Metabolism Disadvantage
inhibitors
No penetration to fat tissues
Increased Insulin sensitivity
Carbenoxolone Non selective Requires addition of
Reduced Hepatic gluconeogenesis
amiloride
Increased Insulin sensitivity
Compound 544 Increased glucose tolerance test
Selective No human studies
Merck Reduced fasting glucose
Reduced food intake
Increased Insulin sensitivity
BVT.2733 Reduced Hepatic gluconeogenesis No human studies
Selective
Biovitrum Reduced circulating glucose
Reduced circulating insulin
12
CHAPTER-1
INTRODUCTION
Drug Discovery
Drug discovery is the process of introducing newer and safe drugs to the population in
order to improve the therapeutic value and eliminates the side effects of already
present drugs. Discovering any new drug needs a keen understanding of the disease at
the molecular level. Genomics, proteomics and computational approaches have made
it easier to understand a disease. This understanding leads to rise of “targets,” to
which promising new drugs might be able to act on. This whole process from
understanding the disease to launching a successful drug candidate in the market;
takes an average of 10-15 years. In fact out of 5,000-10,000 compounds entering the
research and development (R&D) pipeline, only one compound succeeds to get
approval as a drug candidate The whole process can cost $800 million to $1 billion.
This cost includes cost of several pitfalls while discovering the drugs.74 Figure 1.11
describes the entire process of drug discovery. The journey of drug discovery does not
end with the introduction of the drug in to the market. The post-market survey and
pharmacovigilance studies may eliminate the drug out of the market.75 Hence, there is
an urge of a careful planning even before the project is underway.
Recent technologies can predict the possible outcomes with the help of computers and
which make this process less risky. This will help in minimizing the efforts in clueless
directions.
13
CHAPTER-1
INTRODUCTION
14
CHAPTER-1
INTRODUCTION
15
CHAPTER-1
INTRODUCTION
Description
Screening Strategies
Large numbers of compounds analyzed in a assay generally designed to run in
High throughput screening
plates of 384 wells and above
Structural aided
Use of crystal structures to design molecules
drug design
16
CHAPTER-1
INTRODUCTION
are responsible for imparting specific property and nature to the drug. Representation
of these specific functional groups along with their interatomic distances is known as
pharmacophore. The functional groups or atoms which interfere with the binding of
the lead compounds with the receptors are known as auxophores. Out of all those
functional groups present in the lead compound some may be interfering with the
binding of the pharmacophore, and such functional groups need to be optimized.
Homologation
A group of compounds generally differing by methylene group is termed as a
homologous series. A small change in the substituent can affect the polarity, alter the
pKa, & change the electronic properties of a molecule. Optimization of homologous
series is one of method used method to induce the optimization in the lead compound.
17
CHAPTER-1
INTRODUCTION
Bioisosterism
Certain substituents or groups having similar physical or chemical properties and
hence similar biological pattern are known as isosters. Isosteric groups have same
number of electrons in their outermost electron shell. Bioisosteric replacement may
affect various physiological actions of the lead compound, for example molecular
size, steric shape, electron distribution, lipid solubility, water solubility, the pKa, the
chemical reactivity to cell compartments, capacity to undergo H- bonding.82
Preclinical Studies
Preclinical study is meant to check the safety profile of the lead compound. A newly
developed molecule has to clear various pharmacological and toxicological tests
based on animal studies before its testing on the human body. Preclinical phase
includes elucidation of the pharmacokinetics and pharmacodynamic profile of the
molecule by depicting its mode of action. However, priority is given to the animal
study data, which roughly estimate the possible adverse reactions and side effects
during treatment. These preclinical studies are performed in two ways, in vitro studies
and in vivo studies. The in vitro studies are carried out on distinct cell-lines and tissue
preparations. The in vivo studies include use of live animals and then changes in the
their behavior is analyzed.83
Clinical Trials
Preclinical studies are followed by the clinical studies, which involve evaluation of
the lead compound in the human volunteers. This stage helps to analyze the toxic
effects in the human body which cannot be analyzed in animals. But, further to this
stage an “Investigational New Drug (IND)” application should be filed in order to get
FDA approval based on the preclinical data. Clinical trials is manly consists of three
stages, stage I, stage II and stage III. There is one more stage 4 study which is carried
out post marketing of the drug.
18
CHAPTER-1
INTRODUCTION
19
CHAPTER-1
INTRODUCTION
20
CHAPTER-1
INTRODUCTION
Accurate
Required for both the ligand and protein Required for ligand only
Structures
21
CHAPTER-1
INTRODUCTION
molecules having some desired biological activity onto the conformation of a lead
compound, with the objective of improved activity or drug like properties.88 A series
of compound having diverse structure and activity is prerequisite feature of the
methodology. For this approach the lowest possible conformers of the rigid
compounds are generated and superimposed on them. Conformations of the flexible
compounds are searched by applying distance constraints of the rigid compounds.
Finally, all the types of compound structures are aligned to develop a pharmacophore
model. This model may be used to identify and synthesize novel compounds with
specific functional groups at specific positions. The most common tool in a LBDD is
2D and 3D QSAR of which the widely used method is CoMFA, HypoGen and
Hiphop.
QSAR
QSAR studies have become a boon for medicinal chemist in modern chemistry and
biochemistry. It speeds up the process of drug designing by saving the money and
time. The basic theory behind QSAR states that there is a connection between the
physical/chemical properties of a compound with their biological activity i.e.
biological activity is a function of structure of the compounds and an equation can be
deduced by relating the biological activity to the physical/chemical parameters.89
These physical/chemical parameters, which relates to hydrophobicity, topology,
electronic properties, and steric effects, are calculated by computer softwares. The
correlation between the chosen parameters and the structure-activity helps in selecting
the promising chemical entities to be synthesized and evaluated in the laboratory.
Activities used in QSAR include in vitro biochemical assays. The most applicable
approach of QSAR is that it helps in designing new chemical entities based on in vivo
situation.
The linear equation for QSAR is depicted below-
BA = Const + (C1 * P1) + (C2 * P2) + (C3 * P3) +...+ (Cn * Pn) Eq. (1)
Where, P1, Pn are the parameters of all the compounds of the series and C1, Cn are
coefficients of the parameters and the biological activity.
22
CHAPTER-1
INTRODUCTION
History of QSAR
Crum-Brown and Fraser were the first to hypothesize the relationship between the
physiological actions of a substance as a function of its chemical composition and
constitution.90 They postulated the first generalized equation suggesting that
physiological activity Φ of a series of quaternized strychnine derivatives depends on
constitution (c), and suggested the following mathematical equation 2.
Φ= f(c) Eq. (2)
In 1893, Richet found inverse correlations between the toxicities of simple
compounds (alcohol, ether) and water solubility.91 In the late 19th century, Meyer and
Overton showed a relationship between the narcotic actions of organic compounds
with their oil/water partition coefficients.92 In 1939, Ferguson published a
thermodynamic equation to show the correlation between depressant actions of
volatile compounds and their relative saturation.93
In the era of physical organic chemistry, Hammett, introduced to the "sigma-rho"
culture, based on the substituent effects on organic reactions.94 Taft introduced the
first steric parameter, Es and developed a technique to isolate polar, steric, and
resonance effects.95 In 1962, Hansch and Muir correlated SAR of plant hormones to
their Hammett constants and hydrophobicity dependency.96 Hansch and Fujita came
up with the Hansch’s equation by combining hydrophobic constants with Hammett's
electronic constants and Taft’s steric constants.97
Log 1/C = aσ + bπ + ck Eq. (3)
23
CHAPTER-1
INTRODUCTION
24
CHAPTER-1
INTRODUCTION
• Single point determinations (i.e. the single occurence of a certain structural feature
in the whole data set) obscure the statistical results;
• Substituents are described by using many degrees of freedom.
Combined Approach100
The two approaches, Hansch and Free-Wilson, are practically interrelated. Free-
Wilson approach suffers by its inability to attribute physical/chemical significance to
the substituent contribution and fails to yield good correlations in the cases where
parabolic model of Hansch is applicable. Likewise, sometimes it is difficult to
correlate the biological activity only in terms of known physical/chemical parameters,
which has necessitated the use of an indicator variables (I) which constitutes a part of
correlation equation so the most commonly used equations now contain a Hansch
parameters are used and of indicator variables based on the assumption of Free-
Wilson as described in equation (9) contains Hansch parameters and Free-Wilson
assumption.
BA = ∑ai, + ∑cj j + constant Eq. 9
The term ai denotes the contribution for each ith substituent, whereas j is any
physicochemical property of a substituent Xj.
QSAR Methodology
Approach 1: Based on Dimensions of the Descriptors/Models
QSAR methods can be classified on the basis of the structural representation or the
way of deriving the descriptor values101:
1. 0D QSAR characterizes molecular formula e.g. molecular weight, number and
type of atoms etc.
2. 1D QSAR only reflects a composition of a compound. This technique includes
correlation of activity with global molecular properties like pKa, log P etc. It
can be used as a tool for independent virtual screening.102
3. 2D QSAR correlates biological activity to the chemical features like
connectivity indices, 2D-pharmacophores etc. These models characterize only
the topology of the molecule. 2D QSAR helps in finding the necessary
features required for the biological activity of the compounds.103
4. 3D QSAR provides full information of structural composition, topology, and
spatial shape of molecule for one conformer only. These models are
25
CHAPTER-1
INTRODUCTION
Electronic Parameters
Electronic features of compounds are closely linked to their chemical and biological
activities. The electronic need of a reaction is decided by its mechanism. The
26
CHAPTER-1
INTRODUCTION
substituent groups involved in the reaction and its effect on the reaction rates helps in
determining the mechanism of reaction. The first and still most widely used electronic
substituent parameter σ was developed by Hammett in 1935 on the basis of ionization
constants for benzoic acid derivatives.109 Hammett’s equation encompasses solution
effects like hydrogen bonding, dipole interactions etc.110 But, Hammet’s equation has
certain pitfalls which are as follows:111
1. The Hammett relationship only applies to systems where the substituents are
attached to the reaction centre via aromatic rings and are situated meta or para.
2. Ortho- substituted aromatic rings cannot be calculated due to steric effects.
3. Aliphatic acids do not follow the equation, because of steric hindrance in the
transition state and their flexibility may affect correlation in negative manner.
4. The substituent constant (σ) for meta- and para-substituents in benzene
derivatives as defined, on the basis of the ionization constant of substituted
benzoic acid in water at 25 oC. ie. X=H.
5. σ is a collective measure of the total electronic effects of a substitutent
[resonance and inductive effects].
6. σ does not depend on the nature of the reaction site.
Where rі is the distance of the ith atom from the origin and qі is the atomic charge of
the ith atom. Total dipole moment is calculated in Debye units. It is often correlated
with the polarity of a compound.113
theory (FMO) of a chemical reaction states that HOMO and LUMO of reactants
interact to form a transition state.115
The HOMO energy is analogous to the ionization promising and correlates with the
likeliness of the electrophilic attack on the molecules. The LUMO energy is
analogous to the electron affinity and correlates with the likeliness of the nucleophilic
attack on the molecules. Both HOMO and the LUMO energies govern radical based
reactions. Strong electrophilic or nucleophilic natures of the molecules are governed
by the relative energies of the HOMO/LUMO orbitals. Strong nucleophiles have a
low HOMO energy and vice versa; strong electrophiles have a high LUMO energy
and vice versa.116
The gap between two orbitals, i.e. the energy difference between the HOMO and
LUMO, is an important indicator of stability. Larger the gap higher is the stability and
hence less reactive is the molecule.
Steric Parameter
Quantitative determination of steric effects is quite an intriguing process, particularly
at the molecular level. However, steric effects are major role player in the drug-
receptor interactions and also in cellular transport systems. Taft’s ES constant is the
first steric parameter of QSAR.117
ES = log(kX/kH)A Eq.(11)
Where, kX and kH are the rates of acid hydrolysis of esters, X=CH2COOR and
CH3COOR
Taft quantified the steric (spatial) effects using the hydrolysis of esters: Here, the size
of R limits the reaction kinetics by obstructing the nucleophilic attack of water.
ES = log kRCO2CH2-log kCH2CO2CH2 Eq. (12)
=log (kRCO2CH2/ kCH2CO2CH2)
In this case, the steric effects are quantified by the Taft parameter Es: k is the rate
constant for ester hydrolysis.
28
CHAPTER-1
INTRODUCTION
solution. The molecular surface area is determined by removing these gaps and
cervices.118
Verloop
Verloop parameters are comprised of multi-dimensional steric descriptors defined in a
box for characterizing the shape and volume of the substituent. The shape and volume
of a substituent plays a critical role in deciding the steric effects produced by the
substituent in the ligand receptor interaction. The Verloop parameters B1-B5
represent the width of the substituent in the direction perpendicular to the length of
the substituent.119
Larger the molecular weight of the compound, larger is the steric effect, while larger
is the density, the smaller will be the steric. A smaller molar refrativity for a similar
molecular weight indicates stronger interactions in the crystal and hence stronger
dispersion interactions with the receptor.120
Hydrophobic Parameter
The absorption and distribution of any compound inside the biological systems is
determined by its hydrophilic or hydrophobic properties, which are represented by
partition coefficient (P). It is defined by:
P= [drug]octanol Eq. (14)
[drug]water
29
CHAPTER-1
INTRODUCTION
Log P
Log P is hydrophobic parameter. It explains the effect of the substituents on the
hydrophobicity of the main compound.122 To understand the effect of relative
hydrophobicity of substituents the hydrophobicity substituent constant, p was
introduced. The parameter p has been defined analogous to the Hammett parameter
Topological Indices
Topological indices are techniques to convert chemical constitution of the compounds
into numerical values which further can be correlated with physical/chemical
properties and for QSAR studies. It represents the compounds in molecular graphs,
where the atoms are denoted by dots (vertices) linked to each other by lines (edges),
which are chemical bonds.124 With the help of this molecular graphs, certain path
lengths can be determined. A specific path length (m) is the distance between two
atoms connected with m bonds.
Topological indices are easy to calculate on the molecular two dimensional structures
only, thus ditching the need of conformational analysis of the compounds.
Topological indices suffer the drawback in their complexity in interpretation.
Wiener Number
Wiener number was introduced in 1947. It is so named after the inventor Harry
Wiener (1924–1998). His life and his achievements first in chemistry and later in
medicine have been well–presented by Rouvray.125
Wiener number is the sum of all the distances between the carbon atoms. The smaller
this number, the larger is the compactness of the molecule. It can be calculated by
multiplying the number of carbon atoms on one side of any bond by those on the other
side; W is the sum of these two values for all bonds. W can also be obtained by
simply adding all the elements of the graph distance matrix above the main diagonal.
30
CHAPTER-1
INTRODUCTION
Balaban
The Balaban index was proposed by A. T. Balaban which also called the average
distance-sum connectivity or J index. It appears to be a very useful molecular
descriptor with attractive properties.126
The Balaban index is a graph index defined for a graph G with vertex-set V (G) and
edge-set E(G), dG(u, v) denotes the distance between vertices u and v in G, and
DG(u) = Σ dG(u, v) is the distance sum of vertex u in G, i. e., the row
vεV(G) Eq. (16)
sum of distance matrix of G corresponding to u. The Balaban index of G is defined as
J(G) =m/μ + 1 Σ 1/√DG (u)DG(v) Eq. (17)
uvεE(G)
Randic
Randic introduced molecular connectivity Randic index (c) to characterize the
molecular branching. Its quantitation is based on the degree Di of the vertex i in the
hydrogen-suppressed molecular graph. The Di is number of bonds from the atom
(vertex) i to non-hydrogen atoms.
Connectivity Indices
The connectivity index order k is derived from the adjacency matrix and is normally
written as, kχt, The order k is between 0 and 4 and is the number of connected non-
hydrogen atoms which appear in a given sub-structure.127
k
nt
k
χt= Σ 1/2
[Пδi ]- Eq. (18)
j-1 iεSj
In above equation, δi is the number of simple (i.e., sigma) bonds of the atom i to on
hydrogen atoms, Sj represents the jth sub-structure of order k and type t, and knt is the
total number of sub-graphs of order k and type t that can be identified in the molecular
structure. The types used are path (p), cluster (c), and path-cluster (pc).
31
CHAPTER-1
INTRODUCTION
where the summation is over the remaining atoms in the molecule. The intrinsic state
(Ii) and the perturbation (∆Iij) terms are calculated as follows:
where Ni is the principal quantum number of valence electrons (the row, at which the
chemical element is placed in the periodic table)
Where, rij is the topological distance between atoms i and j, equal to the number of
atoms in the shortest path between them.
The quantification of E-state indices for atoms (such as >C<, >N–, =O, –Cl) in a
molecule, and for every hydride group (such as –CH3, >NH, –OH) is possible. The
32
CHAPTER-1
INTRODUCTION
atom-type E-state indices have recently been introduced as an extension of the E-state
indices. These indices are the integration of the E-state values of each atom type. E-
state indices calculation of hydrogen atoms for deriving hydrogen atom-type is also
possible. Atom-type E-state indices determine the nature of electron accessibility for
the atoms of same types. Thus, the E-state values describe the feasibility of a
molecule to take part in a non-covalent intermolecular interaction.
procedure effective when the numbers of descriptors are more than the number of
compounds modeled. Normally, it is calculated by the leave one out cross
validation procedure.
34
CHAPTER-1
INTRODUCTION
quality of fit and the number of degrees of freedom. The smaller the value of SD the
better is the QSAR.
DF=n-k-1. Eq. (25)
2
SD= (yobs –y cal) / n-k-1 Eq. (26)
4. F-value: It calculates the statistical significance of the regression model, the
influence of the number of variables included in the model is larger than the standard
deviation.
F-value= r2 (n-k-1)/k (1-r2) Eq. (27)
5. Predictive residual sum of squares (PRESS): The sum of overall compounds, of
the square difference between the actual and the predicted value dependent variables.
P=Σ(yobs-y calc) 2 Eq. (28)
6. Outliers: An outlier is defined as a data point with a residual greater than two
times the standard deviation of residual.
7. Cross Validated r2 (q2): This is squared correlation coefficient generated using the
equation during a validation procedure.
r2Cv = SD-PRESS/SD Eq. (29)
Where SD is standard deviation
A cross-validated r2 is smaller than the overall r2. It is used to evaluate the
predictibility of an equation.
35
CHAPTER-1
INTRODUCTION
HypoGen
The HypoGen is similar to HipHop in using the same features but is a quantitative
hypothesis. The pharmacophore generation by HypoGen is divided into three
stages.137
• Constructive Stage constitutes generation of pharmacophore models in which the
most active compounds fit.
• Subtractive Stage removes the pharmacophore models common to the inactive
compounds.
• Optimization Stage randomly modifies the generated pharmacophore models by
moving features, rotating vectors, or adding or removing features. The best
modification having the lowest error cost values are kept and finalized as best
hypothesis. The cost values are the number of binary bits to generate
pharmacophore hypotheses. Lower the cost values, stable the hypothesis.
36
CHAPTER-1
INTRODUCTION
Error Cost138
The error cost is directly linked to the RMS difference. It increases with the increase
in root mean-square (RMS) difference between the estimated and the actual affinity
for the training set. It calculates the quality of prediction of the model.
Weight Cost138
The weight cost relates to the actual and ideal weight of the features. It is directly
proportional to the difference between the actual and ideal weights of the features.
The ideal value of the weight is 2. Weight values more than 2 causes fitting of
unrealistic conformations of the compounds to the features.
The error and the weight components are estimated using the probability distribution
shown in the equation 30
Eq. (30)
The error and weight components of the hypothesis cost also be estimated by taking -
lnP of as shown in eq. 31
Eq. (31)
Where, sigma and delta are constant parameters derived from expected values of
weights and errors, and x is the deviation from the expected values for each. The
sigma values are log (uncertainty) and 0.302 for the error and weight components,
respectively. The delta values are log (1.1) and 0.2 for the error and weight
components. The deviation for the errors is the actual minus the predicted scaled
activity values for the training set members, and the deviation for the weights is the
difference between the average of the feature weights and the ideal weight.
Configuration Cost138
The configuration cost is a fixed cost representing the complexity of the hypothesis
space to be optimized. It is expressed as log2 ‘P’, where P is the number of hypothesis
generated during the constructive stage and existed in the subtractive stage. The cost
value should not cross the value of 17. A configuration value of more than 17 leads to
a chance correlation as Catalyst software has the ability to form 217 hypotheses only.
37
CHAPTER-1
INTRODUCTION
Where, e, w, and c are the coefficients associated with the error (E), weight (W), and
configuration (C) components, respectively.
Fixed Cost138
The fixed cost represents the ideal model fitting all the data perfectly i.e. all
compounds lay on a single line of slope. It is a sum of lowest possible error cost and
weight cost and the constant configuration cost
38
CHAPTER-1
INTRODUCTION
39
CHAPTER-1
INTRODUCTION
Molecular Docking
The fate of a successful drug candidate depends on its interaction with suitable
receptor at a suitable orientation. Molecular docking is a technique which determines
40
CHAPTER-1
INTRODUCTION
the possibility of binding of ligand to the receptor and also evaluates the proper
orientation of ligand receptor complexes during binding.144 Molecular docking
comprises of two basic steps: search algorithm and scoring.
Search Algorithm
A search algorithm searches all the possible binding poses for the drug-receptor
complex. This phenomenon includes exploration of all the degree of freedoms for the
ligand, which creates all the possible conformers for the complex.145 Lowest energy
conformers are considered as best and presented as binding poses.146 There are
numerous algorithms used for the process; Monte Carlo simulations and Genetic
algorithms are the most commonly used.
Scoring
Evaluation of the binding poses is done on the basis of the computed ligand–receptor
interaction energy known as docking score of the pose.147 The calculation of this score
is based on energy functions such as energy of interaction (pKd), electrostatics, Van
der Waal’s interactions, hydrogen bonds, solvation effects, loss of entropy, active site
waters. Score represents the number given to the goodness or the energy of the
conformer.
Virtual screening
Identification of novel and promising leads is one of the approaches of drug discovery
and designing process. Pharmacophore models are used to search the compound
databases and library on the basis of perfect fit. The compounds perfectly fitted onto
the pharmacophore model are developed or optimized further as a lead compound.
Two search methods Fast or Best flexible methods are used to retrieve known
compounds having unidentified biological activities.
41
CHAPTER-2
Literature Review
CHAPTER-2
LITERATURE REVIEW
Overview
Diabetes comprises of a group of disorders having the phenotype of hyperglycemia.
Insulin is the key player and a number of insulin products are available in the market
but majority of patients are still fighting to bring the normal glucose level in the
plasma. This demands search for some biological targets that would shift the focus of
diabetes treatment from insulin dependency. The role of glucagon in the
pathophysiology of diabetes has drawn attention towards glucagon as a rational
therapeutic target. Attempts have been done to develop clinically usefull GRA.
Ability of 11β HSD1enzyme to convert inactive cortisone to active cortisol, which has
a major role in mobilization of energy in the form of glucose in the liver, makes it a
promising therapeutic approach to treat type 2 diabetes induced hyperglycemia.
Inhibitors of 11β HSD1 block the synthesis of local cortisol which results in less
hepatic glucose production.
Coy, DH and others (1984), used solid-phase methodology, preparative medium and
high performance reverse phase liquid chromatography to synthesize glucagon and its
Arg12 analogue in approximately 5% yields.149
Hruby, VJ and others (1986), suggested that hepatic cells comprises of two distinct
glucagon receptors, GR-1 receptor which triggers inositol phospholipid breakdown
and GR-2 receptor which triggers adenylate cyclase activity.151
Hruby, VJ and others (1987), synthesized two new glucagon analogues having
antagonistis activities using solid-phase methodology. These two new analogues,
42
CHAPTER-2
LITERATURE REVIEW
Hruby, VJ and others (1995), synthesized a new glucagon analog [des His1, des
Phe6, Glu9] glucagon amide with IC50 value of 48 nM.155
Azizeh, BY and others (1997), designed, and synthesized five new glucagon
analogues, having excellent antagonistic properties.157
Hruby, VJ and others (1997), designed and synthesized fifteen glucagon analogues
by modifying N-terminal region of glucagon, in particular histidine-1, phenylalanine-
6, and aspartic acid-9. Modifications were done to study the effect of phenylalanine at
6th position on the glucagon action.158
Hruby, VJ and others (1998), synthesized eight glucagon analogs. The compounds
were reported to have full or partial adenyl cyclase agonistic activity.160
Madsen, P and others (1998), synthesized the first non peptide competitive human
GRA, 2-(benzimidazol-2-ylthio)-1-(3,4-dihydroxyphenyl)-1-ethanone, NNC 92-1687
with binding affinity of 20 μM (IC50) and Ki = 9.1 μM.33
43
CHAPTER-2
LITERATURE REVIEW
Hruby, VJ and others (2000), designed and synthesized six cyclic glucagon
analogues, c[Asp9, Lys12][Lys17,18, Glu21] glucagon-NH2, c[Asp9, Lys12] glucagon-
NH2, c[Lys12, Asp15] glucagon-NH2, c[Asp15, Lys18] glucagon-NH2, [Lys17-c[Lys18,
Glu21] glucagon-NH2, and c[Lys12, Asp21] glucagon-NH2.163
Sivarajah, P and others (2001), cloned glucagon-like receptors from two species of
frogs to understand the biology of proglucagon-derived glucagon-like sequence.164
Smith, AR and others (2002), established a narrow SAR for the 4-aryl group in 4-
aryl-pyridine glucagon antagonists.169
44
CHAPTER-2
LITERATURE REVIEW
Ladouceur, GH and others (2002), evaluated the SAR of a new class of 4-aryl-
pyridine glucagon receptor antagonists bearing the hydroxyl moiety.171
Madsen, P and others (2002), modified the core (proximal) dimethoxyphenyl group,
the benzyl ether linkage, as well as the (distal) benzylic aryl group of the lead 2, 3-
cyano-4-hydroxybenzoic acid (3,5-dimethoxy-4-isopropylbenzyloxybenzylidene)-
hydrazide to generate a promising human glucagon receptor (hGluR) antagonists.172
Unson, CG and others (2002), reported that for high affinity binding of any ligand
with the glucagon receptor multiple contacts with the residues of N-terminal tail and
first extracellular loop domain of the receptor are required.173
Sapse, AM and others (2003), performed ab initio calculations on the two series of
non-peptide glucagon receptor antagonists (pyrrolo[1,2-a] quinoxalines and
mercaptobenzimidazoles). The results suggested that substituted phenyl ring in the
pyrrolo[1,2-a]quinoxaline series does not affect binding affinity efficiently.174
Hruby, VJ and others (2003), determined the solution structure of the promising
glucagon antagonist, [desHis1, desPhe6, Glu9] glucagon amide by homonuclear 2D
NMR spectroscopy at pH 6.0 and 37 °C in perdeuterated dodecylphosphocholine
micelles.175
45
CHAPTER-2
LITERATURE REVIEW
Jiang, G and others (2004), designed an in vivo glucagon receptor binding assay
125
using I labeled glucagon as a radiotracer. The results revealed that glucagon
receptor is chiefly located in liver, and to some extent in kidney; and peptidyl and
non-peptidyl GRAs bind to the hepatic glucagon receptor in vivo.177
Hsu, WH and others (2004), investigated the function of arginine vasopressin (AVP)
on glucagon secretion in both normal and diabetic rats.178
Potterat, O and others (2004), isolated a new bicyclic 19-peptide, BI-32169 (Figure
2.2), from the culture broth of Streptomyces sp. (DSM 14996), which showed human
glucagon receptor antagonistic activity (IC 50 440 and 320 nM, respectively) in a
functional cell-based assay.179
Duffy, JL and others (2005), discovered a novel human GRA (Figure 2.3), with high
activity (IC 50 = 34 nM) and moderate bioavailability (36% in mice).181
46
CHAPTER-2
LITERATURE REVIEW
47
CHAPTER-2
LITERATURE REVIEW
Mortensen, OH and others (2007), characterized the promoter regions of the human
glucagon receptor gene.186
Lau, J and others (2007), optimized a weak human GRA with an IC50 of 7 μM to a
promising glucagon antagonist. . The GRA efficiently reduced plasma glucose levels
in a diabetic animal model and exhibited oral bioavailability in animals.187
Kim, RM and others (2008), described the discovery and optimization of promising
and selective aminobenzimidazole based GRAs.188
Kodra, JT and others (2008), optimized a series of small molecule human GRAs by
counter screening against the human GIT inhibitory polypeptide receptor. Through
SAR studies, they were successful in finding compounds with nanomolar affinities
against hGluR, selective hGIPR and human glucagon-like peptide-1 receptor (hGLP-
1R).189
Madsen, P and others (2009), designed and synthesized compounds with cyclic
cores (5-aminothiazoles) as promising human GRAs with advanced pharmacokinetic
(PK) properties as a therapeutic approach towards type 2 diabetes.190
Zhuo JL and others (2009), reviewed the role of glucagon and its receptor signaling
in the pathogenesis of Type 2 diabetic renal injury, and explored the possibility of
targeting glucagon receptor signaling as a anti diabetic strategy in Type 2 diabetes.191
Marahiel, MA and others (2010), performed tandem mass spectrometric and nuclear
magnetic resonance spectroscopic studies on the GRA BI-32169. The studies revealed
that BI-32169 is a lasso-structured peptide constituting the new class III of lasso
peptides.192
Rafferty, EP and others (2010), examined the efficacy and stability of two novel
glucagon antagonist peptides in vitro and their duration of action in vivo in normal
mice.193
48
CHAPTER-2
LITERATURE REVIEW
Filipski, KJ and others (2012), discovered a novel series of pyrazole ethers and
aminopyrazoles as GRAs.196
Kieffer, TJ and others (2012), reviewed the biology of GIP, GLP-1 and glucagon
and examined the different therapeutic strategies to activate and antagonize the
receptors of these peptides.197
Xiong, Y and others (2012), discovered a promising, selective GRA 9m, N-[(4-
{(1S)-1-[3-(3,5-dichlorophenyl)-5-(6-methoxynaphthalen-2-yl)-1H-pyrazol-1-
yl]ethyl}phenyl)-carbonyl]-β-alanine.198
Irwin, N and others (2013), evaluated two novel glucagon analogues, desHis1Glu9-
glucagon-[mPEG] and desHis1Glu9(Lys30PAL)-glucagon for GRA properties.201
O’Harte, FPM and others (2013), tested in vitro and in vivo biological activities of
four novel structural analogues of glucagon, desHis1Pro4-glucagon, desHis1Pro4Glu9-
glucagon, desHis1Pro4Glu9Lys12FA-glucagon and desHis1Pro4-Glu9Lys30FA-
glucagon.202
Siu, FY and others (2014), reported the crystal structure of the seven transmembrane
helical domain of human glucagon receptor at 3.4 A˚ resolutions, complemented by
extensive site-specific mutagenesis, and a hybrid model of glucagon bound to
49
CHAPTER-2
LITERATURE REVIEW
glucagon receptor to understand the molecular recognition of the receptor for its
native ligand.27
Alberts, P and others (2003), evaluated the 11β HSD1 inhibitory effects on plasma
glucose levels, glucose tolerance and insulin sensitivity in type 2 diabetes mouse
models.205
Tomlinson, JW and others (2005), described the role of 11β HSD1 as an enzyme
that liberates active glucocorticoid, cortisol, within liver and adipose tissue.207
Xiang, J and others (2005), described the unanticipated discovery of β-keto sulfones
as promising 11β HSD1 inhibitors.210
50
CHAPTER-2
LITERATURE REVIEW
Richards, S and others (2006), discovered that dichloroaniline amide are novel and
potent 11β HSD1 inhibitors.213
Yeh, VSC and others (2006), synthesized and biologically evaluated a series of
metabolically stable butyrolactam as 11β HSD1 inhibitors.214
Yeh, VSC and others (2006), synthesized and biologically evaluated a series of
metabolically stable adamantane amide as 11β HSD1 inhibitors.215
Fujisawa, Y and others (2007), investigated the cause of metabolic disorders in the
infants of diabetic mothers and proposed that 11β HSD1 might be a key player behind
this.217
Potter, BVL and others (2007), discovered and synthesized three structurally
different series of novel 11β HSD1 inhibitors that inhibit human 11β HSD1 in the
micromolar range.218
Potter, BVL and others (2007), reported the discovery and synthesis of some 18β-
glycyrrhetinic acid (18β-GA) derivatives and their inhibitory activities against rat
hepatic11β HSD1 and rat renal 11β-HSD2.219
Patel, JR and others (2007), discovered a novel class of adamantane ethers as 11β
HSD1 inhibitors.220
51
CHAPTER-2
LITERATURE REVIEW
Uckaya, G and others (2008), determined 11β HSD1 mRNA expression levels in
adipose tissues in diabetic patients. The down regulation of 11β HSD1 activity
suggested its contribution to the pathogenesis of type 2 diabetes mellitus.225
Iwasaki, Y and others (2008), examined the effects of multiple humoral factors
related to the metabolic syndrome on the transcriptional activity of 11β HSD1 gene in
hepatocytes in vitro.226
O
N
N S
H
OH
F
Xiang, J and others (2008), synthesized, in vivo evaluation and performed structure-
activity relationship, of piperazine sulfonamides as 11β HSD1 inhibitors.230
52
CHAPTER-2
LITERATURE REVIEW
Kang, NS and others (2008), performed 3D-QSAR CoMFA and CoMSIA studies on
70 11β HSD1 inhibitors, based on molecular docking conformations obtained by
using FlexX-Pharm.231
Shen, J and others (2008), discovered novel inhibitors of 11β HSD1 by using
docking and pharmacophore modeling. Several compounds, with large structural
diversity and good potency against 11β HSD1, were found and their potency was
determined by the enzyme assay. New scaffolds of 11β HSD1 inhibitors were also
reported.236
Wang, H and others (2008), identified several series of pyridine amides as selective
and promising 11β HSD1 inhibitors.238
Gumy, C and others (2009), evaluated six traditional anti diabetic medicinal plants
extracts for inhibition of 11β HSD1 activity and glucocorticoid receptor (GR)
activation in transfected HEK-293 cells. Leave extracts of Eriobotrya japonica
inhibited 11β HSD1 over 11β-HSD2. Extracts of roasted but not native coffee beans
53
CHAPTER-2
LITERATURE REVIEW
Potter, BVL and others (2009), reported the discovery of a series of novel
adamantyl carboxamides as selective inhibitors of human 11β HSD1 in HEK-293
cells transfected with the 11β HSD1 gene.241
Srivastava, RAK and others (2009), reported that fenofibrate down regulated
expression of hepatic and adipose 11β HSD1 gene, which contributed in attenuating
diabetic state.243
Stewart, PM and others (2009), reported the contribution, of 11β HSD in generation
of cortisol from liver.244
Odermatt, A and others (2009), evaluated the impact of the NADPH/NADP+ ratio
and the extracellular glucose on 11β HSD1 activity by applying microsomal
preparations and intact HEK-293 cells expressing 11β HSD1 in the presence or
absence of hexose-6-phosphate dehydrogenase (H6PDH). Measurements in intact
cells suggested that the ER-luminal NADPH concentration is highly sensitive to
fluctuating extracellular glucose levels.246
54
CHAPTER-2
LITERATURE REVIEW
Lloyd, DJ and others (2009), examined the in vivo effects of a specific 11β HSD1
inhibitor on atherosclerosis while assessing glucose homeostasis. The results
suggested that 11β HSD1 inhibition leads to improved glucose metabolism.247
Sun, D and others (2009), described the synthesis and SAR of a series of
arylsulfonylpiperazine inhibitors of 11β HSD1.249
Karlsson, C and others (2010), investigated the relationship between 11β HSD1
mRNA levels in adipose tissue and in skeletal muscle and anthropometric and
metabolic measurements in humans with and without impaired glucose
homeostasis.256
55
CHAPTER-2
LITERATURE REVIEW
Stewart, PM and others (2010), reviewed the role of 11β HSD1 in the pathogenesis
of obesity, type II diabetes and the metabolic syndrome.258
Huber, R and others (2010), evaluated the efficacy and safety of the 11β HSD1
inhibitor INCB13739, in combination therapy with metformin in patients with type 2
diabetes exhibiting inadequate glycemic control (A1c 7-11%).259
Edgerton, DS and others (2010), investigated the effect of acute 11β HSD1
inhibition on HGP and fat metabolism during insulin deficiency in conscious dogs.260
Ge, R and others (2010), reported novel compounds which inhibits 11β HSD1
reductase activity by increasing its oxidase activity without inhibition of 11β-HSD2.
They proposed dual modulation on 11β HSD1 as a better therapeutic strategy for type
II diabetes.262
Yan, X and others (2010), reported a novel series of diarylsulfones as human 11β
HSD1 inhibitors.263
Shah, U and others (2010), reported the discovery of novel azabicyclic sulfonamide
based 11β HSD1 inhibitors.264
Cheng, H and others (2010), reported the design and development of a series of
highly selective pyrrolidine carboxamide 11β HSD1 inhibitors.265
56
CHAPTER-2
LITERATURE REVIEW
Ahn, JH and others (2010), synthesized and evaluated a new series of cyclic
sulfonamide derivatives for their ability to inhibit 11β HSD1.267
Ye, XY and others (2011), designed and synthesized a series of polycyclic acids as
novel, promising, and selective inhibitors of human 11β HSD1.269
Liu, Y and others (2011), observed that 11β HSD1 expression in livers increases
under non-fasting state. They suggested that insulin is a regulator of 11β HSD1
expression and inhibition of this enzyme can be an effective therapeutic approach.270
Simunkova, K and others (2011), reported the cortisone acetate test in 12 young
adult type 1 diabetic patients. The results showed adaptive changes of 11β HSD
activity associated with altered cortisol tissue supply.271
Tice, CM and others (2011), obtained hit 1,3-oxazinan-2-one (9a) from structure
based design with an in vitro IC 50 of 42 nM against 11β HSD1. Optimization of 9a
for improved in vitro enzymatic and cellular potency yielded 25f (Figure 2.6) with IC
50 values of 0.8 nM for the enzyme and 2.5 nM in adipocytes.272
57
CHAPTER-2
LITERATURE REVIEW
Kim, KY and others (2011), investigated the anti diabetic and anti adipogenic effects
of 2-(3-benzoyl)-4-hydroxy-1,1-dioxo-2H-1,2-benzothiazine-2-yl-1-phenylethanone
(KR-66344) as 11β HSD1 inhibitor.273
Sun, D and others (2011), described the synthesis and SAR studies of 4,4-
disubstituted cyclohexylbenzamide as 11β HSD1inhibitor.274
Shen, J and others (2011), reported discovery of seven hit compounds as novel 11β
HSD1 inhibitors from in silico screening of the the commercially available Maybridge
database. They further optimized the lead compounds to have two promising
compounds for further development.275
Maletic, M and others (2011), reported the discovery of a promising ethyl sulfone as
11β HSD1 inhibitors.277
Venier, O and others (2011), identified a novel class of ureas as 11β HSD1
inhibitors by high throughput screening.278
Ahn, JH and others (2011), synthesized and evaluated a new series of thiazolidine
derivatives with an adamantyl group for their ability to inhibit 11β HSD1.280
Xiong, B and others (2012), designed and synthesized a novel class of 1-arylsulfonyl
piperidine-3-carboxamides derivatives as human 11β HSD1 selective inhibitor.282
Maser, E and others (2012), showed that M. charantia extract capsules have
selective 11β HSD1 inhibitory activity.283
58
CHAPTER-2
LITERATURE REVIEW
Wan, ZK and others (2012), described the SAR studies of a series of piperazine
sulfonamide-based 11β HSD1 inhibitors. (R)-3,3,3-Trifluoro-2-(5-(((R)-4-(4-fluoro2-
(trifluoromethyl)phenyl)-2-methylpiperazin-1-yl)sulfonyl)thiophen-2-yl)-2-
hydroxypropanamide 18a (HSD-621) was identified as a promising and selective 11β
HSD1 inhibitor.288
Kim, KY and others (2012), investigated the anti diabetic and anti adipogenic effects
of 4-(2-(1,1-dioxido-6-(2,4,6-trichlorophenyl)-1,2,6-thiadiazinan-2-yl)acetamido)
adamantane-1-carboxamide (KR-67183), a novel selective 11β HSD1 inhibitor.289
59
CHAPTER-2
LITERATURE REVIEW
Shen, J and others (2012), reported the design, synthesis and biological evaluation of
novel amide and urea as 11β HSD1 inhibitors.294
Anagnostis, P and others (2013), reviewed the role of 11β HSD1 inhibition in the
treatment of metabolic disorders and provided rationale for the development of 11β
HSD1 inhibitors in diabetes.296
Harno, E and others (2013), hypothesized that increasing the substrate for 11β
HSD1 (11-dehydrocorticosterone, 11-DHC) would adversely affect metabolic
parameters. They analyzed that chronic administration of 11- dehydrocorticosterone
to male C57BL/6J mice results in increased circulating glucocorticoids, and down-
regulation of the hypothalamic-pituitary-adrenal axis.298
60
CHAPTER-2
LITERATURE REVIEW
Odermatt, A and others (2013), discovered a novel class of 11β HSD1 inhibitors
bearing an arylsulfonylpiperazine scaffold using molecular modeling approach and
classical bioisosteric studies.303
Akiyama, N and others (2014), elucidated adipose dysfunction via 11β HSD1
activation in the development of obesity-related diabetes.307
61
CHAPTER-2
LITERATURE REVIEW
Problem Envisaged
Diabetes is a chronic glucose metabolic disorder affecting millions of people
worldwide. The number of affected population is continuously increasing. Various
therapies have been exercised for the treatment of type 2 diabetes induced
hyperglycemia, but tight glycemic control is still a challenge. Hence there is a
pressing need for additional therapeutic agents which could give a fight to this life
threatening metabolic disorder. In earlier days researchers completely focused on
insulin supplement or enhancing β cell mass; but type 2 diabetes mellitus is not just
insulin impairment. It is actually caused due to imbalanced insulin-glucagon action.
Insulin is responsible for conversion of plasma glucose into its stored form, glycogen
whereas glucagon acts contrary to insulin by converting glycogen back into free
glucose. If insulin impairment is responsible for non conversion of free glucose into
glycogen, glucagon worsens the situation by converting back already stored glycogen
into free glucose, which leads to hyperglycemia. Relative insulin deficiency with
glucagon excess apparently bestows to afflicted glucose production and
hyperglycemia. Thus, targeting reducing glucagon signaling can be a promising
therapeutic approach for treatment of type 2 diabetes mellitus. Up to now, three GRAs
have been reported to be effective in human subjects. The first attempt of glucagon
receptor blockade to reduce the glucose output was achieved with (+)-BAY 27-9955.
Later on, MK0893 were disclosed as a promising GRA in preclinical and human
clinical trial results. Similar to (+)-BAY 27-9955 and MK-0893, LY2409021 was also
reported to block exogenous glucagon-stimulated glucose production in humans. (+)-
BAY 27-9955 and MK-0893 succeeded in short term human studies by safely
blocking exogenous glucagon actions, however clinical development for these
compounds were not pursued and LY2409021 is currently in phase II clinical trial.
Similarly, there are several other biochemical processes in the human body which
increases the plasma glucose concentration by metabolizing other nutrients into
glucose. Gluconeogenesis is a similar process in which glucocorticoid circulates
glucose by accelerating the transcription of rate limiting gluconeogenesis and
glycolysis enzymes.310 Cortisol is the centrally acting glucocorticoid and is the main
player of gluconeogenesis. The activity of cortisol is regulated by 11β HSD isozymes.
11β HSD1 converts inactive cortisone to active cortisol, whereas 11β HSD2 converts
62
CHAPTER-2
LITERATURE REVIEW
cortisol back into cortisone. Hence, inhibiting 11β HSD1 can be a potential target to
reduce hyperglycemia. 11β HSD1 inhibitors block the circulating concentration of
glucocorticoid and thus interfere with the process of gluconeogenesis and hence,
result in less hepatic glucose production (HGP). Liver selective 11β HSD1 inhibitors
actions include decreased hepatic glucose production, enhanced insulin sensitivity,
retardation of progressive β cell dysfunction, and lower risk of hypoglycemia when
compared to sulfonylurea or insulin treatment. Several attempts have been made to
synthesize a promising 11β HSD1 inhibitor; but till now only carbenoxolone has
exhibited some efficacy in humans. But, carbenoxolone also found to inhibit 11β
HSD2 and also have some side effects like hepatic ulcers.
Thus, there is a compelling need to identify a promising GRA and a promising 11β
HSD1 inhibitor that can be considered as alternative clinical development candidates
for the treatment of type 2 diabetes.
Work Plan
The utility of virtual screening in lead identification is unquestionable and the success
stories can be traced in previous literature. In view of this we have made an effort to
identify novel 11 βHSD1 inhibitors and GRA through sequential use of ligand based
pharmacophore modeling, virtual screening, molecular docking and biological
evaluation protocols.
The specific objectives of the present research work:
1. To develop QSAR based models that establish the correlation among
physical-chemical properties of compounds with the biological activity of
GRA and 11β HSD1 inhibitor by using TSAR 3D version 3.3 software.
2. To generate pharmacophoric features essential for ligand receptor/enzyme
interaction.
3. To elucidate important amino acid involved in ligand receptor/enzyme
interaction.
4. To identify new promising GRA and 11β HSD1 inhibitor from chemical
compound databases.
5. Biological evaluation of compounds retrieved from databases.
63
CHAPTER-2
LITERATURE REVIEW
Scheme of Work
Scheme 1: Descriptor based QSAR (Hansch analysis)
64
CHAPTER-2
LITERATURE REVIEW
4. Ring-Aromatic
Hypogen
Algorithmm
Test set Training set Pharmacophore generation
No
Statistically relevant ?
Yes
Best model obtained Model validated ?
65
CHAPTER-2
LITERATURE REVIEW
66
CHAPTER-2
LITERATURE REVIEW
67
CHAPTER-2
LITERATURE REVIEW
68
CHAPTER-2
LITERATURE REVIEW
69
CHAPTER-3
Global Descriptor
Based QSAR
CHAPTER-3
calculates molecular energies by summing bond length, bond angle, torsion angle,
Van der Waals and coulombic terms for all appropriate sets of atoms.. Charges were
evaluated by charge 2 option.312
Defining Substituents
Since substituents, i.e., atoms or group of atoms present at different positions of the
parent structure affect the biological activity, the substitutions of each series were
defined using “define substituent option available within TSAR”.
1. Series 1 comprised of three classes of compounds i.e. β alanine, isoserine and
thiazole. In TSAR package, it is necessary to have same nucleus and same
number of substitution for all the compounds. Out of 70 compounds, 66 had
common substructure of 4-(aminomethyl)-N-methylbenzamide, with three
substituents which were defined (Table 3.1). Four compounds, a61, c24, c27
and c28 were excluded from the series.
2. Series 2 comprised of 30 pyrrolidine carboxamide derivatives as 11β
HSD1inhibitors. 28 compounds appeared with common substructure of 2-
(formylamino)adamantane. 2-(formylamino) adamantane with two substitutes
which were defied (Table 3.1).
3. Series 3 consisted of bicyclo[2.2.2]octyltriazole derivatives as 11β
HSD1inhibitors. 1-methylbicyclo[2.2.2.]octane was considered as the main
nucleus and around it two substitutes were defined (Table 3.1).
Descriptor Calculation
TSAR offers calculation of number of descriptors which includes molecular surface
area, molecular volume, moments of inertia, ellipsoidal volume, Verloop parameters,
lipole moments, dipole moments, molecular mass, molecular connectivity indices,
molecular shape indices, Wiener index, electrotopological state indices, log P, number
of defined atoms (carbon, nitrogen, etc.), rings (aromatic and aliphatic) and groups
(methyl, hydroxyl, etc.). VAMP parameters (electrostatic parameters) like electronic
energy, nuclear repulsion energy, total energy, accessible surface area, atomic charge,
heat of formation, total dipole, polarizability, and dipole components can also be
calculated using TSAR. In the initial approach more than 250 molecular descriptors
for the whole molecule as well as substituent’s belonging to each series were
calculated.313
71
CHAPTER-3
Table 3.1: Lead structures and the substitution pattern of all three series used in QSAR
study
O O
HO N R1
H
N R2
O
Series 1( β O O
alanine,
HO N R1
H
isoserine and H O
R1 N N
thiazole ) R3
O
R3 N R1
(GRA) H
O O N
R2
HO N R1
H
N N R2
R3
Series 2 H
N H
N N R1
(Pyrrolidine R1
carboxamide) O R2 O
(11β HSD1
inhibitors)
N
Series 3
(Bicyclo[2.2.2] N N
R1
octyltriazole) R2
(11β HSD1 X
n N
inhibitors)
N N
Data Reduction
Out of large number of descriptors only few are significantly correlated with the
activity and many of the descriptors are inter-correlated which causes negative effect
on interpretability of the final model. Reduction of irrelevant descriptor is an
72
CHAPTER-3
important step to obtain the statistically optimized and meaningful QSAR model. The
correlation matrix was generated to observe the inter-correlation between the
descriptors and correlation between descriptors and biological activity. The
descriptors with high inter-correlation were examined for their correlation with
biological activity and the descriptors with low correlation were discarded. This
restricted false prediction of the QSAR model, as high co-linearity between
descriptors can cause statistical instability and over prediction which makes the
interpretation of mechanism difficult. Descriptors having distributions of values
clumped about a few distinct values were also removed since these do not explain
continuous variation of activity.
Table 3.2: Training and test set data for all three series under investigation
Number of Number of
Number of
Total number compounds in compounds
Series compounds in
of compounds the training identified as
the test set
set outliers
73
CHAPTER-3
Statistical Analysis
TSAR provides a wide range of statistical methods which can be used in the
development predictive QSAR models. The QSAR techniques used in TSAR includes
multiple linear regression (MLR), partial least square regression (PLS) and neural
network analysis (ANN). The output is displayed in the form of a model highlighting
substituent points that are strongly correlated with the pharmacological properties
under investigation. The TSAR methodology assumes that a proper distribution of
chosen descriptors facilitates all the information required to understand the
mechanism of action.314
74
CHAPTER-3
Validation of Model
Validation is a vital feature of any QSAR modeling. Reliability and relevance of the
QSAR model is established by this specific process. It judges the predictive ability of
a model on a different data and helps in determining the complexity of an equation. A
model developed by TSAR can be validated by following methods.
1. Cross Validation (r2cv): It is the most common method of validating a QSAR
model. This method includes repeating the regression many times on subsets
of data by leaving out once in turn and r2 is calculated using the predicted
values of the left out molecule. Cross-validation test (r2cv,) value for very
robust model is usually smaller than the overall r2 for a QSAR equation and
75
CHAPTER-3
should be greater than 0.50.319 Cross validation was carried out for all the
developed models and the values were analyzed.
2. Test Set Validation: This method includes comparison of the predicted and
observed activities of the test set of compounds. Structure generation,
optimization and charge derivation for the test set compounds were done in the
same way as that of the training set compounds. The predicted activities of the
test set were calculated using the model produced by the training set.
Predictivity of the developed models was assessed on the basis of r2 value. In
general, r2 test value should be greater than 0.6 and smaller than r2 of the
training.320
3. Overfitting: A model can be suffer with over-fitting if it includes more
descriptors than required. In order to avoid over-fitting, small number of
descriptors were used for model development.321
4. Outliers: Compounds having unpredicted biological activity and are incapable
to fit in a QSAR model are known as outliers. Removing outliers from the
main data set can settle the problem. Calculation of residual values and
standardized residuals helped to find the outliers in data set. The compounds
with higher residual values were deleted as outliers.
76
CHAPTER-3
R3 N R1
H
N
R2
Basic nucleus
S
O
a24 - O 116
HO
Cl
H
N O
F
a25 Trans O 55
O F
HO
F
H
N O
F
a26 Cis O 47
O F
F HO
H
N O
a27 Trans O
1045
N HO
H
N Cl O
a28 Trans 226
O
Cl HO
H
N
O
a29 Trans O 35
HO
F
H
N F
F O
a30 Trans O 56
HO
F F F
77
CHAPTER-3
H
N O
a31 O N 2429
O HO
H
N O
a32 140
O
O HO
F F
F
O O
H
a33 N 215
HO
O
N
Br
H
N O
a34 O
F 153
O F HO
F
H O
N
a35 1088
O HO
F F
F
O
H
a36 N 182
O HO
O
N
O
H
a37 N 56
F
F
O
O
HO
F
H Cl
N O
a38 O 80
HO
Cl
H
a39 N O
141
O
OCF3 HO
O N H O
N
a44 782
O
OCF3 HO
78
CHAPTER-3
O H
N O
a45 1984
O
OCF3 HO
H
N O
a46 59
O
OCF3 HO
H
N O
a47 122
O
OCF3 HO
H O
a48 O N
528
O
OCF3 HO
O H O
O N
a49 1207
O
OCF3 HO
H
N O
a50 96
O
OCF3 HO
H
N O
a51 137
O HO
OCF3
H
N O
a52 571
O
OCF3 HO
H
N O
a53 O + O 167
N
O
HO
O
F
a54 F 262
S F HO
79
CHAPTER-3
O
a55 179
HO
O
F
a56 F 61
O F
HO
Cl
O
a57 27
HO
Cl
H
N O
a62 1700
O
OCF3 HO
H
N
H
N
a65* HO 2960
O
OCF3 O
Me
Me Me H
N O
b11 36
O HO
OCF3
H
N Cl O
b13 O 12
HO
Cl
H O
N Cl
b15* R O HO 3.9
Cl OH
H
N Cl
O
b16 S O HO 51
Cl OH
80
CHAPTER-3
Me
H
Me Me N CF3
O
b17 Trans O 16
CF3
HO
Me H
Me Me N CF3 O
b18 R, trans O HO 8
CF3 OH
Me
Me Me H
N CF3
O
b19 S,trans O HO 82
CF3 OH
H O
N Cl
b20 R O HO 693
Cl OH
Me
Me Me H O
N Me
b21 O HO 27
Me
OH
O
H
N Br
b22 HO 8
O
OH
H O
N CF3
b23 O HO 7
Br OH
H
O
N CF3
b24 O
HO 14
OMe
OH
81
CHAPTER-3
H O
N CF3
b25 O HO 9
OMe OH
Me
Me Me H O
N CF3
b26 Trans O HO 10
Me OH
H
O
N
b27 HO 166
O
OH
H
O
N Cl
b28 HO 14
O
OH
H O
N
b29 HO 6
O
SCF3
OH
H
O
N CF3
b30 HO 9
O
CN
OH
O
H
N
b31 HO 18
O
OH
H O
N Cl
b32 O
HO 4
Cl
OH
H F F O
N
O
b33 O F HO 6
O
F
OH
82
CHAPTER-3
H
N CF3
O
b34 O HO 13
F OH
O
H
N SMe
b35 HO 9
O
OH
H
O
N SO2CF3
b36 HO 5
O
OCF3
OH
OCF3
N
O
c6 59
S HO
H
CF3
N
O
c7 64
S
HO
H
OCF3
O
N
c8 96
S
HO
H
Cl
Cl O
c10 N
54
S
H
HO
CF3
O
N
c11 48
S
HO
H
83
CHAPTER-3
Cl CF3
N
O
c16 53
S HO
H
Cl OCF3
O
N
c17 53
S
HO
H
CF3 Cl
N
O
c18 93
S HO
H
OCF3 CF3
O
N
c19 51
S
HO
H
MLR Analysis
The linear regression analysis was executed on the dataset having GRA activity as
dependent variable and reduced pool of descriptors as the independent variable. The
three descriptors namely Verloop L, Dipole moment X component and Log P; having
minimum inter-correlations among them were left after data reduction (Table 3.4).
Table 3.4: Correlation matrix of the independent variables used in the final model illustrating
the degree of correlation
X2: Dipole
Moment X
0.16488 1 -0.10984 0.36158
Component
(Subst.2)
X3: Log P
0.16488 -0.10984 1 0.26705
(Whole Molecule)
During the whole processes of model development, five molecules (a36, a45, b11,
b13 and c12) were detected as outliers having standardized residuals greater than
84
CHAPTER-3
±1.7. Removal of these five molecules yielded the model having the |r2-r2cv| of just
0.04 exhibiting good predictive ability of the model (Equation 1 and 2).
Original Data: Y = = -1.147*X1 + 0.101*X2 + 0.344*X3 + 1.143 Eq. (1)
Standardized Data: Y = -0.56*S1 + 0.25*S2 + 0.32*S3 - 1.827 Eq. (2)
Where, X1 = Verloop L (Subst. 3)
X2 = Dipole Moment X Component (Subst. 2)
X3 = Log P (Whole Molecule)
r = 0.87, r2 = 0.76, r2cv = 0.72, F = 52.88, s = 0.37
Residual sum of square = 6.921, Predictive sum of square = 8.119
r2 of 0.76 explains 76% variance in biological activity, which is a measure of good fit
by the regression equation. The model was cross-validated by leave one out method
resulting in r2cv value of 0.72 which is an excellent statistical validation of the
developed model. All other statistical value such as t-value, jackknife SE and
Covariance SE as shown in Table 3.5 also confirmed the significance of each and
every descriptor for GRA activity. The exclusion of any of the descriptor from the
model resulted in a very poor model.
Table 3.5: Statistical data of the independent variable illustrating the significance in terms of
statistical parameters
Dipole Moment X
Component 0.021454 0.021499 4.6931 2.2729
(Subst.2)
The 9 test set compounds which were left a side during model development were used
to check the external predictive ability of QSAR model. A QSAR model with r2 value
of more than 0.6 for the test set is considered to be good and in present case r2 value
for test set compounds was found to be 0.74, which clearly indicates that the
developed model is highly predictive and statistically significant.
85
CHAPTER-3
PLS Analysis
To further confirm the robustness and prognostic ability of generated model PLS
analysis was executed on the same data set. The results of the PLS as shown in
Equation 3 also were evaluated on the basis of r2 and statistical significance of the
model.
NN Analysis
The multiple-layer NN functionality, under a supervised training by the back
propagation, was used. The number of neurons in the hidden layer and the number of
rows in the training set were balanced to retrieve the best predictive power for the
neural network. In the present case the neural network run with 1 hidden nodes and
70% of training set and 30% of the test set generated the best correlation coefficient
(r2) value. The statistics obtained for the NN treatment were N = 52, input columns
(descriptors) = 3, net configuration = 3-1-1 (3 input nodes, 1 processing node, 1
output node), with test rms = 0.11, best rms = 0.086 and r2 = 0.76 for training and r2 =
0.73 for test. It is noteworthy that NN with same set of descriptors produced model
with equal statistical strength to that of MLR and PLS models. The QSAR model
obtained exhibited negative dependency on the Verloop L (Subst. 3); positive
dependencies on Dipole moment X component (Subst. 2) and Log P (Whole
molecule). The dependency plots are depicted in figure (Figure 3.4-3.6). The actual
and estimated value obtained from MLR, PLS and FFNN analysis of the training and
test set of compounds are shown in Table 3.6-3.7 and their related graphs are shown
in (Figure 3.1-3.3).
86
CHAPTER-3
Table 3.6: Actual and predicted activity data obtained from MLR, PLS and NN analysis of
training set compounds
87
CHAPTER-3
88
CHAPTER-3
Table 3.7: Actual and predicted activity data obtained from MLR, PLS and NN analysis of test
set compounds
Figure 3.1: Plot of actual activity versus estimated activity of training and test set obtained
from MLR analysis
89
CHAPTER-3
Figure 3.2: Plot of actual activity versus estimated activity of training and test set obtained
from PLS analysis
Figure 3.3: Plot of actual activity versus estimated activity of training and test set obtained
from NN analysis
90
CHAPTER-3
Figure 3.4: Dependency graph illustrating correlation between the Verloop L (subst.3) used
to train the neural network architecture versus the actual activity data
Figure 3.5: Dependency graph illustrating correlation between the Dipole moment X
component (subst.2) used to train the neural network architecture versus the actual activity
data
91
CHAPTER-3
Figure 3.6: Dependency graph illustrating correlation between the Log P (whole molecule)
used to train the neural network architecture versus the actual activity data
Verloop L (Subst. 3)
It is a steric parameter that describes the bulkiness of the compound and is based on
the standard substitution of bond lengths and angles of the compounds.322 Verloop L,
the length parameter, is defined as the length of the substituent along the axis of its
substitution with the parent. From the regression equation and dependency plot it is
clear that Verloop L parameter is negatively correlated with the biological activity
indicating that decrease in the length of substituent 3 will increase the GRA activity
because high length of substituent orients the molecule in such a way that there is
minimum binding of ligand to receptor. It can be explained with the help of the
92
CHAPTER-3
Table 3.8: Correlation of biological activity of active and inactive molecules with Verloop L
descriptors
Biological
Name of Verloop L Substituent on R3
activity IC50
compound (Subst.3) position
(µM)
b15 3.9 3.96829 2-hydroxyacetic acid
Active
compounds b32 4 3.87144 2-hydroxyacetic acid
93
CHAPTER-3
Table 3.9: Correlation of biological activity of active and inactive molecules with Dipole
moment X component descriptors
Dipole
Biological
Name of moment X Substituent on R2
activity IC50
compound component position
(µM)
(Subst.2)
H
N Cl
Cl
Active H
N Cl
compounds b32 4 2.44226 O
Cl
H
N SO2CF3
b36 5 5.41165
O
OCF3
H
N
H
Inactive N
a45 1984 -2.65035
O
compounds OCF3
H
N
a65 2960 -4.6015
O
OCF3
94
CHAPTER-3
b15, b32 and b36 and small log P value of the least active compounds like a31 and
a45 supports the theory (Table 3.10).
Table 3.11: Correlation of biological activity of active and inactive molecules with Log P
descriptors
Biological
Name of Log P
activity IC50
compound (Whole molecule)
(µM)
b36 5 6.7739
Conclusion
Specific molecular features important for binding of 66 antagonists to the glucagon
receptor have been identified by the reported 2D QSAR models. The equations of
MLR, PLS and dependency plots obtained from NN provide highly valuable
95
CHAPTER-3
H
N R1
R2 O
Basic Nucleus
Comp. R1 R2 IC50(nm)
4 H 4.15
N
7 H 2.2
11 N
H 22
12 N H 1.2
96
CHAPTER-3
13 N H 3.5
14 N H 0.03
15 N H 0.03
16 N H 0.03
17 N H 0.32
18 N H 0.32
CF3
19 N H 0.32
CF3
Cl
20 N H 0.32
CN
21 N H 0.03
22 N H 0.32
CN
23 N H 20
OH
97
CHAPTER-3
24 N H 5
N
25 N H 10
O
OH
26 N H 0.34
OH
N
27 H 0.03
OH
35 H 34
N
OH
36 H 0.8
N
OH
37 H 0.173
N
OH
38 H 1
N
OH
39 H 0.03
N F
98
CHAPTER-3
OH
40 H 30
N O
H
H
41 H 19.4
N
42 N
OH 23
43 N OH 31.3
MLR Analysis
The linear regression analysis was executed on the compounds showing 11β HSD1
inhibitory activity as dependent variable and reduced pool of descriptors as the
independent variable. The two descriptors namely Dipole moment X component and
Log P; having minimum inter correlations among them and maximum correlation
with the biological activity were left after data reduction (Table 3.12).
Table 3.12: Correlation matrix of the independent variable used in the final model illustrating
the degree of correlation
X1:Dipole Moment X
Descriptors Component (Whole X2: Log P Y: -log Ic50
Molecule) (Whole Molecule)
X1: Dipole
Moment X
1 -0.170075 -0.612715
Component (Whole
Molecule)
X2: Log P
(Whole Molecule)
-0.170075 1 0.821642
During the whole processes of model development, three molecules (15, 17 and 24)
were detected as outliers having residual value greater than ±1. Removal of these
99
CHAPTER-3
three molecules yielded the model having the |r2-r2cv| of just 0.03 exhibiting good
predictive ability of the model. Equation 4 and 5 shows good correlation between 11β
HSD1 inhibitors and selected set of two descriptors. Small difference between r2 and
r2cv defines the good prediction ability of the model.
Original Data: Y = -0.211*X1 + 1.418*X2 - 3.091 Eq.4
Standardized Data: Y = -0.558*S1 + 0.847*S2 + 0.179 Eq.5
Where, X1 = Dipole Moment X Component (Whole molecule)
X2 = Log P (Whole Molecule)
r = 0.95, r2 = 0.91, r2cv = 0.87, F = 67.04, s = 0.38
Residual sum of square = 1.9877, Predictive sum of square = 2.76722
r2 of 0.91 explains 91% variance in biological activity, which is a measure of good fit
by the regression equation. The model was cross-validated by leave one out method
resulting in r2cv value of 0.87 which is an excellent statistical validation of the
developed model. All other statistical value such as t-value, jackknife SE and
Covariance SE (Table 3.13) also confirmed the significance of each and every
descriptor for 11β HSD1 inhibitory activity. The exclusion of any of the descriptor
from the model resulted in a very poor model.
Table 3.13: Statistical data of the independent variable illustrating the significance in terms
of statistical parameters
Log P (Whole
0.155433 0.160054 8.85972 4.09371
Molecule)
The 8 test set compounds were used to check the prediction ability of the developed
QSAR models. These compounds were treated in a manner analogous to the
compounds in the training set. A QSAR model having r2 value of more than 0.6 for
the test set is considered to be a sound model. The r2 value for test set compounds was
found to be 0.84, which indicate that the developed model is highly predictive and
statistically significant.
100
CHAPTER-3
PLS Analysis
To further confirm the robustness and predictive ability of generated model PLS
analysis was executed on the same data set. For a well-defined problem, both MLR
and PLS should have comparable results. The results of the PLS (Equation 6) also
were evaluated on the basis of r2 and statistical significance of the model. The
statistical output of developed PLS model in terms of r2cv (0.88) clearly shows that all
the two descriptors identified during model development are highly significant and
are contributing heavily towards 11β HSD1 inhibitory activity of the compounds
under study. Moreover statistical significance value of 1.09 further testified the
model.
NN Analysis
The multiple-layer NN functionality, under a supervised training by the back
propagation, was used. The number of neurons in the hidden layer and the number of
rows in the training set were balanced to retrieve the best predictive power for the
neural network. In the present case, the neural network run with 1 hidden nodes and
75% of training set and 25% of the test set generated the best value of correlation
coefficient (r2).The statistics obtained for the NN treatment were N = 17, input
columns (descriptors) = 2, net configuration = 2-1-1 (2 input nodes, 1 processing
node, 1 output node), test rms = 0.12, best rms = 0.11 and r2 = 0.88 for training and r2
= 0.82 for test. With the same descriptors as used for the MLR and PLS models, the
NN approach showed almost similar results in terms of predictions abilities. The
model exhibited negative dependency on Dipole moment X component (Whole
molecule) and positive dependency on Log P (Whole molecule). The dependency
plots are depicted in figure (Figure 3.11-3.12). The actual and estimated value
obtained from MLR, PLS and FFNN analysis for the training and test set of
compounds are shown in Table 3.14-3.15 and their related graphs are shown in
(Figure 3.8-3.10).
101
CHAPTER-3
Table 3.14: Actual and predicted activity data obtained from MLR, PLS and NN analysis of
training set compounds
Table 3.15: Actual and predicted activity data obtained from MLR, PLS and NN analysis of test
set compounds
102
CHAPTER-3
Figure 3.8: Plot of actual activity versus estimated activity of training and test set obtained
from MLR analysis
Figure 3.9: Plot of actual activity versus estimated activity of training and test set obtained
from PLS analysis
103
CHAPTER-3
Figure 3.10: Plot of actual activity versus estimated activity of training and test set obtained
from NN analysis
Figure 3.11: Dependency graph illustrating correlation between the Dipole moment X
component (whole molecule) used to train the neural network architecture versus the actual
activity data
104
CHAPTER-3
Figure 3.12: Dependency graph illustrating correlation between the Log P (whole molecule)
used to train the neural network architecture versus the actual activity data
105
CHAPTER-3
Table 3.16: Correlation of biological activity of active and inactive molecules with Dipole
moment X component descriptors
14 0.03 0.754892
15 0.03 -2.59325
27 0.03 -3.22158
39 0.03 0.754892
43 31.3 2.62094
Inactive
compounds 35 34 1.72068
106
CHAPTER-3
Table 3.17: Correlation of biological activity of active and inactive molecules with Dipole
moment X component descriptors
14 0.03 3.1472
15 0.03 2.8586
27 0.03 2.5224
39 0.03 2.737
43 31.3 1.6206
Inactive
compounds 35 34 1.1634
Figure 3.13: Effect of various descriptors on the whole molecule of 11β HSD1 inhibitory
activity
Conclusion
Specific molecular features important for binding of 28 11β HSD1 inhibitors have
been identified by the reported 2D QSAR models. The equations of MLR, PLS and
dependency plots obtained from NN provide highly valuable information about the
107
CHAPTER-3
Table 3.18: Structures and biological activities of the 32 11β HSD1 inhibitors
Bicyclo[2.2.2]octyltriazole derivatives
R1
R2
Basic Nucleus
Comp. R1 R2 IC50(nm)
1 n-butyl N 4
N N
2 Isobutanol N 35
N N
CF3
3 n-butyl N 1.8
N N
CF3
5 O N 1.2
N N
108
CHAPTER-3
CF3
13 MeO N 2
N N
CF3
14 OCH2CH3 N 4
N N
CF3
Cl
15 N 1.1
O
N N
CF3
SO2CH3
16 N 1
O
N N
CF3
17 NHSO2CH3 N 30
N N
CF3
18 NHSO2CH2CH3 N 20
N N
CF3
19 NHSO2(CH2)2CH3 N 8.1
N N
CF3
20 CH2SCH2CH3 N 3.3
N N
CF3
21 CH2SOCH2CH3 N 4.1
N N
CF3
22 CH2SO2CH2CH3 N 7
N N
CF3
23 CH2SO2CH3 N 11
N N
109
CHAPTER-3
CF3
24 CH2SO2CH2CH2CH3 N 5.1
N N
CF3
25 CH2SO2CH(CH3)2 N 4.1
N N
CF3
26 CH2SO2C(CH3)3 N 5.4
N N
CF3
O
27 S N 4.4
O
N N
CF3
F O
28 S N 5.5
O
N N
CF3
29 CH2SO2CF3 N 6.1
N N
CF3
30 CH2SO2CH2CF3 N 3.7
N N
CF3
31 SO2CH2CH3 N 25
N N
CF3
32 CH2CH2SO2CH2CH3 N 3.2
N N
CF3
33 CH2CH2CH2SO2CH2CH3 N 1.6
N N
Cl
34 CH2SO2CH2CH3 N 7.5
N N
110
CHAPTER-3
Cl
35 CH2SO2CH2CH3 N 900
N N
Cl
N
36 CH2SO2CH2CH3 2800
N N
Cl
OH
37 CH2SO2CH2CH3 N 2.8
N N
Br
38 CH2SO2CH2CH3 N 10
N N
39 CH2SO2CH2CH3 N 20
N N
40 CH2SO2CH2CH3 N 140
N N
MLR Analysis
The linear regression analysis was performed using the 11β HSD1 inhibitory activity
as dependent variable and reduced pool of descriptors as the independent variable.
The three descriptors namely Inertial moment length, Log P and VAMP polarization
XX; having minimum inter correlations among them and maximum correlation with
the biological activity were left after data reduction (Table 3.19).
During the whole processes of model development, two molecules (35 and 37) were
detected as outliers having residual value greater than ±0.65. Removal of these two
molecules yielded the model having the |r2-r2cv| of just 0.09 signifying the statistical
strength of the model. Equation 7 and 8 shows correlation between 11β HSD1
inhibitors and selected set of two descriptors
Original Data: Y = -0.163*X1 + 0.271*X2 + 0.037*X3 - 3.095 Eq.7
Standardized Data: Y = -0.491*S1 + 0.250*S2 + 0.192*S3 - 0.928 Eq.8
Where, X1 = Inertia moment 1 length (Subst.2)
X2 = Log P (Whole Molecule)
111
CHAPTER-3
Table 3.19: Correlation matrix of the independent variable used in the final model illustrating
the degree of correlation
X1:Inertial
X2: Log P X3:VAMP Y:
Moment
Descriptors (Whole Polarization XX
Length -log Ic50
Molecule) (Whole Molecule)
(Subst.2)
X1:Inertial
Moment
1 -0.265945 -0.271725 -0.810608
Length
(Subst.2)
X2: Log P
(Whole -0.265945 1 0.485858 0.625431
Molecule)
X3:VAMP
Polarization
-0.271725 0.485858 1 0.587932
XX (Whole
Molecule)
r2 of 0.94 explains 94% variance in biological activity, which is a measure of good fit
by the regression equation. The model was cross-validated by leave one out method
resulting in r2cv value of 0.89 which is an excellent statistical validation of the
developed model. All other statistical value such as t-value, jackknife SE and
Covariance SE (Table 3.20) also confirmed the significance of each and every
descriptor for 11β HSD1 inhibitory activity. The exclusion of any of the descriptor
from the model resulted in a very poor model.
Table 3.20: Statistical data of the independent variable illustrating the significance in terms
of statistical parameters
Log P
0.0599812 0.0751529 3.61263 0.00199047
(Whole Molecule)
VAMP Polarization
XX (Whole 0.0143 0.0133546 2.76035 0.0128846
Molecule)
112
CHAPTER-3
The 8 test set compounds were used to check the prediction ability of QSAR model.
All the test compounds were treated in a manner analogous to the compounds in the
training set. A QSAR model having r2 value of more than 0.6 for the test set is
considered to be a sound model. The r2 value for test set compounds was 0.86, which
clearly indicates that the developed model is highly predictive and statistically
significant.
PLS Analysis
To further confirm the robustness and predictive ability of generated model, PLS
analysis was performed using the same data set. For a well-defined problem, both
MLR and PLS should have comparable results. The results of the PLS as shown in
Equation 9 were also evaluated on the basis of r2 and statistical significance of the
model. The statistical output of developed PLS model in terms of r2cv (0.81) clearly
shows that all the three descriptors identified during model development are highly
significant and are contributing heavily towards 11β HSD1 inhibitory activity of the
compounds under study. Moreover statistical significance value of 1.15 further
testified the model.
NN Analysis
The multiple-layer NN functionality, under a supervised training by the back
propagation, was used. The number of neurons in the hidden layer and the number of
rows in the training set were balanced to retrieve the best predictive power for the
neural network. In the present case the neural network run with 1 hidden nodes and
70% of training set and 30% of the test set generated the best value of correlation
coefficient (r2).The statistics obtained for the NN treatment were N = 22, input
columns (descriptors) = 3, net configuration = 3-1-1 (3 input nodes, 1 processing
node, 1 output node), test rms = 0.08, best rms = 0.05 and r2 = 0.91 for training and r2
= 0.75 for test. With the same descriptors as used for the MLR and PLS models, the
NN approach exhibited almost similar predictions abilities. The QSAR model
obtained showed negative dependency on Inertia moment 1 length (Substituent 2) and
113
CHAPTER-3
Table 3.21: Actual and predicted activity data obtained from MLR, PLS and NN analysis of
training set compounds
114
CHAPTER-3
Table 3.22: Actual and predicted activity data obtained from MLR, PLS and NN analysis of test
set compounds
Figure 3.14: Plot of actual activity versus estimated activity of training and test set obtained
from MLR analysis
115
CHAPTER-3
Figure 3.15: Plot of actual activity versus estimated activity of training and test set obtained
from PLS analysis
Figure 3.16: Plot of actual activity versus estimated activity of training and test set obtained
from NN analysis
116
CHAPTER-3
Figure 3.17: Dependency graph illustrating correlation between the Inertia moment1 length
(Subst.2) used to train the neural network architecture versus the actual activity data
Figure 3.18: Dependency graph illustrating correlation between the Log P (whole molecule)
used to train the neural network architecture versus the actual activity data
117
CHAPTER-3
Figure 3.19: Dependency graph illustrating correlation between the VAMP Polarization XX
(whole molecule) used to train the neural network architecture versus the actual activity data
Table 3.23: Correlation of biological activity of active and inactive molecules with Inertia
moment 1 length descriptors
Biological
Name of Inertia moment 1 Substituent on R2
activity IC50
compound length (Subst.2) position
(µM)
CF 3
16 1 5.74142 N
N N
CF3
Active
15 1.1 5.74547 N
compounds N N
CF 3
5 1.2 5.72429 N
N N
40 140 7.69677 N
N N
Cl
Inactive
35 900 11.8791 N
compounds
N N
Cl
36 2800 19.6661 N
N N
119
CHAPTER-3
Table 3.24: Correlation of biological activity of active and inactive molecules with Log P
descriptors
Biological
Name of Log P
activity IC50
compound (Whole molecule)
(µM)
16 1 5.9216
Active
compounds 15 1.1 7.2946
5 1.2 6.2234
40 140 4.3435
Inactive 900 4.722
35
compounds
36 2800 4.722
120
CHAPTER-3
Table 3.25: Correlation of biological activity of active and inactive molecules with VAMP
polarization XX descriptors
Biological
Name of VAMP Polarization XX
activity IC50
compound (Whole molecule)
(µM)
16 1 62.6568
Active
compounds 15 1.1 50.0554
5 1.2 46.4158
40 140 42.432
Inactive 900 42.103
35
compounds
36 2800 41.9509
Figure 3.20: Effect of various descriptors on the whole molecule of 11β HSD1 inhibitory
activity
Conclusion
Specific molecular features important for binding of 32 11β HSD1 inhibitors have
been identified by the reported 2D QSAR models. The equations of MLR, PLS and
121
CHAPTER-3
dependency plots obtained from NN provide highly valuable information about the
importance of structural features encoded by physical/chemical molecular descriptors,
such as Inertia moment 1 length, Log P and VAMP polarization XX for anti diabetic
activity. The present study will be useful in designing more promising novel 11β
HSD1 inhibitors containing lead structure.
122
CHAPTER- 4
Ligand Based
Pharmacophore (LBP)
Modeling
CHAPTER-4
123
CHAPTER-4
124
CHAPTER-4
Table 4.1: Training and test set data for all three series under investigation
Number of Number of
Number of
Total number compounds in compounds
Series compounds in
of compounds the training identified as
the test set
set outliers
Series 1( β alanine, isoserine and
70 56 13 01
thiazole antagonist of GRA)
Series 2 (Pyrrolidine carboxamide
30 20 10 00
Inhibitors of 11β HSD1)
Series 3 (Bicyclo[2.2.2]
octyltriazole Inhibitors of 11β 32 24 8 00
HSD1)
125
CHAPTER-4
the root mean-square (RMS) difference between the estimated and the actual affinity
for the training set increases. The RMS value predicts the quality of prediction of the
model. The weight cost is directly proportional to the difference between the actual
and ideal weights of the features. The ideal value of the weight is reported to be 2
because higher weight values leads to fitting of abnormal conformations of the
compounds to the features. The third function, configuration cost characterizes
complexity of the model. The configuration cost is a fixed cost that represents the
complexity of the hypothesis space to be optimized. It should not exceed a maximum
value of 17 for a significant pharmacophore model. A difference of 20 bits or more
between null cost and total cost signifies the good correlation of the model. An ideal
hypothesis is characterized by low RMS value, high correlation coefficient (r) and the
highest difference between null and total cost. In addition to these statistical
parameters, the soundness and predictability of the generated pharmacophore models
were also evaluated by fisher randomization test, internal and external test set
validation.
126
CHAPTER-4
geometric fit of the entire test set compounds mapped onto the best pharmacophore.
The geometric fit value is calculated on the basis of number of pharmacophore
features successfully superimposed to the corresponding functional groups, the weight
of the corresponding hypothesis features spheres, the distance between the center of a
particular pharmacophoric sphere (feature centroid) and the center of the
corresponding superimposed chemical moiety of the fitted compound.324 The squared
correlation coefficient (r2) values were determined and evaluated. Moreover the
mapping pattern of the most active compound was compared with least active
compound and the results were analyzed.
Pharmacophore Generation
A series of 70 structurally diverse compounds having GRA activity from 3.9 nM to
2960 nM was used for development of pharmacophore model (Table-4.2). Out of 70
compounds a training set of 57 compounds was used to build the pharmacophore
model, while 13 compounds were kept as internal test set to validate the predictivity
of the generated model. Compound a24 showed an error ratio, so it was removed from
the dataset (training set) during the course of pharmacophore model development.
127
CHAPTER-4
Table 4.2: Structure and biological activity of β-lanine, isoserine and thiazole core
derivatives with glucagon receptor antagonistic activity
O O O
HO
HO N N
H H H
N N O N N
F F
F F
O O
O F O F
O O
O
H HO N
N H H
HO N OMe N N Cl
H H
O N N
F O
F
O
O F Cl
β alanine derivatives
O O
HO N R1
H
N R2
β alanine moiety O
a24 - O 116
Cl
H
N
F
a25 Trans O 55
O F
F
128
CHAPTER-4
H
N
F
a26 Cis O 47
O F
F
H
N
a27 Trans O
1045
N
H
N Cl
a28 Trans 226
O
Cl
H
N
a29 Trans O 35
F
H
N F
F
a30 Trans O 56
F F F
H
N
a31 O N 2429
O
H
N
a32 140
O
O
F F
F
O
H
a33 N 215
O
N
Br
H
N
a34 O
F 153
O F
F
H
N
a35 1088
O
129
CHAPTER-4
F F
F
H
a36 N 182
O
O
H
a37 N
F 56
F
O
O F
H Cl
N
a38 O 80
Cl
H
a39 N
141
O
OCF3
O N H
N
a44 782
O
OCF3
O H
N
a45 1984
O
OCF3
H
N
a46 59
O
OCF3
H
N
a47 122
O
OCF3
H
a48 O N
528
O
OCF3
130
CHAPTER-4
O H
O N
a49 1207
O
OCF3
H
N
a50 96
O
OCF3
H
N
a51 137
O
OCF3
H
N
a52 571
O
OCF3
H
N
a53 O + O 167
N
O
F
a54 F 262
S F
a55 179
F
a56 F 61
O F
Cl
a57 27
Cl
131
CHAPTER-4
Me
Me Me H
N
b11 36
O
OCF3
H
N Cl
b13 O 12
Cl
Me
H
Me Me N CF3
b17 Trans O 16
CF3
Isoserine derivative
O O
HO N R1
H
OH N R2
Isoserine moiety O
H
N Cl
b15* R O 3.9
Cl
H
N Cl
b16 S O 51
Cl
Me H
Me Me N CF3
b18 R, trans O 8
CF3
132
CHAPTER-4
Me
Me Me H
N CF3
b19 S,trans O 82
CF3
H
N Cl
b20 R O 693
Cl
Me
Me Me H
N Me
b21 O 27
Me
H
N Br
b22 8
O
H
N CF3
b23 O 7
Br
H
N CF3
b24 O 14
OMe
H
N CF3
b25 O 9
OMe
Me
Me Me H
N CF3
b26 Trans O 10
Me
133
CHAPTER-4
H
N
b27 166
O
H
N Cl
b28 14
O
H
N
b29 6
O
SCF3
H
N CF3
b30 9
O
CN
H
N
b31 18
O
H
N Cl
b32 O 4
Cl
H F F
N
O
b33 O F
6
O
F
H
N CF3
b34 O 13
F
134
CHAPTER-4
H
N SMe
b35 9
O
H
N SO2CF3
b36 5
O
OCF3
O O
HO N R1
H
X N
n R2
β alanine moiety S
Thiazole core
OCF3
c6 1 N 59
CF3
c7 1 N 64
OCF3
c8 1 N 96
Cl
Cl
c10 1 N 54
135
CHAPTER-4
CF3
c11 1 N 48
CH3
c12 1 N 93
CF3
Cl
c16 1 N 53
Cl OCF3
c17 1 N 53
CF3 Cl
c18 1 N 93
OCF3 CF3
c19 1 N 51
Cl
c24 0 N 34
CF3
Cl
c27 1 C 38
Cl OCF3
c28 1 C 25
136
CHAPTER-4
Table 4.3: Results of the top 10 pharmacophore hypotheses generated by the HypoGen
algorithm
Correlation
Hypothesis Total Cost Cost Difference RMSD Features
(r)
137
CHAPTER-4
Correlation
Hypothesis Total Cost Cost Difference RMSD Features
(r)
Hypothesis1, identified as the best hypothesis, was used to estimate the activity of the
training set molecules. All the training set compounds were classified by their activity
as highly active (<100 nM, +++), moderately active (100-300 nM, ++) and inactive
(>300 nM, +).. Among 56 training set compounds, two active compounds (a30, a37)
were predicted as moderate and inactive respectively; three moderate compounds
(a53, a36, a28) were predicted to be active and inactives respectively; and two
inactive compounds (b20, a45) were predicted as moderate by Hypothesis1.
Consequently, for 49 of 56 training set compounds, the predicted IC50 (nM) values
were within the same activity scale as the experimental values in the training set.
Table 4.4 and Figure 4.2 represent the actual and estimated GRA activity of the 56
training set molecules based on the best hypothesis
Table 4.4: Actual and estimated values of the training set based on the pharmacophore
hypothesis
138
CHAPTER-4
139
CHAPTER-4
140
CHAPTER-4
Figure 4.2: Plot of correlation coefficient between actual and estimated activity value of
training set
Pharmacophore Mapping
The obtained pharmacophoric features and their interfeature distances are shown in
Figure 4.3 (a) and 4.3(b). Mapping of the most active compound b15 on the
pharmacophore (Figure 4.3 (c)) reveals that the carboxyl group is mapped to negative
ionizable group, hydroxyl groups to hydrogen bond donor, benzene ring adjacent to
the bulky group to the hydrophobic and benzene ring near the electron withdrawing
141
CHAPTER-4
region to ring aromatic group, whereas the inactive compound a65 (Figure 4.3 (d))
missed negative ionizable feature due to absence of carboxyl group.
actual and estimated activities of the test set compounds. The scored estimated
activities and actual value of test set compounds are shown in Table 4.5. The
agreement between actual and predicted activity of test set compound testifies the
soundness of hypothesis 1.
Table 4.5: Actual and estimated values of the test set based on the pharmacophore hypothesis
143
CHAPTER-4
Figure 4.4: Plot of correlation coefficient between actual and estimated activity value test set
ii) Cat Scramble Validation: 99 spreadsheets were generated in order to have a 99%
confidence level (Figure 4.5). The data of cross validation clearly indicates that all
values generated after randomization produced hypotheses with no significant value.
Out of 99 runs, all trials had a correlation value less than 0.89, and also RMS
deviation and total cost were very high, which is not desirable for a good hypothesis.
Thus Cat Scramble validation also provided strong confidence in the generated model.
iii) External Test Set Validation: An external test set of 9 compounds comprising
thiophene derivatives and alkylidine hydrazide derivatives with activity range of 89
nm to 60000 nm showed a squared correlation coefficient (r2) of 0.65 (Figure 4.6).
144
CHAPTER-4
Figure 4.6: Plot of correlation coefficient between actual and estimated activity value of
external test set
As an additional external validation step some clinical trial candidates like BAY-27-
9955 and LY2409021 were mapped onto the developed model. BAY-27-9955 showed
only two feature mapping with a fit value of 4.02. The hydroxyl moiety was mapped
by HBD and methyl group by HY (Figure 4.7 (a)). This may be the reason why BAY-
27-9955 was failed in clinical trials. LY2409021 showed two features mapping with
a fit value of 5.238. The amino group was mapped by HBD and carboxyl moiety was
mapped by NI (Figure 4.7 (b)).
Figure 4.7: Pharmacophore mapping of clinical trial drugs: (a) BAY-27-9955 showed
mapping with HBD and HY (b) LY2409021 showed mapping with HBD and NI
145
CHAPTER-4
Conclusion
The present study has been focused on the development of pharmacophore model to
reveal structural and physical/chemical requirement for glucagon receptor
antagonistic activity. Best pharmacophore model generated using 56 molecules of
training set comprised of one hydrogen bond donor (HBD), one negative ionizable
(NI), one hydrophobic (HY) and one ring aromatic (RA) feature. The robustness of
the model was characterized by a high correlation coefficient of the training set r
value of 0.89 and test set r2 0.73. The developed model was validated by three
methods i.e. internal test set validation, Fischer’s validation and external test set
validation. All validation process suggested that the developed model is a good and
reliable pharmacophore model with high statistical significance.
Pharmacophore Generation
A series of 30 structurally diverse compounds having having 11β HSD1 inhibitory
activity from 0.03 nM to 290 nM was used for development of pharmacophore model
(Table-4.6). Out of 30 compounds a training set of 20 compounds was used to build
the pharmacophore model, while 10 compounds were kept as internal test set to
validate the predictivity of the generated model.
Table 4.6: Structure and biological activity of Pyrrolidine carboxamide derivatives with 11β
HSD1 inhibitory activity
H
N R1 N
N
N N
R2 O O
O O
4, 7, 11-27, 35-43 5 6
Comp. R1 R2 IC50(nm)
4 H 4.15
N
146
CHAPTER-4
7 H 2.2
11 N
H 22
12 N H 1.2
13 N H 3.5
14 N H 0.03
15 N H 0.03
16 N H 0.03
17 N H 0.32
18 N H 0.32
CF3
19 N H 0.32
CF3
Cl
20 N H 0.32
147
CHAPTER-4
CN
21 N H 0.03
22 N H 0.32
CN
23 N H 20
OH
24 N H 5
N
25 N H 10
O
OH
26 N H 0.34
OH
N
27 H 0.03
OH
35 H 34
N
OH
36 H 0.8
N
OH
37 H 0.173
N
148
CHAPTER-4
OH
38 H 1
N
OH
39 H 0.03
N F
OH
40 H 30
N O
H
H
41 H 19.4
N
42 N
OH 23
43 N OH 31.3
coefficient (r = 0.92, r2 = 0.85), root mean- square deviation (1.02), weight (2.01) and
error costs (77.82) and a cost difference of 47.89. The configuration cost was 10.95,
which indicates that no over fitting of the data has been exercised. The cost difference
between total and fixed costs for the best hypothesis was only 11.6 bits, indicating the
high probability of the true correlation of the data. Thus, hypothesis1 with four
features, viz., one H-bond donor (HBD), one positive ionizable (PI) and two
hydrophobes (HY1 and HY2) was considered as the best pharmacophore model for
11β HSD1 inhibitory activity. The cost values, correlation coefficients (r), RMSD,
and features for the top ten hypotheses are listed in Table 4.7.
Table 4.7: Results of the top 10 pharmacophore hypotheses generated by the hypoGen
algorithm
Hypothesis1, identified as the best hypothesis, was used to estimate the activity of the
training set molecules. All the training set compounds were classified by their activity
as highly active (<1 nM, +++), moderately active (1-20 nM, ++) and inactive (>20
nM, +). Among 20 training set compounds, only three inactive compounds (35, 40,
43) were predicted as moderate by Hypothesis1. Consequently, for 49 of 56 training
set compounds, the predicted IC50 (nM) values were within the same activity scale as
the experimental values in the training set. Table 4.8 and Figure 4.8 represent the
150
CHAPTER-4
actual and estimated 11β HSD1 inhibitory activity of the 20 training set molecules
based on the best hypothesis.
Table 4.8: Actual and estimated values of the training set based on the pharmacophore
hypothesis
23 20 5.658 6.787 ++ ++
24 5 9.396 6.567 ++ ++
25 10 7.269 6.679 ++ ++
35 34 12.891 6.43 + ++
40 30 3.094 7.049 + ++
151
CHAPTER-4
Figure 4.8: Plot of correlation coefficient between actual and estimated activity value of
training set
Pharmacophore Mapping
The obtained pharmacophoric features and their inter feature distances are shown in
Figure 4.9 (a) and 4.9 (b). Most active compounds 14, 15, 21, 27 and 39 showed full
four feature mapping to the pharmacophore (Figure 4.10 (a)-(e)). Adamantane ring of
all the active compounds showed mapping with HY1 whereas amino groups was
mapped onto HBD and pyrrollidine moieties were mapped by PI features. The
pyrrollidine ring substitutions of all the active compounds were mapped over HY2
features. Inactive compound 6 showed only two feature mapping (Figure 4.11).
cyclohexane ring on HY1whereas cycloheptane ring on HY2.
Figure 4.9: Pharmacophore features (Hypothesis 1): (a) Four pharmacophore features: one
HBD (pink color), one PI (red color), two HY1 and HY2 (light blue colors) and four excluded
volumes (grey color) (b) Interfeature distances between the pharmacophore features
152
CHAPTER-4
Figure 4.10: Pharmacophore mapping with active compounds: (a) Compound no. 14, (b)
Compound no. 15, (c) Compound no. 22, (d) Compound no. 27 and (e) Compound no. 39
scored estimated activities and actual value of test set compounds are shown in Table
4.9. The agreement between actual and predicted activity of test set compound
testifies the soundness of hypothesis 1.
Table 4.9: Actual and estimated values of the test set based on the pharmacophore hypothesis
11 22 5.001 6.841 + ++
42 23 37.007 5.972 + +
Figure 4.12: Plot of correlation coefficient between actual and estimated activity value of test
set
154
CHAPTER-4
ii) Cat Scramble Validation: 99 spreadsheets were generated in order to have a 99%
confidence level (Figure 4.13). The data of cross validation clearly indicates that all
values generated after randomization produced hypotheses with no significant value.
Out of 99 runs, all trials had a correlation value less than 0.92, and also RMS
deviation and total cost were very high, which is not desirable for a good hypothesis.
Thus Cat Scramble validation also provided strong confidence in the generated model.
iii) External Test Set Validation: An external test set of 9 compounds comprising
disubstituted cyclohexylbenzamide derivatives with activity range of 0.5 nm to 14 nm
showed a squared correlation coefficient (r2) of 0.62 (Figure 4.14).
Figure 4.14: Plot of correlation coefficient between actual and estimated activity value of
external test set
155
CHAPTER-4
Pharmacophore Generation
A series of 32 structurally diverse compounds having having 11β HSD1 inhibitory
activity from 1 nM to 2800 nM was used for development of pharmacophore model
(Table-4.10). Out of 32 compounds a training set of 24 compounds was used to build
the pharmacophore model, while 8 compounds were kept as internal test set to
validate the predictivity of the generated model.
156
CHAPTER-4
Bicyclo[2.2.2]octyltriazole derivatives
R1
R2
1-40
Comp. R1 R2 IC50(nm)
1 n-butyl N 4
N N
2 Isobutanol N 35
N N
CF3
3 n-butyl N 1.8
N N
CF3
5 O N 1.2
N N
CF3
13 MeO N 2
N N
CF3
14 OCH2CH3 N 4
N N
CF3
Cl
15 N 1.1
O
N N
CF3
SO2CH3
16 N 1
O
N N
157
CHAPTER-4
CF3
17 NHSO2CH3 N 30
N N
CF3
18 NHSO2CH2CH3 N 20
N N
CF3
19 NHSO2(CH2)2CH3 N 8.1
N N
CF3
20 CH2SCH2CH3 N 3.3
N N
CF3
21 CH2SOCH2CH3 N 4.1
N N
CF3
22 CH2SO2CH2CH3 N 7
N N
CF3
23 CH2SO2CH3 N 11
N N
CF3
24 CH2SO2CH2CH2CH3 N 5.1
N N
CF3
25 CH2SO2CH(CH3)2 N 4.1
N N
CF3
26 CH2SO2C(CH3)3 N 5.4
N N
CF3
O
27 S N 4.4
O
N N
158
CHAPTER-4
CF3
F O
28 S N 5.5
O
N N
CF3
29 CH2SO2CF3 N 6.1
N N
CF3
30 CH2SO2CH2CF3 N 3.7
N N
CF3
31 SO2CH2CH3 N 25
N N
CF3
32 CH2CH2SO2CH2CH3 N 3.2
N N
CF3
33 CH2CH2CH2SO2CH2CH3 N 1.6
N N
Cl
34 CH2SO2CH2CH3 N 7.5
N N
Cl
35 CH2SO2CH2CH3 N 900
N N
Cl
N
36 CH2SO2CH2CH3 2800
N N
Cl
OH
37 CH2SO2CH2CH3 N 2.8
N N
Br
38 CH2SO2CH2CH3 N 10
N N
159
CHAPTER-4
39 CH2SO2CH2CH3 N 20
N N
40 CH2SO2CH2CH3 N 140
N N
160
CHAPTER-4
Table 4.11: Results of the top 10 pharmacophore hypotheses generated by the hypoGen
algorithm
Hypothesis1, identified as the best hypothesis, was used to estimate the activity of the
training set molecules. All the training set compounds were classified by their activity
as highly active (<10 nM, +++), moderately active (10-30 nM, ++) and inactive (>30
nM, +). Among 24 training set compounds, one active compound (1) was predicted as
moderate; and five moderately active compounds (17, 18, 23, 31, 39) were predicted
as active by Hypothesis1. Consequently, for 19 of 24 training set compounds, the
predicted IC50 (nM) values were within the same activity scale as the experimental
values in the training set. Table 4.12 and Figure 4.16 represent the actual and
estimated 11β HSD1 inhibitory activity of the 24 training set molecules based on the
best hypothesis.
161
CHAPTER-4
Table 4.12: Actual and estimated values of the training set based on the pharmacophore
hypothesis
162
CHAPTER-4
Figure 4.16: Plot of correlation coefficient between actual and estimated activity value of
training set
Pharmacophore Mapping
The obtained pharmacophoric features and their inter feature distances are shown in
Figure 4.17 (a) and 4.17 (b). Most active compounds 16 showed full five features
mapping to the pharmacophore (Figure 4.17 (c)). Triazole ring showed mapping with
HBA_Li, carbon trifluoride moiety was mapped over HY1, benzene ring attached to
carbon trifluoride on HY2 and bicycle ring onto the HY3; whereas benzene ring
attached to sulfonyl group showed mapping over RA feature. Inactive compound 36
showed only three feature mapping (Figure 4.17 (d)). Methyl ring attached to the
triazole ring on HY1, the bicycle ring onto HY2 and HBA_Li showed mapping with
oxygen of sulfonyl group.
163
CHAPTER-4
4.13. The agreement between actual and predicted activity of test set compound
testifies the soundness of hypothesis 1.
Table 4.13: Actual and estimated values of the test set based on the pharmacophore
hypothesis
2 35 861.551 6.265 + +
164
CHAPTER-4
Figure 4.18: Plot of correlation coefficient between actual and estimated activity value of test
set
ii) Cat Scramble Validation: 99 spreadsheets were generated in order to have a 99%
confidence level (Figure 4.19). The data of cross validation clearly indicates that all
values generated after randomization produced hypotheses with no significant value.
Out of 99 runs, all trials had a correlation value less than 0.89, and also RMS
deviation and total cost were very high, which is not desirable for a good hypothesis.
Thus Cat Scramble validation also provided strong confidence in the generated model.
165
CHAPTER-4
iii) External Test Set Validation: An external test set of 33 compounds comprising
4-Methyl-5-phenyl triazoles derivatives with activity range of 1 nm to 650 nm showed
a squared correlation coefficient (r2) of 0.67 (Figure 4.20).
Figure 4.20: Plot of correlation coefficient between actual and estimated activity value of
external test set
167
CHAPTER-5
Virtual Screening
&
Structure Based
Pharmacophore (SBP)
Modeling
CHAPTER-5
Virtual Screening
Virtual screening represents a fast & cost effective tool for identification of novel lead
by screening chemical compound libraries. Virtual screening method has emerged as
powerful technique in drug discovery and now a day most of the research based
pharmaceutical companies are relying on computational tools for lead discovery and
optimization. Lipinski’s rule, estimated activity and fit values are used as
computational filters to obtain the most significant compounds from the large
compound databases.
of potential hits. In addition to this the database compounds were also screened on the
basis of the estimated values. Only the hits which had the estimated activity very
close to the most active compounds of the series taken to develop the pharmacophore
model were retained.
Table 5.1: Number of compounds identified through pharmacophore based sequential virtual
screening
Database Number of
Series
library compounds retrieved
169
CHAPTER-5
Estimated
Name of hits Fit value Mapping of hits compounds
value(µM)
3.227
SKDGRA1 7.091
5.539
SKDGRA2 6.857
9.799
SKDGRA3 6.609
170
CHAPTER-5
Table 5.3: NCI hits obtained from pyrrolidine carboxamide based pharmacophore
Estimated
Name of hits Fit value Mapping of hits compounds
value(µM)
0.443
SKDHSD1 7.893
Table 5.4: NCI hits obtained from bicyclo[2.2.2] octyltriazole based pharmacophore
Estimated
Name of hits Fit value Mapping of hits compounds
value(µM)
5.626
NSC 13385 8.449
171
CHAPTER-5
Estimated
Name of hits Fit value Mapping of hits compounds
value(µM)
9.366
NSC 16653 8.228
172
CHAPTER-5
Figure 5.1: Three dimensional structure of glucagon receptor and 11β HSD1 enzyme
pharmacophores are depicted in Figure 5.2 and Figure 5.3. The developed six
featured structures based pharmacophore models were validated using some well
173
CHAPTER-5
known clinical trial/clinical drug candidates with GRA and 11β HSD1 inhibitory
activity.
Figure 5.3: The inter-feature distances of the 11β HSD1 enzyme pharmacophore
174
CHAPTER-5
were mapped onto the pharmacophore. BAY-27-9955 showed mapping with 2HY and
1HBD features whereas LY2409021, exhibited mapping with 2HBA, 1HY and 1
HBD. The fit values were found to be 0.8 and 1.867 respectively for BAY-27-9955
and LY2409021 (Table 5.5).
Table 5.5: Pharmacophore mapping, fit value and lipinski’s violation of clinical trial drugs
(BAY-27-9955 and LY2409021)
Lipinski
Fit
Name Violation Features Mapped
Value
BAY-27-9955 0 0.8
LY2409021 0 1.867
175
CHAPTER-5
Table 5.6: Pharmacophore mapping, fit value and lipinski’s violation of NCI retrieved hits
(SKDGRA1, SKDGRA2 and SKDGRA3)
Lipinski
Name Violation Fit Value Features Mapped
SKDGRA1 0 1.647
SKDGRA2 0 0.999
SKDGRA3 0 1.561
The mapping of the retrieved hits onto the SBP provided a fair insight into the binding
of the hits with the receptor. SKDGRA1 showed good mapping with the SBP, missing
only one feature. It is interesting to note that, SKDGRA1 mapped onto HY and HBD
176
CHAPTER-5
at the same position of LBP mapping (Figure 5.2). The other hits SKDGRA2 and
SKDGRA3 also showed good mapping with the SBP, missing only two features.
SKDGRA2 mapped onto HBD at the same position of LBP mapping (Figure 5.2) and
SKDGRA3 mapped onto HY at the same position of LBP mapping (Figure 5.2).
Good mapping of SKDGRA1, SKDGRA2 and SKDGRA3 onto the SBP and
commonality in mapping with the LBP confirmed the binding potential of the hits
onto the receptor.
Table 5.7: Pharmacophore mapping, fit value and lipinski’s violation of partial 11β HSD1
inhibitor (carbenoxolone)
Lipinski
Fit
Name Violation Features Mapped
Value
Carbenoxolone 0 1.987
177
CHAPTER-5
Table 5.8: Pharmacophore mapping, fit value and lipinski’s violation of NCI retrieved hits
(SKDHSD1, SKDHSD2 and SKDHSD3)
Lipinski
Fit
Name Violation Features Mapped
Value
SKDHSD1 0 1.634
SKDHSD2 0 2.079
SKDHSD3 0 1.45
SKDHSD1 and SKDHSD2 showed fair mapping with the SBP (three features).
SKDHSD1 showed mapping onto HY at the same position of LBP mapping (Figure
5.3) while SKDHSD2 showed mapping onto HY and HBA at the same position of
LBP mapping (Figure 5.4). SKDHSD3 showed good mapping with the SBP (four
features). It showed mapping onto HY and HBA at the same position of LBP mapping
(Figure 5.4). Good mapping of SKDHSD1, SKDHSD2 and SKDHSD3 onto the SBP
and commonality in mapping with the LBP established the binding potential of the
hits onto the receptor.
178
CHAPTER-5
Conclusion
3D SBP models for GRA and 11β HSD1 inhibitor were developed and used for
validation of NCI hits retrieved through ligand based virtual screening. All the
retrieved hits mapped well to the SBP and also showed commonality in mapping with
LBP at the same position. This provides a confirmation of the possible binding of all
the retrieved hits obtained from LBP based virtual screening.
179
CHAPTER- 6
Molecular Docking
&
Experimental
Validation
CHAPTER-6
Molecular Docking
Molecular docking is a powerful in silico technique often used to visualize the mode
of binding of the ligand to its target protein. The vital information provided by the
molecular docking on drug-receptor interaction can be used in the designing of the
novel ligands with improved efficacy and reduced side effects. Now a days molecular
docking is exercised either before or after binding experiments and in vitro or in vivo
activity assays, to get insight into the precise binding geometry of the compounds
under investigation.326 Molecular docking consist of two essential components;
generation of promising ligand binding orientations known as sampling and prediction
of the binding strength for a specific ligand orientations known as scoring. The best
orientation with the lowest energy score is considered as the preferred binding mode.
Because of precision in predicting the correct binding interactions and orientations (in
some cases at a very high accuracy with reference to existing crystal structure of the
complex studied), molecular docking is being widely used in modern days drug
design projects.
Target Identification
Crystal structures of the recently discovered human glucagon receptor (4L6R-Figure
6.1) and 11β HSD1 in complex with NADP and carbenoxolone (2BEL- Figure 6.2)
was obtained from protein data bank and used for molecular docking studies.
Receptor and Ligand Preparation
The protein structures (glucagon receptor and 11β HSD1 enzyme) obtained from PDB
were checked for valency and the missing hydrogen were added. All the water
molecules from the protein hierarchy were removed and the structures were split into
the protein part and crystal ligand part. In both the cases the protein part was defined
as receptor molecules. The active site of receptor was identified using a sphere whose
location and radius was adjusted to 9.0 A˚, so that the active sites and the key residues
180
CHAPTER-6
of the proteins can be involved in interaction with ligands. The CharmM force field
was applied and the energy was minimized. The hits obtained from pharmacophores
Figure 6.1: Crystal structure of human glucagon receptor (4L6R). Labeled amino acids
(black color) represent the active site in crystal structure.
Figure 6.2: Crystal structure of 11β HSD1 enzyme (2BEL). Labeled amino acids (black
color) represent the active site in crystal structure.
181
CHAPTER-6
based virtual screening were energy minimized and used for docking.
182
CHAPTER-6
(Gln232, Leu386, Phe365, Asn238, Lys187), hence the these compounds can be
further developed as potential GR antagonists.
183
CHAPTER-6
protein (2BEL) using LibDock software implemented in DS. 100 docking poses
were generated for SKDHSD1 and 247 and 260 docking poses were generated each
for SKDHSD2 and SKDHSD3. Top five poses were evaluated for each compound on
the basis of LibDock score. SKDHSD1 docked well with a Lib Dock score of 81.234
(Figure 6.6). It showed 5 strong hydrogen bond interactions with Asn119, Tyr183,
Lys187, Lys44 and Gly41. SKDHSD1 also exhibited 7 weak Van der Waal
interactions with Gly45, Ile46, Leu215, Ile121 and His120. SKDHSD2 showed 2
strong hydrogen bond interactions with Gly41, and Ser169. It also exhibited 7 weak
Van der Waal interactions with Ser170, Asn119, His120, Ser43, Lys44, Thr220 and
Ile46 (Figure 6.7) with a LibDock score of 106.916. SKDHSD3 showed only one
strong hydrogen bond interaction with Ser169 and 10 weak Van der Waal interactions
(Figure 6.8) with Ser170, Lys187, Asn119, Ile121, Tyr183, Gly41, Ser43, Leu215,
184
C
CHAPTER-6
The results of present docking study reveals that all the three 11β HSD1 Inhibitors
have the potential to interact with crucial active site amino acid residues (Ile46,
Lys187, Asn119, Ser169, Ser170 and Tyr183). In view of potential of identified hits
185
CHAPTER-6
In view of good estimated activity, fit value, LibDock score and zero Lipinski
the novelty of the hits. The hits were compared with other ligands crystallized with
11β HSD1 inhibitors. SKDHSD2, SKDHSD3 showed low tanimoto similarity indices
of 0.136 and 0.108 respectively. Since both the compounds exhibited novelty they
Dharmendra et al.327
Experimental
Animals
Adult male wistar mice were used for the biological evaluation of SKDHSD2 and
SKDHSD3. Animals were purchased from Chaudhary Charan Singh University,
Hissar, Haryana. All the animals were approximately 9-10 week’s old and weighed
27-35 grams at the beginning of the experiment. Mice were housed in Central animal
186
CHAPTER-6
house of Banasthali Vidyapith with a light cycle of 12 hours light/ 12 hours dark and
allowed free choice access to standard rodent chow and water at temperature (22°C)-
controlled environment. All the experimental protocols were approved by IAEC (Reg.
Number: BU/BT/179/2011-12) and throughout the experiment the guidelines of
Committee for the Purpose of Control and Supervision of Experiments on Animals
(CPCSEA) and Institutional Animal Ethics Committee were followed. All animals
were cared in compliance with the Principles of Laboratory Animal Care and the
Guide.
Pre-Treatment Methodology
15 adult male wistar mice were divided into the following five groups (n = 3 per
group):
I. Normal control (5% DMSO)
II. STZ (40mg/kg)
III. Carbenoxolone (50mg/kg)
IV. SKDHSD2 (5mg/kg) and
V. SKDHSD3 (5mg/kg)
Group III, IV and V were treated for 3 days with carbenoxolone (50mg/kg),
SKDHSD2 (5mg/kg) and SKDHSD3 (5mg/kg) respectively before STZ diabetes
induction. Plasma samples were collected on 0th, 1st, 3rd and 7th day post STZ
injection, through the tail vein of mice and were immediately used for the estimation
of plasma glucose with a glucometer.
Statistical Analysis
All the experimental values were expressed as mean ± S.E.M and evaluated by
ANOVA.
Table 6.1: Effect of SKDHSD2 and SKDHSD3 on blood glucose level of previously treated
STZ induced albino wistar mice
188
CHAPTER-6
Figure 6.9: Effect of SKDHSD2 and SKDHSD3 on blood glucose level of previously treated
STZ induced albino wistar mice
Post Treatment
The hypoglycemic effect of SKDHSD2 and SKDHSD3 on the plasma glucose level is
shown in table 6.2. Plasma glucose levels were found to be reduced (P<0.05) in
carbenoxolone and SKDHSD2 and SKDHSD3 treated groups (153 mg/dl ± 1.15 vs.
134.33 mg/dl ± 2.40 and 145.33 mg/dl ± 1.78, respectively) in comparison to the STZ
induced control groups (Figure 6.10).
The result clearly shows that SKDHSD2 and SKDHSD3 are more potent inhibitor of
11β HSD1 than standard drug carbenoxolone. Undoubtedly the In-silico and In-vivo
work flow adopted in present study has yielded wonderful results.
Table 6.2: Effect of SKDHSD2 and SKDHSD3 on blood glucose level of STZ induced
albino wistar mice
189
CHAPTER-6
Figure 6.10: Effect of SKDHSD2 and SKDHSD3 on blood glucose level of STZ induced
albino wistar mice
Conclusion
In brief, two promising hits based on ligand pharmacophore based virtual screening
were identified having anti diabetic activity. The order of activity for test drugs and
carbenoxolone in both the models were found to be SKDHSD2 > SKDHSD3 >
carbenoxolone, which is in agreement to pharmacophore based virtual screening
results. It is noticeable that SKDHSD2 and SKDHSD3 produced the response at a
dose of 5mg/KG whereas a comparable response of carbenoxolone was observed at a
dose of 50 mg/KG, which proves that the identified hits are more potent than standard
drug. Eventually, anti diabetic assay validated the finding of virtual screening based
on ligand pharmacophore model. The results clearly show that In-silico methods are
powerful source for identification of novel chemical entity.
190
SUMMARY
SUMMARY
Summary
The present thesis entitled “In Silico Ligand Based Pharmacophore Design of GRA
and 11β-HSDI Inhibitors: An Approach to Database Mining and Identification of
New Lead Compounds”, enunciates exploration of essential structural requirements
for development of GRA and 11β HSDI inhibitor and recognition of promising lead
compounds having glucagon receptor antagonistic and 11β HSDI inhibitory activity.
In order to accomplish the aforesaid objectives, several steps like proper selection of
the compound datasets; statistical evaluation of the correlation between compound
dataset and 2D global descriptor; ligand pharmacophore model development and its
interpretation and validation; identification of lead compounds via virtual screening,
structure based pharmacophore model validation for the lead compounds; molecular
docking, and pharmacological in vivo biological evaluation of the lead compounds
were executed.
This whole objective of present work has been divided into three stages. The first
stage is comprised of the development of 2D QSAR/Hansch models for GRA and 11β
HSD1 inhibitors with best statistics. The second stage constitutes development of
three dimensional spatial arrangements and orientation model known as
pharmacophore model for GRA and 11β HSD1 inhibitors. The model with the best
statistical parameters, i.e., highest correlation, best RMS fit and proper difference
between total cost and null cost was subjected for mapping and validation studies.
After validation the model was used as pharmacophore query to identify lead
191
SUMMARY
compounds from compound libraries, the process called as virtual screening. The last
stage was the confirmation of the identified lead compounds as promising GRA and
11β HSD1 inhibitors has been exercised through structure based pharmacophore
model development and its mapping onto the lead compounds; molecular docking and
in vivo anti-diabetic evaluation of the lead compounds.
Chapter 1
The first part of the chapter 1 provides information on the glucagon and 11β HSD1
enzyme as promising targets, in the fight with type II diabetes mellitus. Moreover this
part also provides insight into three dimensional structures of glucagon and 11β
HSD1 enzymes, working strategies as anti-diabetic target, available drugs in the
market, need of development of new drugs as GRA and 11β HSD1 inhibitors and
their fate. Second part of chapter 1 explains the various drug design techniques such
as ligand based drug design and structure based drug design. The chapter gives a
generalized introduction of CADD and QSAR and its application in designing new
chemical entities. This chapter also describes the brief methodology of 2DQSAR,
ligand based pharmacophore modeling, virtual screening, SBDD and molecular
docking.
Chapter 2
This chapter contains a compilation of reports on GRA and 11β HSD1 inhibitors
synthesized and bio-assayed till date. Apart from this the QSAR studies executed till
date on GRA and 11β HSD1 inhibitors has also been reviewed. This chapter also
highlights the problems underlying behind the development of GRA and 11β HSD1
inhibitors as anti-diabetic targets and the scheme of work which has been followed in
present research endeavour.
Chapter 3
This chapter depicts the 2D global descriptor based QSAR model development by
establishing relationship between most significant descriptors and one GRA series and
two 11β HSD1 inhibitory series i.e. β alanine, isoserine and thiazole (GRA);
Pyrrolidine carboxamide Inhibitors (11β HSD1 inhibitor); and Bicyclo[2.2.2]
octyltriazole Inhibitors (11β HSD1 inhibitor).
192
SUMMARY
QSAR analysis (MLR, PLS and FFNN) of Series 1 antagonists (β alanine, isoserine
and thiazole) clearly ilustrates that the glucagon receptor antagonistic activity depends
on Verloop L, Dipole moment X and Log P parameters as determined by MLR
analysis and further validated by PLS and FFNN methods. Validation of models by
internal test set clearly shows the robust nature of the model. Multivariate analysis of
Series 2 (Pyrrolidine carboxamide inhibitors) as 11β HSD1 inhibitor demonstrated
dependency on dipole moment X component and log P towards the inhibition of 11β
HSD1 enzyme. The statistical outputs (r, r2, r2CV) of MLR, PLS and FFNN analysis
confirmed the robustness of the developed models. The multivariate analysis of Series
3 (Bicyclo[2.2.2] octyltriazole inhibitors as 11β HSD1 inhibitor) exhibited the
importance of Inertia moment length, Log P and VAMP Polarization XX as important
parameters for the desired 11β HSD1 inhibitory activity. The statistical outputs of
MLR, PLS and FFNN analysis revealed the predictability of the generated model. The
generated models can assist the rational design of promising drug entities with
enhanced GRA and 11β HSD1 inhibitory activity.
Chapter 4
Chapter 4 reports the 3D ligand based pharmacophore models (LBP) developed using
the structure activity based hypothesis generation approach (HypoGen). The LBP
models provides spatial orientations and arrangements of chemical features required
for interaction of receptor with the ligands.
3D LBP modeling of Series 1 (β alanine, isoserine and thiazole) entails the
significance of four pharmacophoric features i.e. one hydrogen bond donor (HBD),
one hydrophobe (HY), one negative ionizable (NI) and one ring aromatic (RA). The
statistical significance of the developed model was evaluated on the basis of
correlation coefficient, cost values and RMS. The developed model was cross
validated to avoid chance correlation by Cat scramble validation, internal and external
test set prediction. The external test set of 9 compounds resulted a squared correlation
coefficient value of 0.65 indicating the predictive ability of the model. As an
additional validation step some clinical trial drugs BAY-27-9955 and LY409021 were
mapped onto the generated pharmacophore model. BAY-27-9955 showed mapping
with two features (HBD and HY) and LY409021 mapped with three features (HBD,
HY and NI).
193
SUMMARY
Chapter 5
This chapter depicts the approach adopted to identify promising novel scaffolds with
glucagon receptor antagonistic activity and 11β HSD1 inhibitory property. The
validated LBP models were used as a 3D search query to screen NCI database. A
sequential VS procedure was applied, wherein the pharmacophore-based VS was
followed by estimated activity and fit value pre-filtration, and further drug likeness
screening to reduce the number of hits in each different screening step. The LBP
model of Series1, β alanine, isoserine and thiazole derivatives as GRA, was used to
screen NCI database to retrieve 3 potent hits, SKDGRA1, SKDGRA2 and SKDGRA3
with fits value ranging from 7.09-6.60. Similarly the LBP models of Series 2
(Pyrrolidine carboxamide inhibitors) and Series 3 (Bicyclo[2.2.2]octyltriazole
inhibitors), 11β HSD1 inhibitors produced one potent hit SKDHSD1 of fit value
7.893 and two potent hits SKDHSD2 and SKDHSD3 of fit values ranging 8.449-
8.228 respectively. Most importantly all the retrieved hits exhibited zero Lipinski
violation rule.
This chapter also explains the GRA and 11β HSD1 structure based pharmacophore
(SBP) model development. A SBP model helps in understanding the protein ligand
interaction within the protein structural framework, and helps in designing of ligands.
194
SUMMARY
The crystallized structure of glucagon receptor (4L6R) and 11β HSD1 enzyme
(2BEL) was used for the SBP models development. In order to validate the developed
SBP models; clinical trials drugs, BAY-27-9955 and LY409021 were mapped onto
the GRA model. BAY-27-9955 showed mapping with three features (2HY and
1HBD) and LY409021 mapped with four features (2HBA, 1HY and 1 HBD). The
retrieved hits SKDGRA1, SKDGRA2 and SKDGRA3 were also mapped onto the
SBP model. SKDGRA1 showed five feature mapping (2HBA, 2HBD and 1HY);
SKDGRA2 showed four features (2HBA, 1HBD and 1HY) mapping and SKDGRA3
also showed four features (2HBA, 1HBD and 1HY) mapping. Similarly,
carbenoxolone was also mapped onto the 11β HSD1 structure based model.
Carbenoxolone showed three features (2HBA and 1HY) mapping. The retrieved hits
of 11β HSD1 inhibitors, SKDHSD1, SKDHSD2 and SKDHSD3 were also mapped
onto the SBP model. SKDHSD1 showed three features (1HBA, 1HBD and 1HY)
mapping; SKDHSD2 also showed three features (1HBA, and 2HY) mapping and
SKDHSD3 showed four features (2HBA, and 2HY) mapping.
Chapter 6
This chapter deals with molecular docking and anti-diabetic evaluation of retrieved
lead compounds. Molecular docking is a technique to predict the binding mode of the
ligand molecules. Molecular docking consists of two essential components;
generation of promising ligand binding orientations known as sampling and prediction
of the binding strength for a specific ligand orientations known as scoring. In order to
understand the type of interaction between the chosen hits (obtained by the process of
virtual screening) and the targets glucagon receptor and 11β HSD1 enzyme, molecular
docking was performed using “LibDocker” protocol.
The three hits obtained from modeling of Series 1 (β alanine, isoserine and thiazole)
GRAs were docked into the active site of protein. SKDGRA1 docked well with a
LibDock score of 127.65 with 7 strong hydrogen bond interactions and 10 Van der
Waal interactions. SKDGRA2 showed Van der Waal interactions with a LibDock
score of 106.262. NSC 167411 showed three strong hydrogen bonding interactions
and four Van der Waal interactions with an eminent LibDock score of 126.279.
195
SUMMARY
196
REFERENCES
REFERENCES
References
2. Menge BA, Gruber L, Jørgensen SM, Deacon CF, Schmidt WE, Veldhuis JD et al
(2011) Loss of inverse relationship between pulsatile insulin and glucagon secretion
in patients with type 2 diabetes. Diabetes 60:2160-2168.
3. Bishop DB, O'Connor PJ, Desai J (2010) Diabetes In: Remington PL, Brownson R,
Wegner MV (ed) Chronic disease epidemiology and control, 3rd edn. American
Public Health Association, Washington, DC, pp 291--334.
4. Hex N, Bartlett C, Wright D, Taylor M, Varley D (2012) Estimating the current and
future costs of type 1 and type 2 diabetes in the UK, including direct health costs and
indirect societal and productivity costs. Diabetic Medicine 29: 855–62.
5. Kaveeshwar SA, Cornwall J (2014) The current state of diabetes mellitus in India.
Australas Med J 7: 45–48.
6. Danaei G, Finucane MM, Lu Y, Singh GM, Cowan MJ, Paciorek CJ et al (2011) National,
regional, and global trends in fasting plasma glucose and diabetes prevalence since 1980:
systematic analysis of health examination surveys and epidemiological studies with 370
country-years and 2.7 million participants. Lancet 378:31-40.
197
REFERENCES
12. Mayo KE, Miller LJ, Bataille D, Dalle S, Göke B, Thorens B, Drucker DJ (2003)
International union of pharmacology XXXV. The glucagon receptor family.
Pharmacol Rev 55:167-94.
13. Aronoff SL, Berkowitz K, Shreiner B, Want L (2004) Glucose metabolism and
regulation: beyond insulin and glucagon. Diabetes Spectrum 17:183-190.
14. Holst JJ (2010) Glucagon and glucagon-like peptides 1 and 2. Results Probl Cell
Differ 50:121-35.
16. Quesada I, Tuduri E, Ripoll C, Nada A (2008) Physiology of the pancreatic a-cell
and glucagon secretion: role in glucose homeostasis and diabetes. J Endocrin 199:5–
19.
17. Thorens B (2011) Brain glucose sensing and neural regulation of insulin and
glucagon secretion. Diabetes Obes Metab 1:82-88.
19. Rivera N, Everett-Grueter CA, Edgerton DS, Rodewald T, Neal DW, Nishimura
E et al (2007) A novel glucagon receptor antagonist, NNC 25-0926, blunts hepatic
glucose production in the conscious dog. J Pharmacol Exp Ther 321:743-752.
23. Wakelam MJ, Murphy GJ, Hruby VJ, Houslay MD (1986) Activation of two
signal-transduction systems in hepatocytes by glucagon. Nature 323:68–71.
25. Hoare SR (2005) Mechanisms of peptide and nonpeptide ligand binding to class B
G-protein-coupled receptors. Drug Discov Today 10:417–427.
27. Siu FY, He M, De Graaf C, Han, GY, Yang D, Zhang Z et al (2013) Structure of
the human glucagon class B G-protein-coupled receptor. Nature 499: 444-449.
28. Cho YM, Merchant CE, Kieffer TJ (2012) Targeting the glucagon receptor family
for diabetes and obesity therapy. Pharmacol Ther 135:247–278.
30. Bregman MD, Trivedi D, Hruby VJ (1980) Glucagon amino groups. Evaluation of
modifications leading to antagonism and agonism. J Biol Chem 255:11725–11731.
31. Unson CG, Macdonald D, Ray K, Durrah TL, Merrifield RB (1991) Position 9
replacement analogs of glucagon uncouple biological activity and receptor binding. J
Biol Chem 266:2763–2766.
33. Madsen P, Knudsen LB, Wiberg FC, Carr RD (1998) Discovery and structure–
activity relationship of the first non-peptide competitive human glucagon receptor
antagonists. J Med Chem 41:5150–5157.
199
REFERENCES
34. Cascieri MA, Koch GE, Ber E, Sadowski SJ, Louizides D, de Laszlo SE (1999)
Characterization of a novel, non-peptidyl antagonist of the human glucagon receptor.
J Biol Chem 274:8694-8697.
35. Chang LL, Sidler KL, Cascieri MA, De Laszlo S; Koch G, Li B (2001)
Substituted imidazoles as glucagon receptor antagonists. Bioorg Med Chem Lett 11:
2549–53.
36. Parker JC, McPherson RK, Andrews KM, Levy CB, Dubins JS, Chin JE (2000)
Effects of skyrin, a receptor-selective glucagon antagonist, in rat and human
hepatocytes. Diabetes 49:2079–2086.
39. Deeg MA, Abu-Raddad EJ, Tham LS, Fu H, Pinaire JA, Kelly RP (2011) Single
dose of the glucagon receptor antagonist LY2409021 reduces blood glucose in
healthy subjects and patients with type 2 diabetes mellitus. Diabetologia 54:S362.
40. Brekke OH, Sandlie I (2003) Therapeutic antibodies for human diseases at the
dawn of the twenty-first century. Nat Rev Drug Discov 2:52–62.
41. Brand CL, Rolin B, Jorgensen PN, Svendsen I, Kristensen JS, Holst JJ (1994)
Immunoneutralization of endogenous glucagon with monoclonal glucagon antibody
normalizes hyperglycemia in moderately streptozotocin-diabetic rats. Diabetologia
37:985-993.
42. Brand CL, Jorgensen PN, Svendsen I, Holst JJ (1996) Evidence for a major role
for glucagon in regulation of plasma glucose in conscious, nondiabetic, and alloxan-
induced diabetic rabbits. Diabetes 45:1076–1083.
200
REFERENCES
43. Nader N, Chrousos GP, Kino T (2010) Interactions of the circadian CLOCK
system and the HPA Axis. Trends Endocrinol Metab 21:277–286.
45. Wilcox G (2005) Insulin and Insulin Resistance. Clin Biochem Rev 26:19-39.
46. Adam TC, Epel ES (2007) Stress, eating and the reward system. Physiol Behav
91:449–458
47. Van Rossum EF, Russcher H, Lamberts SW (2005) Genetic polymorphisms and
multifactorial diseases: facts and fallacies revealed by the glucocorticoid receptor
gene. Trends Endocrinol Metab 16:445–450.
48. Vetere A, Choudhary A, Burns SM, Wagner BK (2014) Targeting the pancreatic
β-cell to treat diabetes. Nat Rev Drug Discov 13: 278–289.
49. Dickmeis T (2009) Glucocorticoids and the circadian clock. J Endocrinol 200:3–
22.
53. Klemcke HG, Kumar RS, Yang K, Vallet JL, Christenson RK (2003) 11β-
hydroxysteroid dehydrogenase and glucocorticoid receptor messenger rna expression
in porcine placentae: effects of stage of gestation, breed, and uterine environment.
Biol Reprod 69:1945-1950.
201
REFERENCES
54. Bates HE, Sirek A, Kiraly MA, Yue JT, Riddell MC, Matthews SG et al (2008)
Adaptation to intermittent stress promotes maintenance of beta-cell compensation:
comparison with food restriction. Am J Physiol Endocrinol Metab 295:947–958.
58. White PC, Mune T, Rogerson FM, Kayes KM, Agarwal AK (1997) 11β-
Hydroxysteroid dehydrogenase and its role in the syndrome of apparent
mineralocorticoid excess, Pediatr Res 41:25–29.
61. Wang M (2005) The role of glucocorticoid action in the pathophysiology of the
Metabolic Syndrome. Nutrition & Metabolism, 2:3
62. Tomlinson JW, Stewart PM (2007) Modulation of glucocorticoid action and the
treatment of type 2 diabetes. Best practice & Research. Clinical Endocrinology &
Metabolism 21:607-619.
202
REFERENCES
64. Hosfield DJ, Wu Y, Skene RJ, Hilgers M, Jennings A, Snell GP, Aertgeerts K
(2005) Conformational flexibility in crystal structures of human 11β hydroxysteroid
dehydrogenase type 1 provide insights into glucocorticoid interconversion and
enzyme regulation. J. Biol. Chem 280:4639-4648.
66. Nixon M, Wake DJ Livingstone DE, Stimson RH, Esteves CL, Seckl JR et al
(2012) Salicylate downregulates 11β-HSD1 expression in adipose tissue in obese
mice and in humans, mediating insulin sensitization. Diabetes 61:790-796.
68. Morris DJ, Souness GW, Latif SA, Hardy MP, Brem AS (2004) Effect of
chenodeoxycholic acid on 11 beta hydroxysteroid dehydrogenase in various target
tissues. Metabolism 53:811-816.
71. Alberts P, Nilsson C, Selen G, Engblom LO, Edling NH, Norling S et al (2003b)
Selective inhibition of 11 beta-hydroxysteroid dehydrogenase type 1 improves hepatic
insulin sensitivity in hyperglycemic mice strains. Endocrinology 144:4755-4762.
76. Herrling PL (2005) The drug discovery process Progress in Drug Research. In
Rudin M (ed) Birkhäuser Verlag, Basel, Switzerland.
77. Marton MJ, Derisi JL, Bennett HA, Iyer VR, Meyer MR et al. (1998) Drug target
validation and identification of secondary drug target effects using DNA microarrays.
Nature Medicine 4:1293-1301.
79. Martis EA, Somani RR (2012) Drug Designing, Discovery and Development
Techniques. In Basnet Purusotam (Ed.) Promising pharmaceuticals. Intech, Europe.
80. Bleicher KH, Bohm HJ, Muller K, Alanine AI (2003) Hit and Lead generation:
Beyond, High-Throughput Screening. Nature Rev Drug Discov 2:369-378.
83. Caldwell GW, Ritchie DM, Masucci JA, Hageman W, Yan Z (2001) The new pre-
preclinical paradigm: compound optimization in early and late phase drug discovery.
Curr Top Med Chem 1:353-366.
84. Singh R, Nagori BP, Soni B, Singh JV (2011) A Review: Clinical Trial and Data
Management. Pharmacophore 2:200-209.
204
REFERENCES
85. Snyder JP (1991) Computer-assisted drug design. Part I. Conditions in the 1980s.
Med Res Rev 11:641-62.
86. Beddell CR, Goodford PJ, Norrington FE, Wilkinson S, Woottonet R (1976)
Compounds designed to fit a site of known structure in human haemoglobin. Br J
Pharmacol 57:201–209.
87. Hansch C, Fujita T (1964) p-σ-π Analysis. A method for the correlation of
biological activity and chemical structure. J Am Chem Soc 86:1616-1626.
88. Abraham DJ (1998) Burger’s Medicinal Chemistry and Drug Discovery, Vol. 1,
John Willey & Sons. pp.533.
91. Richet MC (1893) Noté sur la Rapport entre la Toxicité et les Propriétés
Physiques des Corps. Compt Rend Soc Biol (Paris) 45:775-776
92. Lipnick RL, Overton CE (1986) Narcosis studies and a contribution to general
pharmacology. Trends Pharmacol Sci 7:161-164.
93. Kuzmin VE, Artemenko AG, Polischuk PG, Muratov EN, Hromov AI,
Liahovskiy AV (2005) Hierarchic system of QSAR models (1D-4D) on the base of
simplex representation of molecular structure. J Mol Model 11: 457-467.
94. Hammett LP (1970) Physical Organic Chemistry 2nd Ed. McGraw-Hill, New
York.
95. Taft RW (1952) Polar and steric substituent constants for aliphatic and o-benzoate
groups from rates of esterification and hydrolysis of esters. J Am Chem Soc 74:3120-
3128.
205
REFERENCES
102. Vedani A, Dobler M, Lill MA (2006) The challenge of predicting drug toxicity
in silico. Basic Clini Pharmacol Toxicol 99:195–208.
103. Myintz KZ, Xie XQ (2010) Recent advances in fragment-based QSAR and
multi-dimensional QSAR methods. Int J Mol Sci 11:3846-3866.
104. Verma J, Khedkar VM, Coutinho EC (2010) 3D-QSAR in drug design a review.
Curr Top Med Chem 10:95-115.
105. Kuzmin VE, Artemenko AG, Muratov EN, Polischuk PG, Ognichenko LN,
Liahovsky AV (2010) Virtual Screening and Molecular Design Based on Hierarchical
Qsar Technology, Recent Advances in QSAR Studies. Chall Adv Comp Chem Phy
8:127-176.
106. Ferreira MMC (2002) Multivariate QSAR. J Brazil Chem Soc 13:742-753.
206
REFERENCES
111. Hammett, LP (1935) Some relations between reaction rates and equilibrium
constants. Chem Rev 17:125.
112. Stanton DT (2012) QSAR and QSPR model interpretation using partial least
squares (PLS) analysis. Curr Comp Aid Drug Design 8:107-27.
113. Assafrao D, Mohallem JR (2007) The isotopic dipole moment of HDO. J Phy
40:85-91.
115. Seward JR, Cronin MT, Schultz TW (2002) The effect of precision of molecular
orbital descriptors on toxicity modeling of selected pyridines. SAR QSAR Environ
Res 13:325-40.
116. Singh PP, Sharma SB, Singh K (2010) Quantum chemical and energy descriptors
based QSAR study of triazine derivatives. J Chem Pharmaceutic Res 2:193-205.
117. Datta D (1991) Taft's substituent constants, σ* and σI, and Huheey's group
electronegativity. J Phy Org Chem 4:96–100.
119. Harper KC, Bess EN, Sigman MS (2012) Multidimensional steric parameters in
the analysis of asymmetric catalytic reactions. Nature Chem 4:366-374.
207
REFERENCES
121. Leo A, Hansch C, Elkins C (1971) Partition coefficients and their uses. Chem
Rev 71:525–616.
122. Reddy KN, Locke MA (1994) Relationships between molecular properties and
log p and soil sorption (koe) of substituted phenylureas: qsar models. Chemosphere
28:1929-1941.
123. Sun H (2004) A universal molecular descriptor system for prediction of LOGp,
LOGs, LOGb, and absorption. J Chem Inf Comp Sci 44:748–757.
124. Buiu AS, Seiman CD, Seiman DD, Borcan C, Mihailescu F (2012) Evaluation of
the pharmacological descriptors related to the induction of antidepressant activity and
its prediction by QSAR/QRAR methods. Mini Rev Med Chem 12:467-476.
125. Hua H (2009) Wiener and Schultz Molecular topological indices of graphs with
specified cut edges. Commun Math Comput Chem 61:643-651.
126. Ghorbani M (2013) Remarks on the balaban index Serdica. J Comput 7:25–34
128. Giri A, Mishra BB, Tripathi P, Agrawal PP, Singh PP (2011) Quantitative
structure-activity relationship of matrix metalloproteinase inhibitors based on
topological descriptors. J Chem Pharmaceutic Res 3:290-305.
130. Lowell HH, Brian M, Lemont BK (1991) The electrotopological state: an atom
index for QSAR. Quantitative Structure Activity Relationships 10: 43-51.
208
REFERENCES
134. Kubinyi H (1993) QSAR: Hansch Analysis and Related Approaches. Willey
Publishers, Weinheim, New York.
136. Mason JS, Good AC, Martin EJ (2001) 3D pharmacophores in drug discovery.
Cur Pharm Des 7:567-597.
137. Milne G W, Nicklaus MC, Wang S (1998) Pharmacophores in drug design and
discovery. SAR QSAR Environ. Res 9:23-38.
141. Stahura FL, Bajorath J (2005) New methodologies for ligand-based virtual
screening. Curr Pharmaceutic Design 11:1189-202.
142. Blundell TL, Patel S (2004) High-throughput X-ray crystallography for drug
discovery. Current Opin Pharmacol 4:490-496.
143. Ziarek JJ, Peterson FC, Lytle BL, Volkman BF (2011) Binding site identification
and structure determination of protein-ligand complexes by NMR. Methods Enzymol
493:241–275.
209
REFERENCES
144. Huang SY, Zou X (2010) Advances and Challenges in Protein-Ligand Docking.
Int J Mol Sci 11:3016–3034.
146. Grigoryan AV, Wang H, Cardozo TJ (2012) Can the Energy Gap in the Protein-
Ligand Binding Energy Landscape Be Used as a Descriptor in Virtual Ligand
Screening? Plos One 7:e46532
147. Kitchen DB, Decornez H, Furr JR, Bajorath J (2004) Docking and scoring in
virtual screening for drug discovery: methods and applications. Nature Reviews Drug
Discov 3:935-949.
148. Gurd RS, Flanders KC, Mar DH, Folz RJ, England RD, Coolican SA et al (1982)
Semisynthetic Derivatives of Glucagon: (Des-His')Ne-acetimidoglucagon and Na-
Biotinyl-N'-acetimidoglucagon. Biochemistry 21:4244-4251.
150. Gysin B, Trivedi D, Johnson DG II, Hruby VJ (1986) Design and synthesis of
glucagon partial agonists and antagonists. Biochemistry 25:8278-8284.
151. Wakelam MJO, Murphy GJ, Hruby VJ, Houslay MD (1986) Activation of two
signal-transduction systems in hepatocytes by glucagon. Nature 323:68-71.
152. Gysin B, Johnson DG, Trivedi D, Hruby VJ (1987) Synthesis of Two Glucagon
Antagonists: Receptor Binding, Adenylate Cyclase, and Effects on Blood Plasma
Glucose Levels. J Med Chem 30:1409-1415.
153. Unson CG, Gurzenda EM, Merrifield RB (1989) Biological activities of des-
His1[Glu9]glucagon amide, a glucagon antagonist. Peptides 10:1171–1177.
154. Post SR, Rubinstein PG, Tager HS (1993) Mechanism of action of des-His1-
[Glu9]glucagon amide, a peptide antagonist of the glucagon receptor system (saponin-
permeabilized hepatocytes/ glucagon-receptor interactions/ adenylyl cydase/ GTP/
Mg2+). Proc Natl Acad Sci USA 90:1662-1666.
210
REFERENCES
155. Hruby VJ, Azizeh BY, Tine BAV, Sturm NS, Hutzler AM, David C (1995) [des
Hisl, des Phe6, GIu9] glucagon amide: a newly designed "pure" glucagon antagonist,
Bioorg Med Chem Lett 5:1849-1852.
156. Hruby VJ, Azizeh BY, Shenderovich MD, Trivedi D, Li G, Sturm NS (1996)
Topographical Amino Acid Substitution in Position 10 of Glucagon Leads to
Antagonists/Partial Agonists with Greater Binding Differences. J Med Chem 39:2449-
2455
157. Hruby VJ, Azizeh BY Tine BAV, Trivedi D (1997) Pure glucagon antagonists:
biological activities and camp accumulation using phosphodiesterase inhibitors.
Peptides 18:633–641.
158. Azizeh BY, Ahn JM, Caspari R, Shenderovich MD, Trivedi D, Hruby VJ (1997)
The role of phenylalanine at position 6 in glucagon's mechanism of biological
action: multiple replacement analogues of glucagon. J Med Chem 40:2555–2562.
160. Hruby VJ, Sturm NS, Lin Y, Burley SK, Krstenansky JL, Ahn JM (1998)
Structure function studies on positions 17, 18, and 21 replacement analogues of
glucagon: the importance of charged residues and salt bridges in glucagon
biological activity. J Med Chem 41:2693–2700.
162. Cascieri MA, Koch GE, Ber E, Sadowski SJ, Louizides D, de Laszlo SE (1999)
Characterization of a novel, non-peptidyl antagonist of the human glucagon receptor.
J Biol Chem 274:8694–8697.
163. Hruby VJ, Trivedi D, Lin Y, Ahn JM, Siegel M, Mollova NN (2000) Design and
synthesis of conformationally constrained glucagon analogues. J Med Chem 43:1714-
1722.
211
REFERENCES
165. Hruby VJ, Ahn JM, Medeiros M, Trivedi D (2001) Development of potent
truncated glucagon antagonists J Med Chem 44:1372-1379.
166. Hruby VJ, Ahn JM, Gitu PM , Medeiros M, Swift JR , Trivedi D (2001) A new
approach to search for the bioactive conformation of glucagon: positional
cyclization scanning. J Med Chem 44:3109–3116.
167. Ladouceur GH, Cook JH, Doherty EM, Schoen WR, MacDougall MR,
Livingston JN (2002) Discovery of 5-Hydroxyalkyl-4-phenylpyridines as a new class
of glucagon receptor antagonists. Bioorg Med Chem Lett 12:461–464
168. Ling A, Plewe M, Gonzalez J, Madsen P, Sams CK, Lau J et al (2002) Human
glucagon receptor antagonists based on alkylidene hydrazides. Bioorg Med Chem Lett
12:663–666.
169. Smith RA, Hertzog DL, Osterhout MH, Ladouceur GH, Korpusik M, Bobko MA
et al (2002) Optimization of the 4-aryl group of 4-aryl-pyridine glucagon antagonists:
development of an efficient, alternative synthesis. Bioorg Med Chem Lett 12:1303–
1306.
170. Ladouceur GH, Cook JH, Hertzog DL, Jones JH, Hundertmark T, Korpusik M et
al (2002) Integration of optimized substituent patterns to produce highly potent 4-
aryl-pyridine glucagon receptor antagonists. Bioorg Med Chem Lett 12:3421–3424.
171. Ladouceur GH, Cook JH, Doherty EM, Giebel D, Schoen WR (2002) 4-
Phenylpyridine glucagon receptor antagonists: synthetic approaches to the sterically
hindered chiral hydroxy group. Tetrahedron Lett 43:4455–4458.
173. Unson CG, Wu CR, Jiang Y, Yoo B, Cheung C, Sakmar TP et al (2002) Roles of
specific extracellular domains of the glucagon receptor in ligand binding and
signaling. Biochemistry 41:11795–11803.
175. Hruby VJ, Ying J, Ahn JM, Jacobsen NE, Brown MF (2003) NMR solution
structure of the glucagon antagonist [deshis1, desphe6, glu9]glucagon amide in the
presence of perdeuterated dodecylphosphocholine micelles. Biochemistry 42:2825–
2835.
176. Kurukulasuriya R, Sorensen BK, Link JT, Patel JR, Jae HS, Winn MX et al
(2004) Biaryl amide glucagon receptor antagonists. Bioorg Med Chem Lett 14:2047–
2050
180. Shen DM, Zhang F, Brady EJ, Candelore MR, Dallas-Yang Q, Ding VDH et al
(2005) Discovery of novel, potent, and orally active spiro-urea human glucagon
receptor antagonists. Bioorg Med Chem Lett 15:4564–4569
181. Duffy JL, Kirk BA, Konteatis Z, Campbell EL, Liang R, Brady EJ et al (2005)
Discovery and investigation of a novel class of thiophene-derived antagonists of the
human glucagon receptor. Bioorg Med Chem Lett 15:1401–1405.
213
REFERENCES
182. Bolli GB, Fanelli CG, Porcellati F, Rossetti P (2006) Glucagon: The effects of its
excess and deficiency on insulin action. Nutri Metab Cardiovasc Dis 16:S28-S34.
183. Duffy JL, Cohen SM, Miller C, Kirk BA, Candelore MR, Ding VDH et al (2006)
Direct observation (NMR) of the efficacy of glucagon receptor antagonists in murine
liver expressing the human glucagon receptor. Bioorg Med Chem 14:1506–1517.
184. Liang R, Abrardo L, Brady EJ, Candelore MR, Ding V, Saperstein R et al (2007)
Design and synthesis of conformationally constrained tri-substituted ureas as potent
antagonists of the human glucagon receptor. Bioorg Med Chem Lett 17:587–592.
185. Qureshi SA, Yang X, Yates ML, Candelore MR, Feeney W, Hora D et al (2007)
Cloning and expression of canine glucagon receptor and its use to evaluate glucagon
receptor antagonists in vitro and in vivo. Eur J Pharmacol 555:8–16.
187. Lau J, Behrens C, Sidelmann UG, Knudsen LB, Lundt B, Sams C et al (2007)
New β-alanine derivatives are orally available glucagon receptorantagonist. J Med
Chem 50:113–128.
188. Kim RM, Chang J, Lins AR, Brady Ed, Candelore MR, Dallas-Yang Q et al
(2008) Discovery of potent, orally active benzimidazole glucagon receptor
antagonists. Bioorg Med Chem Lett 18:3701–3705.
189. Kodra JT, Jørgensen AS, Andersen B, Behrens C, Brand CL, Christensen IT et al
(2008) Novel glucagon receptor antagonists with improved selectivity over the
glucose-dependent insulinotropic polypeptide receptor. J Med Chem 51:5387–5396.
214
REFERENCES
192. Knappe TA, Linne U, Xie X, Marahiel MA (2010) The glucagon receptor
antagonist BI-32169 constitutes a new class of lasso peptides. FEBS Lett 584:785–
789.
193. Rafferty EP, Franklin Z, Irwin N, O'Harte FPM (2010) Stability and efficacy
studies in N-terminally truncated glucagon receptor antagonist peptides. Regulatory
Peptides 164:35–51.
194. Shen DM, Brady EJ, Candelore MR, Dallas-Yang Q, Ding VDH, Feeney WP et
al (2011) Discovery of novel, potent, selective, and orally active human glucagon
receptor antagonists containing a pyrazole core. Bioorg Med Chem Lett 21:76–81.
196. Filipski KJ, Bian J, Ebner DC, Lee ECY, Li JC, Sammons MF et al (2012) A
novel series of glucagon receptor antagonists with reduced molecular weight and
lipophilicity. Bioorg Med Chem Lett 22:415–420.
197. Kieffer TJ, Cho YM, Merchant CE (2012) Targeting the glucagon receptor
family for diabetes and obesity therapy. Pharmacol Therap 135:247–278.
199. Chung JYL, Steinhuebel D, Krska SW, Hartner FW, Cai C, Rosen J et al (2012)
Asymmetric synthesis of a glucagon receptor antagonist via friedel crafts alkylation of
indole with chiral α-phenyl benzyl cation. Org Process Res Dev 16:1832−1845.
215
REFERENCES
200. Guzman-Perez A, Pfefferkorn JA, Lee ECY, Stevens BD, Aspnes GE, Bian J et
al (2013) The design and synthesis of a potent glucagon receptor antagonist with
favorable physicochemical and pharmacokinetic properties as a candidate for the
treatment of type 2 diabetes mellitus. Bioorg Med Chem Lett 23:3051–3058.
202. O’Harte FPM, Franklin ZJ, Rafferty EP, Irwin N (2013) Characterisation of
structurally modified analogues of glucagon as potential glucagon receptor
antagonists. Mol Cell Endocrinol 381:26–34.
203. DeMong D, Dai X, Hwa J, Miller M, Lin SI, Kang L et al (2014) The discovery
of N-((2H-Tetrazol-5-yl)methyl)-4-((R)-1-((5R,8R)-8-(tertbutyl)-3-(3,5-
dichlorophenyl)-2-oxo-1,4-diazaspiro[4.5]dec-3-en-1-yl)-4,4-dimethylpentyl)
benzamide (SCH 900822): a potent and selective glucagon receptor antagonist. J Med
Chem 57:2601−2610.
216
REFERENCES
212. Sua X, Vickera N, Ganeshapillai D, Smitha A, Purohit A, Reed MJ, Potter BVL
(2006) Benzothiazole derivatives as novel inhibitors of human 11β-hydroxysteroid
dehydrogenase type 1. Mol Cell Endocrinol 248:214–217.
215. Yeh VSC, Patel JR, Kurukulasuriya R, Fung S, Monzon K, Chiou W (2006)
Synthesis and biological evaluation of heterocycle containing adamantane 11β-HSD1
inhibitors. Bioorg Med Chem Lett 16:5414–5419.
217
REFERENCES
217. Fujisawa Y, Nakagawa Y, Li R-S, Liu Y-J, Ohzeki T (2007) Diabetic pregnancy
in rats leads to impaired glucose metabolism in offspring involving tissue-specific
dysregulation of 11β-hydroxysteroid dehydrogenase type 1 expression. Life Sciences
81:724–731.
222. Yuan C, Jean DJS, Liu Q, Cai L, Li A, Han N et al (2007) The discovery of 2-
anilinothiazolones as 11β-HSD1 inhibitors. Bioorg Med Chem Lett 17:6056–6061.
223. Flyrena K, Bergquitsa LO, Castroa VM, Fotsch C, Johansson L, Jean DJS Jr. et
al (2007) Piperidine amides as 11β-hydroxysteroid dehydrogenase type 1 inhibitors.
Bioorg Med Chem Lett 17:3421–3425.
218
REFERENCES
229. Fotsch C, Bartberger MD, Bercot EA, Chen M, Cupples R, Emery M et al (2008)
Further studies with the 2-amino-1,3-thiazol-4(5h)-one class of 11β-hydroxysteroid
dehydrogenase type 1 inhibitors: reducing pregnane x receptor activity and exploring
activity in a monkey pharmacodynamic model. J Med Chem 51:7953–7967.
230. Xiang J, Wan Z-K, Li H-Q, Ipek M, Binnum E, Nunez J et al (2008) Piperazine
sulfonamides as potent, selective, and orally available 11β-hydroxysteroid
dehydrogenase type 1 inhibitors with efficacy in the rat cortisone-induced
hyperinsulinemia model. J Med Chem 51:4068–4071.
231. Lee JH, Kang NS and Yoo S-E (2008) Docking-based 3D-QSAR study for 11β-
HSD1 inhibitors. Bioorg Med Chem Lett 18:2479–2490.
219
REFERENCES
236. Yanga H, Doub W, Loub J, Leng Y, Shen J (2008) Discovery of novel inhibitors
of 11β-hydroxysteroid dehydrogenase type 1 by docking and pharmacophore
modeling. Bioorg Med Chem Lett 18:1340–1345.
238. Wanga H, Ruan Z, Lia JJ, Simpkins LM, Smirka RA, Wua SC et al (2008)
Pyridine amides as potent and selective inhibitors of 11β-hydroxysteroid
dehydrogenase type 1. Bioorganic & Medicinal Chemistry Letters 18:11, 3168–3172.
241. Sua X, Vicker N, Trusselle M, Halemb H, Cullerb MD, Potter BVL (2009)
Discovery of novel inhibitors of human 11β-hydroxysteroid dehydrogenase type 1.
Mol Cell Endocrinol 301:169–173.
220
REFERENCES
245. Morgan SA, Sherlock M, Gathercole LL, Lavery GG, Lenaghan C, Bujalska IJ et
al (2009) 11β-hydroxysteroid dehydrogenase type 1 regulates glucocorticoid-induced
insulin resistance in skeletal muscle. Diabetes 58:2506–2515.
246. Dzyakanchuka AA, Balazsa Z, Nasheva LG, Amreinb KE, Odermatt A (2009)
11β-Hydroxysteroid dehydrogenase 1 reductase activity is dependent on a high ratio
of NADPH/NADP+ and is stimulated by extracellular glucose. Mol Cell Endocrinol
301:137–141.
248. Siu M, Johnsona TO, Wanga Y, Naira SK, Taylora WD, Crippsa SJ et al (2009)
N-(Pyridin-2-yl) arylsulfonamide inhibitors of 11β-hydroxysteroid dehydrogenase
type 1: Discovery of PF-915275. Bioorg Med Chem Lett 19:3493–3497.
221
REFERENCES
222
REFERENCES
259. Huber R, Rosenstock J, Banarer S, Fonseca VA, Inzucchi SE, Sun W et al (2010)
The 11-beta-hydroxysteroid dehydrogenase type 1 inhibitor INCB13739 improves
hyperglycemia in patients with type 2 diabetes inadequately controlled by metformin
monotherapy. Diab Care 33:1516–1522.
260. Edgerton DS, Basu R, Ramnanan CJ, Farmer TD, Neal D, Scott M et al (2010)
Effect of 11β-hydroxysteroid dehydrogenase-1 inhibition on hepatic glucose
metabolism in the conscious dog. Am J Physiol Endocrinol Metab 298:1019-1026.
261. Wang M, Veniant MM, Hale C, Hungate RW, Gahm K, Emery MG et al (2010)
Discovery of a potent, orally active 11β-hydroxysteroid dehydrogenase type 1
inhibitor for clinical study: identification of (S)-2-((1S,2S,4R)-Bicyclo[2.2.1]heptan-
2-ylamino)-5-isopropyl-5-methylthiazol-4(5H)-one (AMG 221). J Med Chem 53:
4481–4487.
266. Tice CM, Zhao W, Krosky PM, Kruk BA, Berbaum J, Johnson JA et al (2010)
Discovery and optimization of adamantyl carbamate inhibitors of 11β-HSD1. Bioorg
Med Chem Lett 20:6725–6729.
223
REFERENCES
267. Ahn JH, Kim SH, Ramu R, Kwon SW, Lee SH, Kim CH et al (2010) Discovery
of cyclicsulfonamide derivatives as 11β-hydroxysteroid dehydrogenase 1 inhibitors.
Bioorg Med Chem Lett 20:1065–1069.
269. Ye XY, Chen SY, Nayeem A, Golla R, Seethala R, Wang M et al (2011) Design,
synthesis, and SAR studies of novel polycyclic acids as potent and selective inhibitors
of human 11β-hydroxysteroid dehydrogenase type 1 (11β-HSD-1). Bioorg Med Chem
Lett 21:6699–6704.
270. Liu Y, Fana Z, Dub H, Zhanga M, Menga Z, Chena L (2011) Direct regulation of
glucose and not insulin on hepatic hexose-6-phosphate dehydrogenase and 11β-
hydroxysteroid dehydrogenase type 1. Mol Cell Endocrinol 333:62–69.
273. Kim KY, Park JS, Rhee SD, Kang NS, Jung WH, Kim HY et al (2011) Anti-
diabetic and anti-adipogenic effects of a novel selective 11β-hydroxysteroid
dehydrogenase type 1 inhibitor, 2-(3-benzoyl)-4-hydroxy-1,1-dioxo-2H-1,2-
benzothiazine-2-yl-1-phenylethanone (KR-66344). Biochem Pharmacol 81:1028-35.
275. Shen J, Xia G, Xue M, Liu L, Yu J, Liu H et al (2011) Potent and novel 11β-
HSD1 inhibitors identified from shape and docking based virtual screening. Bioorg
Med Chem Lett 21:5739–5744.
224
REFERENCES
276. Wua SC, Yoon D, Chin J, Kirk KV, Seethala R, Golla R et al (2011) Discovery
of 3-hydroxy-4-cyano-isoquinolines as novel, potent, and selective inhibitors of
human 11β-hydroxydehydrogenase 1 (11β-HSD1). Bioorg Med Chem Lett 21:6693–
6698.
280. Ahn JH, Kim KY, Kwon SW, Kang SK, Lee JH, Bok JH et al (2011) Synthesis
and 11β hydroxysteroid dehydrogenase 1 inhibition of thiazolidine derivatives with an
adamantyl group. Bioorg Med Chem Lett 21:435–439.
glucose homeostasis and obesity in db⁄db mice. Clini Exp Pharmacol Physiol 39:69–
77.
285. Scott JS, deSchoolmeester J, Kilgour E, Mayers RM, Packer MJ, Hargreaves D
et al (2012) Novel acidic 11β-hydroxysteroid dehydrogenase type 1 (11β-HSD1)
inhibitor with reduced acyl glucuronide liability: the discovery of 4-[4-(2-
Adamantylcarbamoyl)-5-tert-butyl-pyrazol-1-yl]benzoic Acid (AZD8329). J Med
Chem 55:10136−10147.
286. Goldberg FW, Leach AG, Scott JS, Snelson W, Groombridge SD, Donald CS et
al (2012) Free-Wilson and structural approaches to co-optimizing human and rodent
isoform potency for 11β-hydroxysteroid dehydrogenase type 1 (11β-HSD1)
inhibitors. J Med Chem 55:10652−10661.
288. Wan ZK, Chenail E, Li HQ, Ipek M, Xiang J, Suri V et al (2012) Discovery of
HSD-621 as a potential agent for the treatment of type 2 diabetes. ACS Med Chem
Lett 4:118−123.
289. Kim KY, Parka JS, Rheea SD, Junga WH, Kang NS, Kim HY et al (2012) Anti-
diabetic and anti-adipogenic effects of a novel selective 11β-hydroxysteroid
dehydrogenase type 1 inhibitor in the diet-induced obese mice. Eur J Pharmacol
691:19–27.
226
REFERENCES
292. Scott JS, Gill AL, Godfrey L, Groombridge SD, Rees A, Revill J et al (2012)
Optimisation of pharmacokinetic properties in a neutral series of 11β-HSD1
inhibitors. Bioorg Med Chem Lett 22:6756–6761.
294. Shen J, Zhang L, Chen J, Ning M, Zou Q, Leng Y (2012) Synthesis and
evaluation of piperidine urea derivatives as efficacious 11β-hydroxysteroid
dehydrogenase type 1 inhibitors in diabetic ob/ob mice. Bioorg Med Chem Lett
22:2748–2752.
295. Ritter K, Bohme T, Engel CK, Farjot G, Gussregen S, Haack T et al (2013) 1,1-
Dioxo-5,6-dihydro-[4,1,2]oxathiazines, a novel class of 11ß-HSD1 inhibitors for the
treatment of diabetes. Bioorg Med Chem Lett 23:4685–4691.
297. Kim KY, Park SB, Jung WH, Kang NS, Park JS, Bae GH et al (2013) Anti-
diabetic and anti-inflammatory effect of a novel selective 11β-HSD1 inhibitor in the
diet-induced obese mice. Eur J Pharmacol 721:70–79.
298. Harno E, Cottrell EC, Keevil BG, DeSchoolmeester JD, Andersen H, Turnbull
AV et al (2013) 11-Dehydrocorticosterone causes metabolic syndrome, which is
prevented when 11β-HSD1 is knocked out in livers of male mice. Endocrinol
154:3599-3609.
299. Nair SK, Matthews JJ, Cripps SJ, Cheng H, Hoffman JE, Smith C et al (2013) N-
(Pyridin-2-yl) arylsulfonamide inhibitors of 11b-hydroxysteroid dehydrogenase type
1: Strategies to eliminate reactive metabolites. Bioorg Med Chem Lett 23:2344–2348.
304. Robl JA, Ye XY, Yoon DY, Chen SY, Nayeem A, Golla R et al (2014) Synthesis
and structure–activity relationship of 2-adamantylmethyl tetrazoles as potent and
selective inhibitors of human 11β-hydroxysteroid dehydrogenase type 1 (11β-HSD1).
Bioorg Med Chem Lett 24: 654–660.
305. Fardella CE, Lagos CF, Vecchiola A, Allende F, Fuentes CA, Tichauer JE et al
(2014) Identification of novel 11β-HSD1 inhibitors by combined ligand- and
structure-based virtual screening. Mol Cell Endocrinol 384:71–82.
310. Rousseau GG, Pierreux CE, Stafford J, Demonte D, Scott DK, Vandenhaute J et
al (1999) Antiglucocorticoid activity of hepatocyte nuclear factor-6. Proc Natl Acad
Sci U S A 96:8961–8966.
312. Muppalaneni NB, Rao AA (2012) Virtual screening of plant derived compounds
for aldose reductase inhibition using molecular docking. Bioinformation 8:980-983.
313. Serra JR, Jurs PC, Kaiser KL (2011) Linear regression and computational neural
network prediction of tetrahymena acute toxicity for aromatic compounds from
molecular structure. Chem Res Toxicol 14:1535-1545.
315. Riahi S, Pourbasheer E, Ganjali MR, Ali PN, Moghaddam Z (2008) QSPR study
of the distribution coefficient property for hydantoin and 5- arylidene derivatives. A
genetic algorithm application for the variable selection in the MLR and PLS methods.
J Chinese Chem Soc 55:1086-1093.
229
REFERENCES
319. Myint KZ, Xie XA (2010) Recent advances in fragment -based QSAR and multi-
dimensional QSAR methods. Int J Mol Sci 11:3846-3866.
320. Wu B, Rega MF, Wei J, Yuan H, Dahl R, Zhang Z et al (2009) Discovery and
binding studies on a series of novel pin1 ligands. Chem Biol Drug Design 73:369–
379.
230
REFERENCES
327. Gupta RK, Kumar D, Chaudhary AK, Maithani M, Singh R (2012) Antidiabetic
activity of Passiflora incarnata Linn. in streptozotocin-induced diabetes in mice. J
Ethnopharmacol 139: 801-806.
328. Eleazu CO, Eleazu KC, Chukwuma S, Essien UN (2013) Review of the
mechanism of cell death resulting from streptozotocin challenge in experimental
animals, its practical use and potential risk to humans. J Diab Metabol Disord 12:1-7.
231
ANEXURE-I
Pharmacophore modeling,
virtual screening, molecular
Tetrahedron
1. docking and biological Under review
Letters
evaluation to identify novel
anti-diabetic lead compounds
Simultaneous Modeling of 4-
(aminomethyl)-N-
methylbenzamide based
Glucagon Receptor Indian Journal of
2. Under review
Antagonist (GRA): Chemistry-B
Application of Equation and
Neural Network Based
QSAR
Quantitative Structure
Activity Relationships
Medicinal
(QSAR) of N6 substituted
3. Chemistry Published
adenosine receptor agonists
Research
as potential antihypertensive
agents
Quantitative Structure
Activity Relationship Studies
Journal of
5. of Topoisomerase I Published
Chemistry
Inhibitors as Potent Anti-
Breast Cancer Agents