You are on page 1of 177

Design, Synthesis and Evaluation of Some Novel

Pyrazoloquinolines as Anticancer Agents

Thesis submitted to

J.S.S. UNIVERSITY, MYSORE


in partial fulfillment of the requirements for the degree of

DOCTOR OF PHILOSOPHY.

by

Ms. Roshini Chandrasekhar, M.Sc.,

July, 2013

Under the Supervision of

Prof. M.J. Nanjan

J.S.S. College of Pharmacy


(Off Campus, JSS University, Mysore)
Udhagamandalam 643 001, The Nilgiris
Tamilnadu, India
ACKNOWLEDGEMENTS

I wish to express my deep sense of gratitude to Prof. M.J. Nanjan, Director, PG Studies
and Research/TIFAC CORE, J.S.S. College of Pharmacy. Ootacamund, under whose
active guidance, innovative ideas, constant inspiration, suggestions and encouragement
this research work was carried out.
I would like to thank Dr. B.Suresh, Vice Chancellor, JSS University, Mysore and Dr. K.
Elango, Principal, J.S.S. College of Pharmacy, Ootacamund, for providing all the
necessary facilities for the successful completion of my work.
I sincerely thank, Prof. S.P. Dhanabal, HOD, Dept. of Pharmacognosy and
Phytopharmacy for all his support. I am especially grateful to Dr. T.K. Praveen, for
helping me at every stage of my work with support and advice for the betterment of my
research.
I would like to thank Prof. S.Chandrasekaran, Dept. of Organic Chemistry, for giving me
an oppurtunity to carry out a part of my work at the Indian Institute of Sciences,
Bangalore and providing me with all the facilities for my research work. It is my pleasure
to thank Mr. Ganesh Venkataraman, Ms. Bandita Datta, Mr. Kishore Gadde, Ms. Divya
for their support and advice throughout my work in IISc., Bangalore.
My sincere thanks to Mrs. Mahalakshmi, Mr. Prashath Francis, Ms. Bincy, Mr. Ashish
Wadhwani, Mr. Viral Patel, Mr. Ankur Gupta, Ms. Bagyashree Kamble, Mr. Pankaj
Masih, Ms. Charitha, Ms. Santilna, my batch mates, Mr. Manikandan and Ms. Rubha,
my lab mates, for their encouragement.
I also would like to thank laboratory technicians and assistants, Mr. Gopal, Mr.
Subramanium and Mr. Madappa for their support and help given throughout my project.
I thank my dad, Mr. C.Chandraseharan, my mom, Mrs. Kumari Chandraseharan and my
sister, Reshna Chandrasekhar for being with me through thick and thin. Their
unconditional love and constant inspiration and blessing given throughout my work was,
no doubt, a great support to me.I am thankful to GOD ALMIGHTY with whose mercy I
have reached so far .

(Roshini Chandrasekhar)
LIST OF TABLES
Table 1: Typical properties of fragments, lead-like and drug-like molecules.

Table 2: Some of the patents related to pyrazoloquinolines.

Table 3: Data on 4,5-dihydro[1,2,3]oxadiaz-olo[3.4-a]quinolin-10-ium-3-olate


and its derivatives.

Table 4: Data on 4,5-dihydropyrazolo[1,5-a]quinolines.

Table 5: Docking parameters of selected ligands.

Table 6:Anticancer activity of 4,5-dihydro[1,2,3]oxadiazolo[3.4-a]quinolin-10-


ium-3-olate, its derivatives and 5FU against selected human cancer and
normal cell lines IC50 (µg/mL). (21-24)

Table 7:Anticancer activity of phenyl-4,5-dihydropyrazolo[1,5-a]quinoline-2-


carboxylate and its derivatives against selected human cancer cell lines
IC50 (µg/mL). (26-29)

Table 8:Anticancer activity of dimethyl 4,5-dihydropyrazolo[1,5-a]quinoline-2,3-


dicarboxylate and its derivatives against selected human cancer cell lines
IC50 (µg/mL). (31-34)

Table 9:Anticancer activity of (4,5-dihydropyrazolo[1,5-a]quinolin-3-yl)methyl


benzoate and its derivatives against selected human cancer cell lines
IC50 (µg/mL). (36-43)
LIST OF FIGURES
Figure 1: The progress of development of new molecular target therapy, gene to
drug.
Figure 2: Human topoisomerase IIα (1ZXM) from the Protetin Data Bank.
Figure 3: Mechanism of action of topoisomerase II.
Figure 4: The catalytic cycle of topoisomerase II. The individual steps of the
catalytic
cycle of topoisomerase II are shown. These are: (1) DNA binding; (2)
pre-strand passage cleavage/religation; (3) DNA strand passage; (4)
post-strand passage cleavage/religation; (5) ATP hydrolysis; and (6)
enzyme recycling.
Figure 5: Synthetic topoisomerase II inhibitors.
Figure 6: Some potent inhibitors of topoisomerase II.
Figure 7: Some biologically active sydnones.
Figure 8: Some important tetrahydroquinolines.
Figure 9: Some important pyrazoloquinolines.
Figure 10: Ramachandran Plot- 1ZXM.
Figure 11: Hydrogen bonding interactions of ANP, the co-crystallised ligand.
Figure 12: Hydrophobic interactions of salvicine-S in the binding site of topo IIα.
Figure 13: Hydrophobic interactions of the ligands in the binding site of topo IIα.
Figure 14: Hydrophobic interactions of the ligands in the binding site of topo IIα.
Figure 15: Hydrophobic interactions of the ligands in the binding site of topo IIα.
Figure 16: IR spectra of Compound 21.
Figure 17: MASS spectra of Compound 21.
Figure 18: 1H NMR spectra for Compound 21.
Figure 19: 13C NMR spectra for Compound 21.
Figure 20: IR spectra for Compound 22 .
Figure 21: MASS spectra for Compound 22.
Figure 22: 1H NMR spectra for Compound 22.
Figure 23: 13C NMR spectra for Compound 22.
Figure 24: IR spectra of Compound 23.
Figure 25: MASS spectra for Compound 23.
Figure 26: 1H NMR spectra for Compound 23.
Figure 27: 13C NMR spectra for Compound 23.
Figure 28: IR spectra for Compound 24.
Figure 29: MASS spectra for Compound 24.
Figure 30: 1H NMR spectra for Compound 24.
Figure 31: 13C NMR spectra for Compound 24.
Figure 32: MASS spectra for Compound 26.
Figure 33: 1H NMR spectra for Compound 26.
Figure 34: 13C NMR spectra for Compound 26.
Figure 35: NOESY spectra for Compound 26.
Figure 36: IR spectra for Compound 27.
Figure 37: MASS spectra for Compound 27.
Figure 39: 1H NMR for Compound 27.
Figure 40: 13C NMR for Compound 27.
Figure 41: IR spectra for Compound 28.
Figure 42: MASS spectra for Compound 28.
Figure 43: 1H NMR for Compound 28.
Figure 44: 13C NMR for Compound 28.
Figure 46: MASS spectra for Compound 29.
Figure 47: 1H NMR for Compound 29.
Figure 48: 13C NMR for Compound 29.
Figure 49: IR spectra for Compound 31.
Figure 50: MASS spectra for Compound 31.
Figure 51: 1H NMR for Compound 31.
Figure 52: 13C NMR for Compound 31.
Figure 53: IR for Compound 32.
Figure 54: MASS for Compound 32.
Figure 55: 1H NMR for Compound 32.
Figure 56: 13C NMR for Compound 32.
Figure 57: IR for Compound 33.
Figure 58: MASS for Compound 33.
Figure 59: 1H NMR for Compound 33.
Figure 60: 13C NMR for Compound 33.
Figure 61: IR for Compound 34.
Figure 62: MASS for Compound 34.
Figure 63: 1H NMR for Compound 34.
Figure 62: 13C NMR for Compound 34.
Figure 63: IR spectra for Compound 36.
Figure 64: MASS spectra for Compound 36.
Figure 65: MASS spectra for Compound 40.
Figure 66: 1H NMR for Compound 36.
Figure 67: 1H NMR for Compound 40.
Figure 68: 13C NMR for Compound 36.
Figure 69: 13C NMR for Compound 40.
Figure 70: NOESY for Compound 36.
Figure 71: NOESY for Compound 40.
Figure 72: IR spectra for Compound 37.
Figure 73: MASS spectra for Compound 37.
Figure 74: MASS spectra for Compound 41.
Figure 75: 1H NMR for Compound 37.
Figure 76: 1H NMR for Compound 41.
Figure 77: 13C NMR for Compound 37.
Figure 78: 13C NMR for Compound 41.
Figure 79: IR spectra for Compound 38.
Figure 80: MASS spectra for Compound 38.
Figure 81: MASS spectra for Compound 42.
Figure 82: 1H NMR for Compound 38.
Figure 83: 1H NMR for Compound 42.
Figure 84: 13C NMR for Compound 38.
Figure 85: 13C NMR for Compound 42.
Figure 86: IR spectra for Compound 39.
Figure 87: MASS spectra for Compound 39.
Figure 88: MASS spectra for Compound 42.
Figure 89: 1H NMR for Compound 39.
Figure 90: 1H NMR for Compound 42.
Figure 91: 13C NMR for Compound 39.
Figure 92: 13C NMR for Compound 42.
Figure 93: Graph representing the IC50 values of compounds 21-24.
Figure 94: Graph representing the IC50 values of compounds 26-29.
Figure 95: Graph representing the IC50 values of compounds 31-34.
Figure 96: Graph representing the IC50 values of compounds 36 to 43.
Figure 97 :(A) HeLa cells: Control cells and cells treated with 23 (B) MCF-7
cells: Control and cells treated with 27 (C) HeLa cells: Control cells
and cells treated with 28 (D) MCF-7 cells: Control and cells treated
with 41.
CONTENTS

Page no.

1. INTRODUCTION
1.1 Cancer and Cancer Therapy 1
1.2 Bioinformatics, Chemoinformatics and Drug Discovery 6
1.3 Some Novel Anticancer Targets 6
1.4 Topoisomerase Inhibitors 10
1.5 Type II Topoisomerase as a Drug Target 11
1.6 Sydnones and Sydnoquinolines 13
1.7 Quinolines and Tetrahydroquinolines 15
1.8 Cycloaddition Reactions 16
1.9 Pyrazoles and Pyrazoloquinolines 18
1.10 Review of Literature 21
2. SCOPE AND OBJECTIVES 63
3. EXPERIMENTAL
3.1 Chemicals and Reagents 69
3.2 Docking and Molecular Modeling Studies 70
3.3 Synthesis and Purification 73
3.4 In vitro Cell Proliferation MTT Assay 81

4. RESULTS AND DISCUSSIONS


4.1 Docking Studies 85
4.2 ADME Studies 91
4.2 Synthesis and Characterization 92
4.3 In vitro Anticancer Studies 148
5. SUMMARY AND CONCLUSIONS 156
6. BIBLIOGRAPHY 160
7. ANNEXURES 170
Introduction

1. INTRODUCTION

1.1 Cancer and Cancer Therapy

The first description of cancer dates back to 1600 BC and was found in an Egyptian

papyrus. It was regarded as an incurable disease until the 19th Century when surgical

removal was made possible. Radiation therapy started being used after 1960. Over time it

was realized that neither surgery nor radiation, or both in combination, was effective in

treating cancer as this could not control metastastic cancer.

Cancer is medically known as malignant neoplasm and is a group of various diseases,

all related to unregulated cell growth. Cancer is not caused by one agent, a virus or a

bacterium, that can be flushed or crushed. It is an intricate and potentially lethal

collaboration of genes gone awry, absence of some growth inhibitors or changing

hormones and epigenomes (Alice Park, 2013). In cancer, cells divide and grow

uncontrollably, forming malignant tumors, and invade nearby parts of the body. Cancer

may also spread to more distant parts of the body through the lymphatic system or

bloodstream. There are over 200 different known cancers that afflict humans

(cancerresearchuk.org). Cancer thus remains one of the most life threatening diseases. A

tremendous amount of research is, therefore, being carried out today for the prevention,

diagnosis and treatment of cancer.

The rationale for the use of conventional cytotoxic drugs is based on the theory that

rapidly proliferating and dividing cells are more sensitive to these compounds than the

normal cells. The hall mark of treatment for cancer has, therefore, been intravenous

cytotoxic chemotherapy. But then, cancer chemotherapy is a very difficult task. The

problems associated with chemotherapy are, non-specific toxicity and multi drug

1
Introduction

resistance (MDR) (who.int/mediacentre/factsheets/fs297/en). Many patients experience

the classic toxicities of alopecia, gastrointestinal symptoms and myelosuppression.

Advances that are taking place today in understanding the molecular mechanisms

underlying cancer development and progression are expected to lead to more efficacious

and less toxic drugs. Investigation are being conducted adapting a rational approach to

drug discovery in which a new generation of small molecules are designed and generated

to inhibit specific proteins or pathways abnormally expressed in cancerous cells. This

approach, termed Targeted therapy, is widely being applied today to discover new targets

and develop new anticancer drugs (Collins and Workman, 2006). Although traditional

cytotoxic chemotherapy remains the treatment of choice for many malignancies, targeted

therapies are now a component of treatment for many types of cancers, including breast,

colorectal, lung, and pancreatic cancers, as well as lymphoma, leukemia and multiple

myeloma.

Of the several new anticancer drugs approved by the U.S. Food and Drug Administration

(FDA) since 2000, 15 are targeted therapeutic agents compared to only five traditional

chemotherapeutic agents (centerwatch.com/patient/drugs/Druglist.html)

A large number of novel targets and approaches to anticancer therapy have emerged in

recent years. These approaches include targeting to the growth signal transduction

machinery within cancer cells, processes involved in cell invasion and metastatic spread,

apoptosis and tumor-related angiogenesis. Other approaches focus on tumor-specific

antigens, targeted poisons, stimulation of the immune system and gene therapy.

2
Introduction

Targeted therapies work by influencing the processes that control growth, division and

spread of cancer cells, as well as the signals that cause cancer cells to die naturally, the

way normal cells do when they are damaged or old. The development of new molecules

that bind to targets of biological interest is a breakthrough in anticancer research. The

knowledge of cell cycle mechanism and pathways of cancer pathogenesis has lead to

novel strategies and targets like Topoisomerases, Check Point Kinases (CPK’s), Cyclin

Dependent Kinases (CDK’s), Oncogenic Human Papiloma virus (HPV), ABC

transporters, etc.(Antony et al.,2009). One way of achieving this is through in silico

methods and Structure Based Drug Design (SBDD) methods.

Newer, safer and effective drugs, however, for cancer are yet to be explored due to the

fact that, drug discovery and development is cost and time intensive. It involves much

considerations right from Structure Based Drug Design (SBDD) or Ligand Based Drug

Design (LBDD) to synthesis, preclinical testing to clinical testing from cellular/molecular

to global effects on population. Only 20% of drug discovery projects are reported to lead

to clinical candidates of which only 10% compounds achieve registration. Analysis of the

reasons for this apparently low success is due to poor pharmacokinetics, toxicity and lack

of efficacy. Issues like target specificity and affinity must also be dealt for improving the

success rates (Shaikh et al.,2007). In recent years, continuous efforts have been made

both via computer aided drug design and reverse pharmacology using natural resources to

discover and develop drugs capable of specifically binding to the proteins/DNA

responsible for cancer pathogenesis (Spychala et al.,2009; Bonjean et al.,1998;

Yamamoto et al.,1990).

3
Introduction

SBDD has been widely used in ligand design and discovery (Kuntz 1992, Klebe 2000). In

particular, ligands can be identified among a large library of small molecules by virtual

screening. Each library molecule is docked into a binding site, then scored and ranked by

its ability to compliment the protein. High-ranking docked molecules are then tested

experimentally (Figure 1). The two critical parts of this process are docking to search

through plausible binding modes for candidate compounds and scoring to distinguish

ligands from nonbinding “decoys” (Collins and Workman, 2006).

Figure 1: The progress of development of new molecular target therapy, gene to drug
(Collins and Workman, 2006)

Not all small molecule leads are suitable as drugs. Profiling of potential leads against

multiple chemical, physicochemical and biological criteria has been adopted to select the

4
Introduction

best chemical starting point to maximize the probability of clinical success (Table 1)

(Davis et al., 2005).

In addition to target potency and selectivity, cellular efficacy and preliminary ADMET
properties are evaluated for promising compounds. This is called lead profiling. Lead
optimization involves addition of functional groups to increase the efficacy and
biological activity (Opera et al., 2001).

Table 1: Typical properties of fragments, lead-like and drug-like molecules. (Collins


and Workman, 2006)
Fragment Lead Drug

Physicochemical
Molecular weight (Da) <300 <400-450 <500
Lipophilicity (LogP) <3 <4 <5
H-bond donors (OH,NH) <3 <4-5 <5
H-bond acceptors (N,O) <3 <8-9 <10
Polar Surface areas NA NA <140-150Å2
Rotatable bonds NA <8 <10
Chemically reactive NA None present None present
groups

Biological
Target activity (IC50 or >>10-5M 10-6-10-7M 10-8-10-9M
K1) >0.3 >0.3 >0.3
Ligand efficiency (Kcal NMR or X-ray data SAR established SAR understood
atom-1)
SAR

ADMET NA Membrane permeable Bioavailable


Absorption NA Distributes to tissues Sufficient tumor
Distribution NA Low intrinsic levels
Metabolism/Excretion No common clearance Sufficient half life
Toxicology toxicophore Off-target Therapeutic window
pharmacology

5
Introduction

1.2 Bioinformatics, Chemoinformatics and Drug Discovery

Bioinformatics in drug discovery has been used for finding new targets. It predicts the

biological functions at the macromolecular level and considers gene as a fundamental

unit of genetic material. Chemoinformatics studies the relation between chemical

structures and properties of molecules, the interaction between small molecules and

macromolecules and the construction of chemical libraries. Today bioinformatics and

chemoinformatics overlap more and more (XU Jun et al., 2012).

Docking is a computational tool of structure–based drug design to predict the protein-

ligand interaction geometries and binding affinities. Docking program can be utilized to

discover and refine leads. Small molecule structures are bound with the target protein, the

energies of the resulting complexes are evaluated and those that show promising values

are tested as lead molecules. Here, the scoring method plays a very crucial role in

evaluating the molecules. The primary forces of attraction are the electrostatic

interactions. Among these, hydrogen bonds are of special interest (Jeffery et al., 1997,

Morimoto et al., 1991, Michael et al., 1997).

1.3 Some Novel Anticancer Targets

Some novel anticancer targets are, Tyrosine Kinase (TK) (Vlahovic and Crawford, 2008),

Farnesyl transferase (FTase) (Rowinsky et al., 1999), DNA topoisomerase.

Tyrosine Kinase (TK): TK are a family of mono-transmembrane -helix proteins

including membrane receptor tyrosine kinase and cytoplasm non-receptor tyrosine kinase.

The membrane receptor TK related to tumorigenesis includes epidermal cell growth

6
Introduction

factor receptor (EGFR) tyrosine kinase and vascular endothelial growth factor receptor

(VEGFR) tyrosine kinase, while the non-receptor tyrosine kinases include Src kinase and

Bcr-abl kinase, etc.

Farnesyltransferase (FTase): Ras protein, a low-molecular-weight GDP/GTP-binding

guanine triphosphatase encoded by Ras gene, plays a critical role in signal transduction of

cell growth and differentiation. In malignant transformation, invasion and spread of

cancer, Ras mutations or constitutive activation have been described. The continuous cell

growth signals are out of control causing cell differentiation and proliferation excessively

resulting in tumorigenesis.

DNA Topoisomerase: DNA is one of the most important molecules in cell biology.

Topoisomerases are the enzymes responsible for the changes in the DNA structure. These

enzymes accomplish this feat by either passing one strand of the DNA through a break in

the opposing strand (type I) or by passing a region of duplex from the same or a different

molecule through a double-stranded gap generated in a DNA (type II). Topoisomerases

are also known to relax only negative supercoils that relax supercoils of both signs or that

introduce either negative (bacterial DNA gyrase) or positive supercoils into the DNA

(reverse gyrase). They play an important role in regulating cellular processes such as

replication, transcription and chromosomal segregation by altering DNA topology. Most

type I DNA topoisomerases modify DNA topology in an ATP independent fashion by

creating single strand breaks in DNA whereas type II DNA topoisomerases do so in an

ATP-dependent fashion by creating double strand breaks in DNA (Schoeffler et al.,

2008). Among topoisomerases, topo II is a well-known anticancer target. Agents that

7
Introduction

target topo II are among the most effective anticancer drugs currently available for the

treatment of human cancers. Nevertheless, topo II based chemotherapy remains

associated with incidences of life-threatening toxic side effects like drug-induced

secondary malignancies. Intriguingly, previous studies suggest that suppressing topo II ,

a subtype of topo II, is responsible for the development of secondary malignancy

associated with etoposide treatment (Azarova et al., 2007). Drugs like etoposide and

doxorubicin may, however, have potent anticancer activity because they target another

subtype of topo II, namely topo II . Hence, agents specifically targeting topo II may

potentially be efficacious and safe chemotherapeutic drugs with a reduced risk of

treatment related to secondary malignancies (Toyoda et al., 2008).

Human Topoisomerase II (topo II ): Topo II is a homodimeric enzyme that requires

ATP for function (Figure 2). Topo II enzymes pass one duplex DNA through a second

duplex. To accomplish this reaction, the enzyme forms a reversible bond with 5’-

Phosphate of the DNA strand thereby creating a transient break in each phoshodiester

backbone. These breaks create a protein linked gate through which the second DNA

duplex can be pass (Bekhit et al., 2005). The whole process and functioning of topo II is

controlled by the hydrolysis of ATP to ADP and inorganic phosphate which leads to an

asymmetric retraction of Lys-378. This opens the enzyme-bridged middle gate of the

protein-DNA complex and movement of the T-segment from the upper cavity formed in

the ATPase domain into the lower cavity formed by the cleavage-reunion core (Figure 3

and 4). The inhibition of topo II leads to cell death and apotosis.

8
Introduction

Figure 2: Human topoisomerase II (1ZXM) from the Protetin Data Bank.

Figure 3: Mechanism of action of topoisomerase II

9
Introduction

Figure 4: The catalytic cycle of topoisomerase II. The individual steps of the catalytic
cycle of topoisomerase II are shown. These are: (1) DNA binding; (2) pre-strand passage
cleavage/religation; (3) DNA strand passage; (4) post-strand passage cleavage/religation;
(5) ATP hydrolysis; and (6) enzyme recycling.

1.4 Topoisomerase inhibitors

The topoisomerases were discovered in 1971 but it was not until 1980 that the

significance of these enzymes and their therapeutic potential were identified (Wang,

1985). Topoisomerases are the enzymes that regulate the overwinding and unwinding of

DNA. They control and modify the extracellular functions involved in replication,

recombination and transcription by catalyzing the passage of individual DNA strands

(topo I) or double helices (topo II) through one another. Topo I causes a break in a single

DNA strand, while topo II breaks both strands of DNA and requires ATP for activity

(Pinar et al., 2004). A variety of anticancer agents used in chemotherapy or those

10
Introduction

evaluated for clinical trials are known to inhibit topo I or topo II. Some synthetic Topo II

inhibitors are given in Figure 5

Figure 5: Synthetic topoisomerase II inhibitors.

1.5 Type II topoisomerase as a drug target

Eukaryotic Topo I and II and bacterial DNA gyrase (topo II) are the targets for a number

of clinically important drugs. Bacterial DNA gyrase is the target for quinolone synthetic

antibiotics. Quinolones potentially interact with the host, topoisomerase, and their affinity

to eukaryotic topoisomerase is thought to be relatively low. The precise mechanism of

quinolone-topoisomerase interaction is unknown. It is believed that the DNA and

11
Introduction

topoisomerase complex causes a conformational change in the "quinolone pocket", and

drugs bound at this site stabilize the complex. The stabilization of this complex inhibits

DNA rejoining and brings about the subsequent DNA damage. It is likely that quinolone

resistance is not transmitted by plasmids or transposons. Rare point mutations at the

quinolone pocket might decrease quinolone affinity and show drug resistance. Topo II is

essential for cell proliferation. In rapidly dividing mammalian cells, the total amount of

Topo II is dramatically increased. Hence topo II is a good target for cancer

chemotherapy. Drugs such as etoposide, doxorubicin, daunomycin and mitoxantrone are

widely used for systemic treatment of malignant neoplasms. Furthermore, the

chemotherapeutic regimens either include or are based on agents targeted to topo II. Topo

II poisons can be categorized into three groups; intercalators (such as doxorubicin and m-

AMSA), non-DNA-intercalators (etoposide,teniposide) and catalytic inhibitors of the

topoisomerase (aclarubicin and bisdioxopiperidines like ICRF-19363) (Figure 6). The

first two groups trap topo II on the DNA in a covalently bound state, often referred to as

the "cleavable complex". Catalytic inhibitors act on topo II without forming the cleavable

complex. It is not entirely clear what causes cell death following topoisomerase

stabilization. DNA damage provoked by topo II poisons has been shown in some cases to

result in apoptosis.

12
Introduction

Figure 6: Some potent inhibitors of topoisomerase II

1.6 Sydnones and Sydnoquinolines

Sydnones were first synthesized by Earl and Mackney in 1935. The structure 2 was

assigned to them (Scheme 1).

Later Baker, Ollis and Poole suggested the resonance stabilized 5-membered heterocyclic

system, 3, as the structure of sydnones (Baker et al.,1949).

13
Introduction

Sydnones are unique, dipolar heteroaromatic members of the general class of mesoionic

compounds. Chemically, they are 1, 2, 3-oxadiazolium-5-olates (Ollis et al.,1976). A

large number of sydnone derivatives have been synthesized (Kier et al., 1967;

Ackermann 1967; Pandey et al., 2006) and reported to possess a wide spectrum of

biological activities such as antiviral, antimicrobial, antiinflammatory, analgesic,

anthelmintic (Kalluraya et al., 2002), antitumor (Dunkley et al., 2003), free radical

scavenging (Mallur et al.,2007) and nitric oxide donor (Satyanarayana et al., 2002)

activity. The potential value of sydnones as biologically active substances is found in

their planar aromatic character, their relatively small size and the variation in electron

density around the ring. It is believed that the ionic resonance structures of sydnones

promote significant interactions with biological molecules. The stimulant drugs,

feprosidine and mesocarb, are substructures of sydnones imine in which the keto group of

sydnones (=O) is replaced by imino group (=NH). Some sydnones of biological

importance are shown in Figure 7.

14
Introduction

Figure 7: Some biologically active sydnones

1.7 Quinolines and Tetrahydroquinolines

Quinoline and tetrahydroquinoline derivatives have attracted a lot of interest due to their

applications as synthetic intermediates for a wide range of biologically active molecules.

Quinolines are an important scaffold in medicinal chemistry due to their occurrence in

natural products (Campbell et al.,1998) and drugs (Jenekhe et. al., 2001, Agrawal et al.,

1991, Jegou et al., 2001), polymer chemistry, electronics and optoelectronics for their

excellent mechanistic properties (Niwa et al.,1987). A large number of

tetrahydroquinolines have gained interest due to their biological activities. They are

found widely in nature. 2-Methyl-1,2,3,4-tetrahydroquinoline is present in the human

brain (Hadden et al., 2001). Dynemycin, a natural antitumor antibiotic based on

tetrahydroquinoline nucleus, the 2,4,6-trisubstituted tetrahydroquinoline, isolated from


15
Introduction

Martinella iquitosensis (Stewert, 1964), exhibits activity as a bradykinin antagonist and

with -adrenergic, histaminergic and muscarinic receptors. Some of the important

tetrahydroquinolines are shown in Figure 8.

Figure 8: Some important tetrahydroquinolines

Though sydnones are molecules which have been widely studied, sydnones fused with

tetrahydroquinolines have been studied only sparingly. There are only a few novel

synthetic routes for the synthesis of fused ring sydnoquinolines.

1.8 Cycloaddition Reactions

Cycloaddition reactions have always played an important role in the field of medicinal

chemistry. Woodward and Hoffmann first laid the fundamental basis for these concerted

16
Introduction

reactions (Woodward et al., 1965). Cycloaddition reactions belong to the class of

pericyclic reactions.

Pericyclic reactions are defined as the reactions that occur by a concerted cyclic shift of

electrons. This definition states two key points that characterize a pericyclic reaction.

Firstly, the reaction is concerted. In a concerted reaction, reactant bonds are broken and

product bonds are formed at the same time without intermediates. Secondly, these

reactions involve a cyclic shift of electrons and thus are characterized by a cyclic

transition state involving the bonds. The energy of activation of pericyclic reactions is

supplied by heat (Thermal Induction), or by UV light (Photo Induction). A cycloaddition

is a reaction between two compounds with bonds to form a cyclic product with two new

bonds as shown in Scheme 2.

Scheme 2

Sydnones undergo cycloaddition reaction with alkenes and alkynes to give pyrazolines

and pyrazoles, respectively. This is one of the most important synthetic applications of

sydnones. Cycloaddition with alkynes delivers pyrazoles via a

cycloaddition/retrocycloaddition with the expulsion of carbon dioxide (Scheme 3).

Recent years have seen a growing interest in pyrazole chemistry from both industrial and

academic standpoint.

17
Introduction

1.9 Pyrazoles and Pyrazoloquinolines

Pyrazoles and their derivatives have gained considerable importance over the years due to

their wide range of biological activities like antibacterial (Tandon et al., 2005), anticancer

(El-Deeb et al., 1962), antiinflammatory (Godaginamath et al., 2003), antitumor (Salgin-

Goksen et al., 2007), anticonvulsant(Onkol et al., 2004), etc. Pyrazoloquinolines,

quinolines fused with pyrazoles, are an important class of compounds which have been

studied extensively. Pyrazoloquinolines are known to possess anticancer (Mark et al.,

1995), antipsychotic (Shu-Wei Yang et al., 2012) and antiviral among other activities

Some biologically active pyrazoloquinolines are shown if Figure 9.

18
Introduction

19
Introduction

Though pyrazoloquinoline drugs are not in the market, KW-2170 and PD-115934 are two

such topoisomerase inhibitors which have reached phase II clinical trials. KW-2170 is a

pyrazoloacridine which is a topo II inhibitor of synthetic origin whereas PD-115934 is a

dual topo I and topo II inhibitor (Avendano and Menendez, 2008).

Although pyrazoloquinolines have been widely studied, reports on 4,5-

dihyropyrazolo[1,5-a]quinolines are very limited.

In the present investigation it was proposed, therefore, to design some fused ring

sydnoquinolines and pyrazolo[1,5-a]quinolines for the target, Topoisomerase II (pdb id:

1ZXM). The designed molecules will be synthesized and evaluated for their

antiproliferative activity on cervical cancer (HeLa), larynx epidermoid carcinoma (HEp-

2) and breast cancer (MCF-7) cell lines. The results of the in vitro studies and docking

studies will then be correlated to establish a structure-activity relationship for the

molecules.

20
Review of literature

1.10 Review of literature

The investigations carried out by earlier workers on the synthesis, pharmacological

properties and structure-activity relationships of sydnoquinolines and pyrazoloquinolines

including some of the related patents will now be reviewed for the period 1977 to 2013.

Chemistry and Pharmacology of Sydnoquinoline Derivatives.

Sydnones are a class of compounds that have been widely studied due to their diverse

biological properties and their betain like character. The synthesis of these compounds

was first carried out by the dehydration of N-nitroso-N-aryl (or –alkyl) glycines (Baker et

al.,1950). Nassoy et al., (2013) have carried out the synthesis and cycloaddition of

sydnones to obtain oxetanyl-substituted sydnones 2 (Scheme 4). They focused on

oxetanes due to their favourable pharmacological properties. The synthesized oxetane

substituted sydnones were further subjected to cycloaddition reaction with commercially

available alkynes (Scheme 5). Microwave assisted cycloaddition was found to result in

higher yields in lesser times with good regiocontrol. A higher yield of the isomer 4b was

observed.

Scheme 4

21
Review of literature

Scheme 5

They also studied the intramolecular cycloaddition using microwave method (Scheme

6)which revealed that all the pyrazoles are formed in good yields in short times.

Scheme 6

The authors thus effectively proved that oxetane-substituted sydnones are effective

precursors for the synthesis of pyrazoles via cycloaddition. They also successfully

synthesized spiro-fused intermediates (6) via intramolecular cycloaddition of sydnones

and alkynes.

22
Review of literature

Nitinchandra et al.,(2012) have synthesized a group of sydnones containing new mannich

21 and schiff’s bases 20 and evaluated them for their analgesic and antiinflammatory

activity (Scheme 8). The compounds containing schiff’s bases showed surprisingly good

antiinflammatory and analgesic activity on the introduction of electron donating groups.

In the case of compounds containing mannich bases significant antiinflammatory and

analgesic activity were observed and the presence of a piperidine or morpholine ring was

found to enhance the activity.

Scheme 7

23
Review of literature

Scheme 8

Chandrasekhar et al.,(2011) have synthesized 4, 5-dihydro[1,2,3] oxadiazolo [3,4-

a]quinolin-10-ium and its fluoro derivative 28 (Scheme 9). This preliminary

24
Review of literature

communication reports the synthesis of fused ring quinolines. There are only a very few

reports on the synthesis of these type of molecules. The authors have also published a

review on the synthesis and biological activites of sydnones and their derivatives

(Chandrasekhar et al.,2012).

Scheme 9

Tegginamath et al.,(2011) have reported a one-pot synthesis of sydnones fused with

coumarin ring via thiazoles (Scheme 10). The reaction was carried out in presence of

reusable silica sulphuric acid. The synthesized compounds 32a-r were tested for their α-

amylase inhibition and DNA cleavage properties. The compounds with electron

withdrawing groups exhibited stronger α-amylase inhibitory activity when compared to

the control enzyme inhibitor. The in vitro studies revealed that among the synthesized

molecules chlorine substituted derivatives exhibit good α-amylase inhibitory and also

DNA cleavage activity.

25
Review of literature

Scheme 10

Taj et al.,(2011) have utilized 3-aryl sydnones and exploited their ability to undergo

electrophilic substitution at the 4th position in order to couple benzotriazole,

benzothiazolin-2-thione, morpholine and diphenylamine to 4-bromo-3-arylsydnones to

obtain the desired molecules 35, 37, 39 and 41 (Scheme 11). The synthesized molecules

were studied for their potential antitubercular activity. The compounds containing

electron withdrawing groups exhibited excellent inhibition at a concentration of less than

or equal to 5μg/mL.

26
Review of literature

Scheme 11

Zhang et al.,(2002) have efficiently synthesized 3-(3-hydroxypropyl)sydnones 50 and

studied their intramolecular electrophilic aromatic substitution (Scheme 12). They

exploited the intramolecular Friedel-Crafts acylation reaction in order to obtain seven

membered fused ring sydnones 51. The Friedel-Crafts reaction was carried out in the

presence of H2SO4 or BF3.Et2O.

27
Review of literature

Scheme 12

Butkovioc et al.,(2011) have synthesized a series of stilbene-sydnones from methyl

anthranilate (Scheme 13) and evaluated them for their cytotoxic properties in five cell

lines, namely HeLa (cervical carcinoma), MCF-7 (breast carcinoma), SW 620 (colon

carcinoma), MiaPaCa-2 (pancreatic carcinoma) and H 460 (lung carcinoma). All the

compounds, however, showed low antiproliferative effect. An interesting observation

made was that though trans stilbenes generally exhibit high biological activity, the trans

derivatives synthesized by them showed low activity when compared to the cis

derivatives. This method was found to be convenient for the synthesis of both

diastereomers.

28
Review of literature

Scheme 13

Deshpande et al.,(2010) have synthesized sydnones containing a chalcone moiety

(Scheme 14) and evaluated them for their antiinflammatory, analgesic and antibacterial

properties. The synthesized molecules exhibited low antibacterial and antiinflammatory

activities.

29
Review of literature

Dunkley et al.,(2003) have synthesized N-(4’-substituted-3’-nitrophenyl)sydnones 69a-d

(Scheme 15) and evaluated them for their anticancer activity. These molecules were

synthesized based on the work of Grynberb et al., who synthesized a series of 4’-

substituted-3’-nitrophenylsydnones, evaluated them for their anticancer activity and

found that the 4’- chloro and 4’-pyrrolidino compounds had good activity when

compared to others.

Scheme 15

The authors also synthesized various substituents of Grynberg’s compounds and studied

the role of inductive effect or the ring size but were not successful in suggesting the

actual mechanism for the activity of these compounds. However, based on the findings of

Fliedner, Dunkley and coworkers suggested a mechanism for the anticancer activity of

sydnones. Fliedner has found that the pKa of 3-sydnonyl acetic acid and nitroacetic acid

are similar which suggests that the 3-sydnonyl moiety has electron withdrawing character

as the nitro group. The nitro group and the sydnonyl moiety thus act as o,p-directing

30
Review of literature

group in a normal nucleophilic aromatic substitution. They suggested that the

nucleophilic groups on DNA may get attached to the sydnones and get deactivated.

Satyanarayana et al.,(1995) have synthesized 3-phenylsydnones (Scheme 16) and

evaluated them for their antiinflammatory, antiarthritic and analgesic activity. The

antiinflammatory activity was evaluated using carrageenen induced paw edema method.

All the synthesized molecules exhibited low activity when compared to the standard,

phenylbutazone. These compounds, when evaluated for their antiarthritic activity in

adjuvant induced arthritis model in rats, exhibited significant activity. The compounds

were also more active compared to the standards, phenylbutazone and indomethacin,

though the bromo derivative showed activity similar to phenylbutazone. The compounds

were also evaluated for analgesic activity using acetic acid induced assay in mice. The

fluoro derivative exhibited very high activity when compared to the standard, aspirin. The

authors also evaluated the active compounds for their ulcerogenic effect as gastric effects

are the major side effects of antiinflammatory drugs. All the compounds were found to be

ulcerogenic though not significantly.

31
Review of literature

Senff-Ribeiro et al.,(2004) have studied the cytotoxic and antiproliferative activity of MI-

D (72) on human melanoma cell lines and found that MI-D could be potent against

human melanoma cells.

Sydnones as synthons for other biologically active molecules

Latthe et al.,(2011) have reported the synthesis of 1,2-diaza-five membered heterocyclic

systems. They synthesized 4-(hydrazinocarbonyl)phenylhydrazine 74 and used it as a

synthon for the synthesis of bismesoionic compounds (Scheme 17). The synthesized

molecules were then evaluated for their antibacterial activity. They reported the synthetic

utility of the bisfunctional compound, for the synthesis of novel bis azide–4-

(azidocarbonyl)phenyl azides 75, that were a synthetic challenge till then. The azides

were then used for the synthesis of compounds 81a-c and 82a-c. These compounds were

found to show moderate antibacterial activity. All the phenylcarbamates 77a-c showed

considerably good antifungal activity when compared to the standard, griseofulvin.

32
Review of literature

Scheme 17

Foster et al.,(2011) have synthesized azine-substituted pyrazoles via the cycloaddition of

sydnones (Scheme 18). This synthesis is novel as not many N-azine-substituted sydnones

have been reported in literature. The authors studied the cycloaddition of sydnones with

alkylboronates for the synthesis of pyrazole boronic acid derivatives 88-91. These studies

are of importance due the the boronate motif. The authors have thus efficiently

33
Review of literature

demonstrated that alkyne/sydnone cycloaddition reactions are a direct and convenient

method for the preparation of azine-substituted pyrazoles.

34
Review of literature

Scheme 18

35
Review of literature

Wu et al.,(2010) have reported the synthesis of 2H-indazoles via the [3+2] cycloaddition

of arynes to sydnones (Scheme 19). They carried out the cycloaddition reaction with

different precursors of arynes and observed that the reaction proceeded to completion

with good yields with single products 107. The reaction conditions were mild. A detailed

study on the mechanism required was also in progress by the authors.

Sateesha Rai et.al.,(2008) have synthesized a series of novel 1-aryl-3-(5-nitro-2-thienyl)-

4-aroyl-pyrazoles 121 (Scheme 22)via 1,3-dipolar cycloaddition of 3-arylsydnones 113

(Scheme 20) with 1-aryl-3-(5-nitro-2-thienyl)-2-propyn-1-ones 118 (Scheme 21). The

synthesized molecules were evaluated for their antibacterial (Escherichia coli ATCC-

25922, Staphylococcus aureus ATCC-25923, Pseudomonas aeruginosa ATCC-27853,

recultured Bacillus subtilis) and antifungal activity [(Candida albicans (NCIM No.

3100)] by serial dilution method. In an attempt to increase the activity of the molecules

they introduced 5-nitrofuran and 5-nitrothiophene moiety. The results revealed that some

of the tested compounds had good activity. The compounds with methyl and chloro

derivatives were found to show excellent antibacerial activity. The compounds with

methyl, chloro and methoxy substituents were found to show exceptionally good

antifungal activity when compared to the standard, fluconazole.

36
Review of literature

Scheme 20

Scheme 21

37
Review of literature

Scheme 22

Ganesha Rai et al.,(2006) have synthesized a series of 1-aryl-3-(5-nitro-2-furyl)-4-

aroylpyrazoles 124 via 1,3-dipolar cycloaddition reaction of 3-arylsydnones 123 and α,β-

acetylenic ketones 122 containing nitrofuran moiety (Scheme 23). Although 1,3-dipolar

cycloaddition of sydnones is well known and well studied, the authors felt that less

attention has been given to the regiochemistry of 1,3-dipolar cycloaddition reaction with

more complex dipolarophiles. Hence, they made an attempt to study the cycloaddition

reaction with more complex dipolarophiles like 1-aryl-3-(5-nitro-2-furyl)propynones 122.

They found that the reaction of these dipolarophiles with 3-arylsydnones resulted in

regiospecific 1-aryl-3-(5-nitro-2-furyl)-4-aroylpyrazoles in good yields. In order to study

the regiospecificity in more detail the authors used different para substituted aryl

compounds and in all the cases the reactions were found to be highly regioselective. They

38
Review of literature

found the formation of single products and this was confirmed using X-ray

crystallographic studies.

Scheme 23

Chemistry and Pharmacology of Pyrazoloquinoline derivatives.

Faidallah et al.,(2013) have studied the DNA binding property of tetrahydroquinolines

125, tetrahydropyrimidino[4,5-b]quinolines 126 and tetrahydropentaazacyclopenta[a]

anthracenes 127. All the synthesized compounds displayed good antitumor activity and

good DNA binding activity. Tricyclic tetrahydropyrimidino [4,5-b]quinolines, however,

showed better activity.

Jun Ya Kato et al.,(2013) have reported a novel method for the synthesis of pyrazolo[1,5-

a]quinolines 130 under transition metal free conditions (Scheme 24). This method

involved the synthesis of pyrazoloquinolines via a combination of aromatic nucleophilic

substitution and knovenagel condensation.

39
Review of literature

Scheme 24

Mishra (2012) has studied the 2D- structure-activity relationship (QSAR) of 2,5-

dihydropyrazolo[4,3-c]quinolines on the inhibition of phosphodiesterase-4 (PDE-4). The

models were checked for the observed biological activity and the predicted activity. The

biological activities of the selected compounds were shown to be due to the

hydrophobicity, electrostatic and topological properties of the molecules. The authors

concluded that the increase PDE-4 inhibitory activity of 2, 5-dihydropyrazolo [4, 3-c]

quinoline-3-one derivatives was due to the presence of groups contributing to flexibility

in chain length and lipophilicity of molecule.

Chang et al.,(2012)have efficiently synthesized two regioisomers of 2-arylpyrazolo[3,4-

c]quinolin-4(5H)-ones 134 and 2-arylpyrazolo[4,3-c]quinolin-4(5H)-ones 135 from 3-

arylsydnones 133, ethyl 3-bromopropynoate, and 2-aminophenylboronic acid pinacol

40
Review of literature

ester using Pd(PPh3)4 as catalyst (Scheme 25). This efficient one-pot synthesis

methodology involved 1,3-dipolar cycloaddition, Suzuki coupling reaction and

intramolecular cyclization.

Abu-Hashem et al.,(2012) have synthesized a series of 2-

hydrazinyltetrahydropyrimido[4,5-b]quinolin-4(3H)-ones 138 by desulphurization of S-

and N-dimethyl derivatives 137 with hydrazine hydrate (Scheme 26).

These molecules on reacting further with malonitrile, carbondisulphide, potassium

thiocyanate, pthalic achydride and aromatic aldehydes gave 3,5-di

aminopyrazolopyrimido[4,5-b]quinolines 139, triazolotetrahydropyrimido[4,5-

b]quinolines 140, aminotriazolopyrimido[4,5-b]quinolines 141,

aminopthalimidopyrimido[4,5-b]quinolines 142 and N-arylidene hydrazinepyrimido[4,5-

b]quinoline derivatives, respectively 146 (Scheme 27). The synthesized molecules were

41
Review of literature

evaluated for their antitumor potential and a few of them proved to be potent antitumor

agents.

Scheme 27

42
Review of literature

The in vitro cytotoxicity studies of the synthesized molecules revealed that the

pyrimidoquinoline when introduced into the basic scaffold helped improve the antitumor

activity. Compounds 139, 140, 141 and 146d showed cytotoxicity against KB, MGC-803

and MCF-7 cell lines and compound 139 showed potent cytotoxicity against CNE2

cancer cell lines. The structure-activity relationship revealed that the presence of 3,5-

diaminopyrazolo, 2-amino-1,3,4-triazolo, 1,3,4-triazolo and triazolothiazolidine moieties

linked to the pyrimido[4,5-b]quinolines enhanced the cytotoxicity of the molecules.

Reis et al.,(2011) have synthesized and evaluated some novel 2-(benzo[d]thiazol-2-yl)-8-

substituted-2H-pyrazolo[4,3-c]quinolin-3(5H)-ones 151 (Scheme 28) for their anticancer

activity against MDA-MB-435, HL-60, HCT-8 and SF-295 cell lines. The results

revealed that compounds 151b and 151c exhibited good anticancer activity on all three

cell lines with IC50 values less than 5μg/mL. Molecular modeling studies were also

carried out by these authors using Osiris programs to evaluate the electronic properties,

hydrogen bonding, molecular weight and theoretical toxicity properties.

Scheme 28

43
Review of literature

Hemolytic assay studies revealed that none of the molecules were capable of causing

hemolysis in mouse erythrocytes even at high concentrations.

Lenzi et al.,(2011) have designed and synthesized 2-phenyl and 2-methylpyrazolo[3,4-

c]quinolines-4-one 155, 156 (Scheme 29) and 4-amine derivatives 163, 167 (Scheme 30)

as adenosine receptor antagonists. The synthesized molecules were evaluated for their

ability to displace specific [3H]DPCPX, [3H]ZM241385 and [125I]AB-MECA binding

from cloned hA1, hA2A and hA3 receptors, respectively. The results revealed that the

synthesized molecules showed A1 receptor affinity and selectivity. Molecular docking

studies were also carried out in order to define the structural features of the binding (pdb

code: 3EML). The docking studies revealed that the introduction of the functional group

at the 6th position leads to enhanced affinity towards the A1 receptor and also selectivity.

44
Review of literature

45
Review of literature

Imbriglio et al.,(2011) have worked on a series of amino-anthranilic acid derivatives as a

new class of low serum-shifted high affinity full agonists of the human orphan G-protein-

coupled receptor, GPR109a, with improved ADME profile. They designed a series of

GPR109a receptor antagonists. A few pyrazoloquinolines based on these series of

compounds were found to show a 10-fold reduction of the serum shift.

The anthranilic acid derivatives were found to have a 10,000 fold shift of serum-potency,

excellent in vitro profile and modest ADME properties. Considering the importance of

the anthranilic acid moiety and the terminal phenol group they designed molecules

keeping these two moieties intact and modifying the rest of the molecule in order to

increase the efficacy. They successfully synthesized a new class of aminoanthranilic acid

agonists of GPR109a with potent agonists, reduced serum shift and excellent ADME

properties.

Hang et al.,(2011) have carried out a facile copper catalyzed tandem reaction for the

synthesis of 4,5-dihydropyrazolo[1,5-a]quinoline 176 (Scheme 32) and pyrazolo[1,5-

46
Review of literature

a]indoles 173 (Scheme 31). They found that the yields pyrazolo[1,5-a]quinolines were

found to be better than the indoles. This was explained on the basis of the steric

hinderance. They also came up with an efficient method for the synthesis of some fused

ring indoles and pyrazoles.

47
Review of literature

Colotta et al.,(2009) have reported a series of pyrazoloquinolines (Scheme 33) and found

them to exhibit a high affinity for adenosine receptors and were active in nanomolar

quantities.

Scheme 33

Scheme 34

48
Review of literature

Scheme 35

The target compounds were synthesized from 2-arylpyrazolo-[3,4-c]quinolines-4-amines,

200-203 (Scheme 34, 35), which were prepared from 3-ethoxalylindole. The final

molecules 178-197, 198 and 199 were synthesized from 4-amino derivatives by reacting

with suitable carboxylic acid in DMF in the presence of 1- hydroxybenzotriazole,

triethylamine, and 4-(dimethylamino)pyridine. 4-Diaroylamino derivatives, 198 and 199,

were obtained by refluxing the 4-amino derivative, 200, with an excess of 3-

49
Review of literature

trifluoromethylbenzoyl chloride and 4-trifluoromethylbenzoyl chloride, respectively, in

anhydrous methylene chloride and pyridine.

The structure-activity relationship of the synthesized compounds revealed that

introducing aroyl ring in place of the benzoyl moiety increases the binding affinity of the

synthesized molecules. They also evaluated the effect of various heterocyclic rings in the

basic scaffold and found that the 2-furyl and 2- or 3- or 4-pyridyl rings were the most

beneficial. The introduction of a methyl group to the furyl moiety increased the affinity

further. The presence of a Me or OMe, either in the para or meta position, while

maintaining a high hA3 affinity, reduced the hA3 versus hA1 selectivity. The authors

also carried out the docking studies in order to obtain a structure based pharmacophore

model (PDB id: 1L9H). The docking scores were compared with the binding assay

results. Based on these results a pharmacophore model was developed which may be of

help in designing molecules for this receptor.

The authors have also reported a novel group of compounds as adenosine receptor

antagonists, efficiently correlated the in silico and in vitro studies and explained the

structure-activity relationships of the synthesized molecules.

Duggineni et al.,(2006) have reported a novel application of a pictet Spengler reaction for

the synthesis of some pyrazoloquinolines and thiazoloquinolines (Scheme 36). Thiazole

and pyrazole based arylamine substrates were used for the reaction unlike the

conventional method. The studies carried out by this group proved that arylamines linked

to an activated heterocyclic ring can lead to a variety of second-generation substrates for

the Pictet–Spengler cyclisation (Scheme 37, 38).

50
Review of literature

Scheme 36

The authors worked on this concept with the hope to synthesize benzannulated

heterosystems and avoid the stereochemical issues associated with the traditionally used

pictet Spengler reactions. Their work helped to prove that aryl amine derived substrates

are likely to undergo pictet Spengler reaction faster than the substrate derived from

aliphatic amines. They also worked out an efficient synthetic strategy for the synthesis of

dihydropyrazoles (pyrazolines) and also successfully modified the problem faced during

cyclisation when an electron withdrawing group is attached to the aldehyde.

51
Review of literature

Scheme 38

Selvi et al.,(2006) have synthesized a series of pyrimido[4,5-b] 239, 240 (Scheme 39)

and pyrazolo[3,4-b]quinolines (Scheme 40,41) and evaluated them for their antimicrobial

activity. They carried out the synthesis using environmentally benign solvent-free

conditions using p-tolylsulphonic acid as catalyst and found that compounds 239a-g and

240a-g had significant effect on the inhibition of Bacillus subtilis, Escherichia coli,

Pseudomonas aeruginosa, compounds 239a–g exhibited good antifungal activity against

Candida albicans whereas the compounds 240a–g were active against Aspergillus flavus.

Compounds 242a–g were found to exhibit good antibacterial activity against E. coli and
52
Review of literature

P. aeruginosa in addition to a pronounced effect on the growth of fungi like Rhodotorula

rubra, C. albicans and Lipomyces lopofera whereas compounds 244a–g were active

against Staphylococcus aureus, Staphylococcus albus, E. coli and P. aeruginosa.

53
Review of literature

Kalayanov et.al.,(1998) have synthesized a series of 1-aryl-1H-pyrazolo[4,3-c]quinolines

and 2-aryl-2H-pyrazolo[4,3-c]quinolines (Scheme 42) and evaluated them for their

H+/K+- ATPase activity. They synthesized these molecules based on the reversible proton

pump inhibitor, SK and F 96067.

The carbonyl group present in the SK and F 96067 was found to be responsible for the

restriction of the NH group conformation by forming a hydrogen bond with the carbonyl

and also by increasing the conjugation between the nitrogen and quinolines ring. 1H-

Pyrazolo[4,3-c]quinolines were, therefore, synthesized in order to reduce the flexibility of

the molecule.

All the synthesized molecules were evaluated for their antiulcer activity using SK and F

97067 was used as the standard. The activity of the synthesized molecules was found to

be lower than that of the standard.

54
Review of literature

Wojciechowska et al.(1998) have reported some pyrazoloquinolines having affinity for

benzodiazepine receptors. They synthesized pyrazoloquinoline based on the scaffold

pyrazolo[4,3-c]quinolin-3-ones, known for their affinity for benzodiazepine receptors.


55
Review of literature

Studies were carried out to explain the effect of different substituents in pyrazolo[4,3-

c]quinolin-3-ones. A series of 6- and 7-substituted-2-arylpyrazolo[4,3-c]quinolin-3-ones

were synthesized (Scheme 43) and evaluated for their benzodiazepine receptor binding in

competition with flunitrazepam. The target molecules were synthesized from diethyl

ethoxymethylenemalonate via condensation with a suitable aniline. The product was

further treated with POCl3 and phenylhydrazine to obtain the final compounds. The

partition coefficient and electronic parameters used in correlation regression were

compared with the experimental data obtained. The results of the QSAR studies revealed

that the hydrophobicity and the position of the bicyclic core are both important for the

binding affinity for the benzodiazepine receptor. The authors also successfully evaluated

the structure-activity relationship of pyrazoloquinolines and explained the effect of the

substituents on the pyrazoloquinoline scaffold.

56
Review of literature

Scheme 43
David Barrett et al.(1996) have reported a novel synthesis of pyrazolo[1,5-a] quinolines

in excellent yields via a tandem Michael reaction of N-methylaminoquinolones with

various acrylate derivatives in the presence of NaH (Scheme 44). The synthesized

compounds were converted to DNA gyrase inhibitors by reaction with secondary amines

(Scheme 45). The in vitro antibacterial studies carried out on the synthesized molecules,

however, revealed that these derivatives were weak when compared to the standard,

levofloxacin.

Scheme 44

57
Review of literature

Scheme 45

Wentland et al.,(1995) have studied the topoisomerase II inhibitory activity of a

quinolone derivative and related compounds (Scheme 46). They demonstrated that

significant enhancement in topoisomerase II inhibition on introducing a keto group and

carboxylate group in the pyrazole ring.

Scheme 46

One of the earliest reports on the synthesis of 4,5-dihydropyrazolo[1,5-a]quinolines was

by Deshayes et al., (1983). They explored the photo reactivity of a series of 5-alkenyl or

dialkenyl-1-phenylpyrazoles and carried out the reaction under N2 atmosphere using

benzene as the solvent (Scheme 47). They did not, however, carry out the biological

evaluation of these molecules

58
Review of literature

Scheme 47

Table 2: Some of the patents related to pyrazoloquinolines are summarized below,

Patent number Biological activity Compound

EP 2 520 577 A1 Central cannabinoid receptor

Nov 7, 2012 (CB1) antagonizing activity.

US 7 863 266 B2 GABA receptor modulator Pyrazolo[4,3-c]quinolin-3-one

Jan 4 2011

US 7 858 614 B2 GABA receptor modulator Pyrazolo[4,3-c]quinolin-3-one

Dec 28, 2010

59
Review of literature

US 7 081 456 B2 Immunomodulation,


Rheumatoid arthritis,
July 25, 2006 multiple sclerosis, diabetes,
asthma, psoriasis.

US 6 642 249 B2 Immunomodulation,


Rheumatoid arthritis,
Nov 4, 2003
multiple sclerosis, diabetes,
asthma, psoriasis.

US 5 442 065 Antiinflammatory agents 1H-pyrazolo[4,3-c]quinol-4(5H)-one-


carboxylicacid
Aug 15, 1995

US 4 560 689 Benzodiazepine Pyrazolo[4,3-c]quinolin-3-one

Dec 24, 1985 receptor modulators

US 4 312 870 Psychoactive drug for 2-arylpyrazolo[4,3-c]quinolin-3-one


treatment of anxiety and
Jan 26 1982 depression.

60
Review of literature

US 4 268 516 Immuno regulators. Benzothiopyrano[4,3-c]pyrazoles

May 19, 1981

US 4 076 818 Bronchodialators, Pyrazolo[1,5-c]quinazoline

Feb 28, 1978 antihistamine,Antiinflammat

ory agent and for

rheumatoid arthritis.

US 4 024 149 Antifertility drug Triazolo[1,5-a]quinolines

May 17, 1977

Conclusions and Future Perspectives

In conclusion, sydnones are versatile and privileged structures which belong to the

mesoionic class of compounds. They possess a wide variety of biological activities and

undergo a large number of reactions like cycloaddition, alkylation, arylation, lithiation,

etc. Cycloaddition reactions have been one of the most exploited reactions of sydnones.

This has been a stepping stone for the discovery of lead molecules. Among these, the

reaction of sydnones to form pyrazoles are of considerable importance. The synthesis of

pyrazoloquinolines have lead to the discovery of several biologically active molecules.

Several pyrazoloquinolines have been evaluated for their anticancer, anticonvulsant,

antibacterial and antifungal activities among others. Although sydnones and

pyrazoloquinolines have been studied, fused ring derivatives of these molecules have

61
Review of literature

been reported only sparingly. In Particular, sydnoquinolines and 4,5-

dihydropyrazoloquinolines are yet to be explored, synthetically as well as biologically.

Pyrazoloquinolines are known for their ability to bind with Adenosine receptor,

benzodiazepine receptor, Chk1kinase, phosphodiesterase, ras and topoisomerase II.

Although sydnones and pyrazoloquinolines have been widely studied, reports on

sydnoquinolines and 4,5-dihydropyrazolo[1,5-a]quinolines are sparingly found in

literature. There is good scope to study these molecules adopting new synthetic strategies.

It was proposed, therefore, to explore the synthetic routes for 4,5-dihydrosydnoquinolines

and 4,5-dihydropyrazoloquinolines. The structural features of these molecules and their

capability to bind with receptors have increased the writer’s interest in them.

62
Scope and Objectives

2. SCOPE AND OBJECTIVES

The problems associated with the current cancer chemotherapy are non-specific toxicity

and multi drug resistance (MDR). Rapid advances are being made today in understanding

the molecular mechanisms underlying cancer development and progression.

Simultaneously, investigations are being conducted adapting a rational approach to drug

discovery in which a new generation of small molecules are designed and synthesized to

inhibit specific proteins or pathways abnormally expressed in cancerous cells. This

approach termed Targeted therapy is widely being studied today to discover new targets

and develop new anticancer drugs.

Targeted therapy offers a number of potential advantages over conventional

chemotherapy including an increased therapeutic index (i.e., effective cancer treatment

with less side effects), better tolerability due to a better toxicity profile, better target

selectivity, availability for chronic treatment and in some cases oral administration.

The principal criterion applied in modern anticancer drug design is the principle of

selective toxicity which requires activity restricted exclusively to the cancer cells. From

the point of view of selective activity directed on tumour cells and mechanisms of

carcinogenesis, different classes of modern drugs can be distinguished like inhibitors of

Cyclin Dependent Kinases (CDK’s), Check Point Kinases (CPK’s), Oncogenic Human

pappiloma virus (HPV), Topoisomerases, Cdc25 Phosphatase and ABC transporters.

DNA topoisomerases are enzymes that control and modify the topological states of DNA.

They can catalyze several interconversions between topological isomers of DNA by

transiently breaking single or double strands, and resealing them after reorganization of

63
Scope and Objectives

the topology. Topoisomerases are important drug targets for anticancer therapeutics due

to the vital role played by them in the survival of the cells.

Topoisomerase I (topo I) breaks a single DNA stand, while topoisomerase II (topo II)

breaks both strands but requires ATP for full activity. Since the activity of

topoisomerases is essential for several cellular processes such as replication, transcription

and chromosome condensation, investigation of the inhibitory activities of eukaryotic

topoisomerases is widely studied in anticancer drug development. Topo II is the target for

some of the most active anticancer drugs such as etoposide, teniposide, and doxorubicin

used in the treatment of human malignancies.

Compounds of the mesoionic class have interesting structural features due to their betain-

like character. They consist of a five membered ring associated with a sextet of p and Π

electrons, supported by a partial positive charge in the heterocyclic ring and

counterbalanced by a formal negative charge. The association of these characteristics

suggests a high probability of strong interactions with biomolecules such as DNA and/or

proteins. Sydnones belong to this class of compounds.

Sydnones, therefore, have been widely studied due to their unique heteroaromatic

character and their biological applications. Apart from their biological activity, another

attractive feature of these molecules is their application as synthetic precursors for other

molecules like pyrazolines and pyrazoles.

64
Scope and Objectives

Various 4-substituted sydnones have been studied over the years for anticancer,

antiinflammatory and analgesic activities among others. Some of these have shown

reasonably good biological activity. Sydnones have also been fused with quinolines or

tetrahydroquinolines. The tetrahydroquinoline ring is present in numerous biologically

active compounds. Due to the importance of these privileged molecules, numerous

synthetic routes have been reported for these molecules. Derivatives containing fused

ring sydnones, however, have been sparingly reported.

Further, a broad spectrum of biological activity has been reported for pyrazoloquinolines.

No pyrazoloquinolines have, however, been identified so far that show clinical promise

although the p-chloro derivative, CGS 9896, has been reported to be a potent

anticonvulsant.

65
Scope and Objectives

Would pyrazoloquinolines be effective against multidrug resistance? Numerous tri- and

tetracyclic planar nitrogen containing heterocycles are well known as topoisomerase

inhibitors. On the basis of some known topoisomerase II catalytic inhibitors like

benzophenanthridine, psorospermin, intoplicine, etc., it was hypothesized that the

presence of a planar ring system and 3 to 4 fused aromatic rings would result in a better

binding to the DNA.

Pyrazoloquinolines are known to be made up of 3 planar ring systems.

Pyrazoloquinolines were, therefore, chosen as the target molecules for topo II because the

fused polycyclic part can bind to the ATP site via van der waals forces and hydrogen

bonding interactions. It is also likely that the pyrazole ring when attached to the quinoline

ring may help in increasing the rigidity of the side chains attached to the quinoline

nucleus thus increasing its the efficacy for the ATP binding site of topoisomerase II.

66
Scope and Objectives

The basic scaffold of pyrazoloquinolines are,

Prazolo[3,4-c]quinolines, pyrazolo[4,3-c]quinolines and pyrazolo[1,5-c]quinolines have

been studied for their antitumor and adenosine receptor antagonist properties. Although a

few attempts have been made to synthesize 4,5-dihydropyrazolo[1,5-a]quinolines their

biological evaluation have not been reported so far.

Based on a detailed literature review, it was hypothesized that the desired

pyrazoloquinolines could be synthesized by the reterosynthetic Scheme 48.

Sydnoquinolines can be synthesized from anilines and crotonaldehyde via Doebner-

miller reaction. Further, the obtained molecules may be reduced and nitrosated. The N-

67
Scope and Objectives

nitroso derivatives so obtained can then be converted to sydnones using acetic anhydride

cyclisation. The sydnoquinolines so obtained can then be converted to some novel

pyrazoloquinolines via cycloaddition reaction. Sydnoquinolines are known to undergo

this reaction.

In the present work it was proposed, therefore, to synthesize some sydnoquinolines,

namely sydnones fused with tetrahydroquinoline ring. Attempts would also be made

to synthesize some fused ring pyrazoloquinolines from the sydnoquinolines via [3+4]

dipolar cycloaddition reaction. The sydnoquinolines and pyrazoloquinolines

obtained would then be evaluated for their anticancer activity in vitro using various

cell lines.

The objectives of the present study are to,

· design molecules for the topoisomerase II target.

· carry out docking studies of the designed molecules on the topoisomerase II (pdb

id:1ZXM)

· synthesize fused ring sydnoquinolines using feasible synthetic strategies.

· synthesize pyrazoloquinolines from sydnoquinolines via the cycloaddition

reaction.

· characterize all the synthesized molecules by MASS, 1H NMR, 13


C NMR,

NOESY spectroscopy.

· evaluate the synthesized molecules for their in vitro anticancer potential using

various cell lines.

68
Experimental

3. EXPERIMENTAL

3.1 Chemicals and Reagents

All the solvents and reagents were purified and dried according to the procedures given

in Vogel’s text book of Practical Organic Chemistry. All the reactions were monitored by

thin layer chromatography (TLC) on aluminum plates precoated with silica gel GF.

Reduction of C=C double bonds were carried out under H2 atmosphere. All the

synthesized compounds were characterized based on TLC, physical nature, melting point

(mp), Infrared Spectroscopy (IR), Nuclear Magnetic Spectroscopy (NMR) and Mass

Spectroscopy (MS).

The melting points were determined in open capillaries using Veego VMP-1 melting

point apparatus and are uncorrected. The temperatures are expressed in °C.

The IR spectra of the compounds were recorded on Shimadzu and Perking-Elmer FT-IR

spectrophotometer using neat technique and were expressed in cm-1. 1H NMR, 13C NMR,
1
H-1H HOMO COSY spectra were recorded on Bruker 400MHz spectrometer, using

CD3OD as solvent and TMS as internal standard. The chemical shifts were expressed in δ

ppm and the following abbreviations were used, s=singlet, bs= broad singlet, d = doublet,

t = triplet, q = quartet, m = multiplet. Attempts were made to quantify and identify the

peaks in 13C NMR spectra.

Mass spectra were recorded using high resolution mass spectrometer (HRMS) and liquid

chromatography mass spectroscopy (LCMS) under electron spray ionization technique,

using time of flight (TOF) and triple quardrupole mass analyzers, respectively.

69
Experimental

3.2 Docking and Molecular Modeling Studies

The molecular modeling studies were carried out on Linux Fedora 7.0 workstation using

GLIDE, version 5.7, Schrodinger suit 2011, LLC, New York, on a Maestro graphical user

interface.

3.2.1 Ligand Preparation

The structures of the ligands were generated using Chem Draw Ultra Version 8.0. These

ligands were converted to mol2 format and subjected to ligprep module of Maestro in

Schrodinger suite of tools. They were converted from 2D to 3D structures by including

stereochemical, ionization, tautomeric variations as well as energy minimization. They

were then optimized for their geometry, desalted and corrected for their chiralities and

missing hydrogen atoms. The bond orders of these ligands were fixed and the charges

neutralized. The ionization and tautomeric states were generated between pH of 6.8 to 7.2

using Epik module. In the final stage of Ligprep, compounds were minimized using

Optimized Potential for Liquid Simulations-2005 (OPLS-2005) force field in Impact

package of Schrodinger until a root mean square deviation of 1.8 Å was achieved.

Steepest descent algorithm was used for minimization, followed by conjugate gradient

method. A single low energy ring conformation per ligand was generated and the

optimized ligands were used for docking.

3.2.2 Protein Preparation

The crystal structure of the topoisomerase II was downloaded from RCSB Protein Data

Bank (Collaboratory for Structural Bioinformatics, PDB) and was used for the modeling

studies. Refinement of bond orders, formal charges and missing hydrogen atoms,

70
Experimental

incomplete and missing residues and terminal amide groups were carried out. Water

molecules beyond 5 Å of the hetero atom were removed. The possible ionization states

were generated for the heteroatoms in the protein and the most stable state was chosen.

The hydrogen bonds were assigned and orientations of the retained water molecules were

corrected. A restrained minimization of the protein molecule was carried out using OPLS

2005 force field to reorient side chain hydroxyl groups and alleviate potential steric

clashes. The minimization was restrained to the input protein coordinated by a predefined

Root Mean Square Deviation (RMSD) tolerance of 0.3 Å.

3.2.3 Receptor Grid Generation

The ligand was retained in the crystal structure of the prepared protein which was used

for the receptor grid construction. The grid dimensions (within which the docked pose is

confined) of the protein was set to 14 Å X 14 Å X 14 Å.

3.2.4 Validation of the docking programme

The accuracy of the docking procedure was determined by comparing the lowest energy

pose of the co-crystallized ligand predicted by the object scoring function, Glide score,

and an experimental binding mode as determined by X-ray crystallography. Extra

precision Glide procedure was validated by removing the co-crystallized ligand from the

binding site and redocking the ligand. The hydrogen bonding interactions and the root

mean square deviation (RMSD) between the predicted conformation and the observed X-

ray crystallographic conformation was used to analyze the results.

71
Experimental

3.2.5 Glide ligand docking

The docking of the molecules were carried out using the prepared receptor grid and the

ligand molecules. The favorable interactions between the ligand and receptor were scored

using Glide. All the docking calculations were performed using extra precision (XP) and

OPLS 2005 force field. The program was run in the flexible mode which automatically

generates conformations for each input ligand. The poses generated were run through a

series of filters to evaluate the receptor-ligand interactions. The initial filter tests the

spatial fit of the ligand to the defined active site and examines the ligand –receptor

interactions using a grid based method patterned after the empirical ChemScore function.

The algorithm recognizes hydrophobic interactions, hydrogen bonding, metal-ligand

interactions. Poses that pass this initial screening enter the final stage which involves

evaluation and minimization of a grid approximation OPLS nonbonded ligand-receptor

interaction energy. Finally the minimized poses were rescored using Glide Score

function. Glide Score is based on chemscore, but includes a steric-clash term, adds buried

polar terms to penalize electrostatic mismatches and has modifications to other terms:

GScore = 0.065 * vdw + 0.1130 * Coul + Lipo +Hbond + Metal + BuryP + RotB +
Site
Where, Vdw = Vander waals energy, Coul = Coloumb energy, Lipo = Lipophilic term,
Hbond = Hydrogen Bonding term, Metal = metal binding term, BuryP = penalty for
buried polar groups, RotB = Penalty for freezing rotatable bonds, Site = Polar
interactions in the active site.

72
Experimental

3.2.6 General procedure for molecular modeling studies for the identification of

potent ligands

Docking studies on ligands were carried out in the active site using Glide. The ligands

prepared were docked using High-Thorughput Virtual Screening (HTVS) which is the

least computationally intense process intended for rapid screening of ligands. From this,

ligands were selected for further steps based on their Glide energy and ranking. The top

100 ligands were selected and docked in the standard precision (SP) mode from which

30 ligands were selected and docked using extra precision (XP). From this result

obtained the ligands with the best score were selected.

3.3 Synthesis and Purification

3.3.1 General procedure for preparation of quinolines

Aniline (1.12g, 4.46mmol) was heated with 6M HCl (22.4mL) at 100°C (bath

temperature). Toluene (5.8mL) and then crotonaldehyde (0.74mL, 8.92mmol) were added

dropwise at 100°C. Stirring was continued for 2h at 100°C and the mixture was allowed

to cool to room temperature. The lower aqueous layer was separated and neutralized with

aqueous NaOH. The neutralized layer was extracted with ethyl acetate (3X5mL). The

ethyl acetate layer was dried and concentrated to afford the crude 2-methylquinoline. The

crude product was purified using flash column to give 70% yield of the pure compound.

73
Experimental

3.3.2 General procedure for preparation of quinoline-2-carboxylic acids

Selenium dioxide (0.36g, 3.3mmol) was added to a solution of 2-methylquinoline (0.63g,

3.1mmol) in dry 1,4-dioxan (30mL). The reaction mixture was refluxed for 2h and then

cooled to room temperature and filtered. The filtrate was evaporated to give a residue

which was dissolved in formic acid (1mL) and cooled to 0°C. Hydrogen peroxide (1.7mL

of 30% solution in water, 15.5mmol) was slowly added and the mixture was allowed to

stand overnight between 0°C and 10°C. The precipitate formed was collected by

filtration, washed with cold water and dried to obtain 3-hydroxyquinaldic acid as an

yellow solid. This was then purified using flash column by elution with CHCl3 to get 75%

yield of the pure compound.

3.3.3 General procedure for preparation of 1,2,3,4-tetrahydroquinoline-2-

carboxylic acids

Quinaldinic acid (1g, 5.77mmol) in methanol (6mL) was hydrogenated over platinum

oxide (0.3g) under atmospheric pressure at room temperature until the theoretical amount

of hydrogen was consumed. The mixture was filtered through celite and the filtrate was

concentrated and used as such for the next step.

74
Experimental

3.3.4 General procedure for preparation of N-nitroso quinoline-2-carboxylic acids

In a 250mL round bottom flask tetrahydroquinoline-2-carboxylic acid (4.60g, 19.1mmol),

THF (10mL), NaNO2 (1.51g, 22.0 mmol) and HCl (3M, 8.8mL) were placed. The

reaction mixture was stirred for 18h at room temperature. The completion of the reaction

was confirmed using TLC. The reaction mixture was concentrated and used as such for

the next step.

3.3.5 General procedure for preparation of sydnoquinolines

The residue obtained from scheme 3.3.4 (3.6g) was treated with acetic anhydride (30mL)

and pyridine (2mL). The completion of reaction was monitored using TLC. Water was

added to the reaction mixture to neutralize the acetic anhydride and extracted with ethyl

acetate (3X5mL). The ethyl acetate layer was dried and concentrated and purified using

flash column to get 70% yield of the pure compound. The structures and data of the

sydnoquinolines obtained are given in Table 3.

75
Experimental

Table 3: Data on 4,5-dihydro[1,2,3]oxadiazolo[3,4-a]quinolin-10-ium-3-olate and its


derivatives

Comp Molecular Rf
Structure IUPAC Name formulae/
no. value
Weight

4,5-dihydro[1,2,3]oxadiaz-
C10H8N2O2
21 -olo[3,4-a]quinolin-10- 0.48
188.1827
ium-3-olate

7-methoxy-4,5-dihydro-
C11H10N2O3
22 [1,2,3]oxadiazolo[3,4- 0.50
218.2087
a]quinolin-10-ium-3-olate

7-fluoro-4,5-dihydro- C10H7FN2O
23
[1,2,3]oxadiazolo[3,4- 2 0.50
a]quinolin-10-ium-3-olate 206.1732

7-methyl-4,5-dihydro-
24 C11H10N2O2
[1,2,3]oxadiazolo[3,4- 0.60
202.2093
a]quinolin-10-ium-3-olate

76
Experimental

3.3.6 General procedure for preparation of pyrazoloquinolines

A solution of the sydnone (0.05g) and ethylphenylacetylene (0.05g) in Xylene (2mL) was

stirred under N2 atmosphere at reflux for 4-5h. The solvent was removed in vacuo to give

the crude product. Further purification by flash chromatography on silica gel gave the

product 66% yield of the pure compound.

The structures and data on pyrazoloquinolines are given in Table 4.

Table 4: Data on 4,5-dihydropyrazolo[1,5-a]quinolines

Comp Molecular Rf
Structure IUPAC name
no. formulae/weight value

ethyl 3-
phenyl-4,5-
dihydropyrazo
C20H18N2O2 0.40
26 lo[1,5-
318.3691
a]quinoline-2-
carboxylate

ethyl 7-
methoxy-3- C21H20N203
27 phenyl-4,5- 348.3951
0.50
dihydropyrazo
lo[1,5-
a]quinoline-2-
carboxylate

77
Experimental

28
ethyl 7-fluoro- C20H17FN2O2
3-phenyl-4,5- 0.50
dihydropyrazo 336.3596
lo[1,5-
a]quinoline-2-
carboxylate

ethyl 7-
methyl-3-
phenyl-4,5- C21H20N2O2
29 dihydropyrazo 0.54
lo[1,5- 332.3957
a]quinoline-2-
carboxylate

dimethyl 4,5-
dihydropyrazo
lo[1,5- C15H14N2O4
31 0.52
a]quinoline- 286.2827
2,3-
dicarboxylate

dimethyl 7-
methoxy-4,5-
dihydropyrazo C16H16N2O5
32 lo[1,5- 0.48
a]quinoline- 316.3086
2,3-
dicarboxylate

78
Experimental

dimethyl 7-
fluoro-4,5-
dihydropyrazo C15H13FN2O4
33 lo[1,5- 0.50
a]quinoline- 304.2731
2,3-
dicarboxylate

dimethyl 7-
methyl-4,5-
dihydropyrazo C17H19N2O4
34 lo[1,5- 0.46
a]quinoline- 315.3438
2,3-
dicarboxylate

Benzyl 4,5-
dihydropyrazo C19H16N2O2
36 lo[1,5- 0.62
a]quinoline-3- 304.3425
carboxylate

Benzyl 4,5-
dihydropyrazo C19H16N2O2
40 lo[1,5- 0.58
a]quinoline-2- 304.3425
carboxylate

79
Experimental

benzyl 7-
methoxy-4,5-
dihydropyrazo C20H18N2O3
37 0.64
lo[1,5- 334.3685
a]quinoline-3-
carboxylate

benzyl 7-
methoxy-4,5-
dihydropyrazo C20H18N2O3
41 0.59
lo[1,5- 334.3685
a]quinoline-2-
carboxylate

Benzyl 7-
fluoro-4,5-
dihydropyrazo C19H15 FN2O2
38 0.60
lo[1,5- 322.3330
a]quinoline-3-
carboxylate

Benzyl 7-
fluoro-4,5-
dihydropyrazo C19H15 FN2O2
42 0.55
lo[1,5- 322.3330
a]quinoline-2-
carboxylate

80
Experimental

Benzyl 7-
methyl-4,5-
dihydropyrazo C20H18N2O2
39 0.60
lo[1,5- 318.3692
a]quinoline-3-
carboxylate

Benzyl 7-
methyl-4,5-
dihydropyrazo C20H18N2O2
43 0.57
lo[1,5- 318.3692
a]quinoline-2-
carboxylate

3.4 In vitro cell proliferation MTT assay

3.4.1 Principle

Measurement of cell viability and proliferation forms the basis of numerous in vitro

anticancer assays. The yellow coloured MTT (3-(4,5-Dimethylthiazol-2-yl)-2,5-

diphenyltetrazolium bromide) is reduced to the purple coloured formazan in living cells.

A solubilization solution (usually either dimethyl sulfoxide, an acidified ethanol solution,

or a solution of the detergent sodium dodecyl sulfate in diluted hydrochloric acid) is

added to dissolve the insoluble purple formazan product into a colored solution. From the

absorbance of this colored solution the product obtained can be quantified by measuring

at a certain wavelength (usually between 500 and 600 nm) using a spectrophotometer.

81
Experimental

The absorption maximum is dependent on the solvent employed. The intensity of the

colour of the product is directly proportional to the number of living cells in the culture.

Reduction of MTT into formazan crystals by mitochondrial dehydrogenase.

3.4.2 Maintenance of cell lines

The cell lines were maintained using Eagle’s MEM supplemented with 10% FBS and 50

μg/mL gentamycin sulphate at 37°C in CO2 incubator in an atmosphere of humidified 5%

CO2. The cells were maintained by routine sub culturing in 25 cm2 tissue culture flasks.

3.4.3. Sub culturing process of cell lines

· The culture media from the flasks containing the monolayer culture was aspirated

and washed with sterile phosphate buffered saline (PBS).

· To the flasks, 2 mL of 0.1% trypsin-EDTA solution was added. After few seconds

it was aspirated and the flask was kept in incubator for 2-3 minutes for cell

detachment.

· The flasks were then removed from the incubator and the cell detachment was

confirmed by observing under an inverted microscope (Olympus).

82
Experimental

· Once the cells were completely detached from the flasks 2-3 mL of MEM media

containing 10% FBS was added and mixed well.

· Cell viability was checked with a small sample of the suspension by tryptan blue

dye exclusion test.

· From the stock cell suspension, 1x104 viable cells/mL suspended in media were

seeded in 25 cm2 tissue culture flask containing about 4 mL of fresh media and

incubated until the flasks attained 60-70% confluence.

3.4.4. Preservation of the tumor cells

Tumor cells from the first and second passage of transplantation were stored in liquid

nitrogen in cryovials at a concentration of 1x106 cells/mL containing respective media

supplemented with 20% serum and 10% DMSO as a preservative. This constitutes the

tumor bank. After every ten passages, the tumor cells were discarded and new passage

was started using the original tumor cells from the tumor bank.

3.4.5. Trypsinisation

To obtain a single cell suspension from monolayer culture, cells were dislodged from the

culture flasks by trypsinisation.

· From a 60-70% confluent flask, the culture media was aspirated out using

micropipette. Cells were washed with 3 mL of PBS to remove the trace amount of

media.

83
Experimental

· To each culture flask 2 mL of trypsin-EDTA was added and after few seconds it

was aspirated and the flask was kept in the incubator for 3-4 minutes for cell

detachment.

· The culture flasks were observed under an inverted microscope to ensure that the

cells were completely dislodged.

· Trypsin activity was stopped by adding 2-3 mL media containing 10% FBS.

3.4.6 In vitro cell proliferation MTT assay method

The selected compound was dissolved in 1mL of distilled dimethylsulphoxide (DMSO)

and the volume was made up to 10mL with maintenance medium to obtain a stock

solution of 1mg/mL concentration, sterilized by filteration and further dilutions were

made from the stock solution. The cytotoxicity assay was carried out using 0.01mL of

cell suspension, containing 10,000 cells/well of a 96 well microtitre plate. Fresh medium

containing different concentrations of the test compounds were added after 24h of partial

monolayer. Control cells were incubated with the test compounds and MEM. The

microtitre plates were incubated at 37°C in a humidified incubator with 5% CO2 for a

period of 72h. MTT solution (20µL, 2mg/mL in PBS) was added to the plates and

incubated for 4h at 37°C. MTT- formazon crystals formed were dissolved in 100 µL of

DMSO and the optical density was read with a microtitre plate reader at 570nm. The

percentage cytotoxicity of each compound at various concentrations was calculated using

the following equation;

84
Results and Discussion

4. RESULTS AND DISCUSSION

4.1 Docking Studies

The Ramachandran diagram was studied to visualize the backbone dihedral angle of the

amino acid residues in the protein (Figure 10). The plot showed about 87.6% residues in

the most favorable regions.

Figure 10: Ramachandran Plot- 1ZXM

The validation results of Glide docking procedure for topo IIα revealed a very good

agreement between the localization of the ligand upon docking as compared to the crystal

structure. The re-docked ligand, phosphoaminophosphonic acid-adenylate ester (ANP),

showed similar hydrogen bonding interactions with Asn91, Asn120, Ser148, Ser149,

Asn150, Arg162, Asn163, Gly164, Tyr165, Gly166, Ala167, Lys168, Gln376, Lys378

and Mg2+ residues of the protein, suggesting the reliability of the Glide docking in

85
Results and Discussion

reproducing the experimentally observed binding mode for ANP (Figure 11). The

parameters set for Glide docking are, therefore, reasonable to reproduce the X-ray

structure.

Figure 11: Hydrogen bonding interactions of ANP, the co-crystallised ligand.

A series of 180 molecules were designed for both sydnoquinolines and 4,5-

dihydropyrazoloquinolines. All these molecules were screened in silico for their ability to

inhibit the topo IIα at the ATP site. Among them a few of them showed a good docking

score (Table 5). Salvicine, a well known potent topo IIα inhibitor, was used as the

standard. Salvicine is currently in Phase II clinical trials. The docking scores of the

designed molecules were compared with the score of salvicine.

86
Results and Discussion

Table 5: Docking parameters of selected ligands.

Title Docking score Glide energy


C49 -7.384656 -45.113769
C36 -7.088328 -52.495523
C45 -7.064577 -42.567197
Salvicine -6.909206 -41.849454
C48 -6.801888 -48.801266
C37 -6.757759 -50.040877
C44 -6.608523 -55.093816
C41 -6.320824 -47.07366
C39 -6.195273 -52.644247
C40 -5.948458 -49.987325
C43 -5.462206 -47.60748
C47 -5.460709 -46.249083
C35 -5.322588 -48.209609
C34 -5.007437 -40.726292
C46 -4.3637 -31.648829
C42 -4.040974 -30.734029
C38 -1.193795 -32.451712

87
Results and Discussion

Both the R and S isomers of salvicine were docked to determine which isomer binds

better with the ATP site. It was observed that the S-isomer (Figure 12) had a better score

(-6.90921) than that of the R-isomer (-4.5461).

Figure 12: Hydrophobic interactions of salvicine-S in the binding site of topo IIα

The molecules used for docking were designed based on the basic scaffold given below.

The results of the docking studies revealed that on the sydnoquinoline ring (3) the change

in the functional group R does not bring any major change to the binding affinity. The

presence of the sydnone ring attached to the tetrahydroquinoline enhances the rigidity of

the molecule. The docking score of the sydnoquinoline was found to be -4.046974.

Although the change in the substituents bring about a slight variation in the glide score, it

was not significant. The scores of the pyrazoloquinoline (1) were found to be higher with

88
Results and Discussion

the presence of a phenyl ring attached to the nucleus at the R1 position (Figure 13). This

could probably be due to the enhanced resonance in the aromatic ring system. The

presence of an electron donating group attached to the –COOH was found to enhance the

binding affinity. On increasing the length of the side chain increased binding affinity than

the standard selected with a glide score of -7.3846. In the case of the pyrazolines (2) the

scores were comparatively low, probably because of the absence of aromaticity in the

pyrazole ring. The aromaticity of the ring is thus a crucial factor for binding.

The results of the docking studies showed very good glide score indicating a good

interactions with the ATP binding site (Figure 13-15).The docking scores revealed that

the designed molecules have good hydrogen bonding interactions with the important

amino acids in the ATP binding site. The molecules were found to interact with Asn 91,

Ser 148, Ser 149, Asn 150 Ala 167 and Lys 168 residues. The scores of a few designed

molecules were better than salvicine, whereas, others were comparable with that of

salvicine. A total of 20 molecules were selected for synthesis based on the glide scores.

The glide scores of the ligand C49 (-7.3846) were higher to that of salvicine and that of

C48 (-6.80188) is similar to the standard, salvicine. In comparison with salvicine, the test

molecules, C49, C36 and C35 showed better glide score and glide energy.

89
Results and Discussion

Figure 13 : Hydrophobic interactions of the ligands in the binding site of topo IIα

Figure 14: Hydrophobic interactions of the ligands in the binding site of topo IIα

90
Results and Discussion

Figure 15: Hydrophobic interactions of the ligands in the binding site of topo IIα

4.2 ADME studies

QikProp, the prediction program designed by Prof. William L. Jorgensen (Jorgenson et

al.,2004), was used to calculate ADME (Absorption, Distribution, Metabolism, and

Excretion) properties. Qikprop is quick, accurate and predicts physically significant

descriptors and pharmaceutically relevant properties of organic molecules. Ligprep

minimized ligands were given as a source in Qikprop 3.2. Qikprop modules provide the

ranges of molecular predicting properties for comparing the properties of a particular

molecule with those of 95% of known drugs. This helps in screening the proposed

molecules for various properties “drug-like” molecules should follow. Using the

compounds designed by SBDD, the physiochemical properties were, therefore, calculated

in Qikprop simulation. All the compounds obeyed the Lipinski's rules: molecular weight

below 500 Da, hydrogen bond donor (less than five) and acceptor (less than ten). Also

91
Results and Discussion

QPlogP o/w (octanol/water partition coefficient) for all the compounds was less than five.

Total solvent accessible surface area (SASA), hydrophobic component of the SASA

(FOSA) and hydrophilic component of the SASA (FISA) were analyzed for the

compounds that were abiding the ranges in Qikprop physiochemical properties.

Qualitative human oral absorption was predicted. Polar nitrogen and oxygen van der

waals surface area (PSA) of SBDD compounds fullfilled the limit in physicochemical

calculations. All the compounds satisfied the values of partition coefficient of octanol/gas

(QPlogPoct), water/gas (QPlogPw) and brain/blood (QPlogBB). Aqueous solubility

(QPlogS) and skin permeability (QPlogKp) predicted for ligands also lie in the allowed

solubility and permeability range.

4.3 Synthesis and Characterization

Scheme 49 gives the synthesis of 4,5-dihydro-[1,2,3]oxadiazolo[3,4-a]quinolin-10-

ium-3-olates:

92
Results and Discussion

2-Methylquinoline was synthesized by Doebner Miller reaction. In this method the

reaction takes place in a two phase solvent system which reduces polymerization of the

aldehyde (Matsugi et al.,2000). Aniline and crontonaldehyde were heated to reflux along

with 6M HCl and toluene for 3h. The product, 2-methyl quinoline, was isolated in good

yields after purification by flash column. The solvent system used was PE:EtOAc (7:3).

The structure of the product was confirmed using 1H NMR and mass. The proton NMR

reveals, three methyl protons (-CH3) at δ 2.75 ppm and 6 aromatic protons appear in the

region δ 7.26 - 8 ppm. In the mass spectra M+1 (144 amu) is observed.

Mechanism of for step 1 of Scheme 49.

2-Methyl quinoline obtained was oxidized to quinoline-2-carboxylic acid using selenium

dioxide and dioxan followed by hydrogen peroxide and formic acid (Riego et al.,2005).

This reaction takes place in two stages. First, conversion of the methyl group of 2-methyl

quinoline to an aldehyde on treatment with SeO2 and dioxan, followed by conversion to

the acid when treated with H2O2 and HCOOH. Generally to carry out the oxidation of the

methyl group, a strong oxidizing agent like KMnO4 would be used. But oxidation with

93
Results and Discussion

KMnO4 leads to cleavage of the double bonds and hence KMnO4 was not used. The

quinoline-2-carboxylic acid obtained was purified by eluting with CHCl3 to obtain the

pure product. The product was confirmed using 1H NMR which shows 6 aromatic protons

between δ 7.72-8.43 ppm.

Mechanism of step 2 of Scheme 49

The double bond of the quinoline-2-carboxylic acid was reduced to afford 1,2,3,4-

tetrahydroquinoline-2-carboxylic acid. This was carried out using platinum oxide as the

catalyst (Kim et al.,2007). The reaction was carried out under hydrogen atmosphere. The

product obtained was used as such for the next step. Efforts were made to isolate the pure

product but the product was found to degrade on eluting with the solvent during column

chromatography. Hence, the catalyst was filtered out and the solvent was removed and

the residue was dried thoroughly and used for the next step. The formation of the product

was confirmed by using ninhydrin solution as the staining agent.

94
Results and Discussion

Mechanism of step 3 of Scheme 49.

The tetrahydroquinoline was converted to its N-nitroso derivative using NaNO2, 3M HCl

and THF (Banerjee et al.,2010). The N-nitroso product was confirmed using ninhydrin

solution as the staining agent. The N-nitroso derivative was isolated by extracting with

EtOAc which was removed in vacuo and dried thoroughly to get a residue which was

used as such for the next reaction.

Mechanism of step 4 of Scheme 49.

The synthesis of the sydnoquinoline was carried out by treating the N-nitroso derivative

with Ac2O and a drop of pyridine. In this reaction, acetic anhydride and pyridine react to

form 1-acetylpyridinium, which in turn reacts with the N-nitroso quinoline-2-carboxylic

acid to form an acetyl derivative of the N-nitroso quinoline-2-carboxylic acid. With the

elimination of –OAc, cyclisation takes place to give the sydnoquinoline. The reaction

mixture was stirred overnight and purified using flash column to give the sydnone in

95
Results and Discussion

good yield. The reaction was carried out under N2 atmosphere. The mechanism for the

above reaction may be given as follows;

Mechanism of step5 of Scheme 49.

The following is the physical and analytical data of the compounds 4,5-dihydro[1,2,3]

oxadiazolo[3,4-a]quinolin-10-ium-3-olates;

4,5-dihydro[1,2,3]oxadiazolo[3,4-a]quinolin-10-ium-3-olate(21): brown sticky solid;

Rf : 0.48 (hexane/EtOAc, 5:5); Purified using flash column; Yield: 70%; IR (neat) :

2936, 2863, 1739, 1507, 1243, 767, 722cm-1; 1H NMR (400 MHz, CDCl3) δ ppm: 7.94-

7.96(d, 1H, J=8Hz), 7.43-7.55(m, 3H), 3.06-3.10(t, 2H), 2.93-2.97(t, 2H);13C NMR (100

96
Results and Discussion

MHz,CDCl3):167.57, 131.69, 130.11, 129.52, 128.29, 117.57, 102.48, 24.7, 16.77; HR-

MS m/z:calcd for C10H8N2O2Na [M+Na]: 211.0483; found: 211.0491.

7-methoxy-4,5-dihydro-[1,2,3]oxadiazolo[3,4-a]quinolin-10-ium-3-olate (22): Light

brown sticky solid; Rf : 0.5 (hexane/EtOAc, 5:5); Purified using flash column; Yield: 72%;

IR (neat) : 2936, 2863, 1741, 1717, 1265, 767, 722cm-1; 1H NMR (400 MHz, CDCl3) δ

ppm: 7.87-7.90(d, 1H, J=12Hz), 6.93-6.95(m,1H, J= 8Hz), 6.89(m, 1H), 3.88(s, 3H),

3.01-3.04 (t, 2H), 2.91-2.95 (t, 2H);13C NMR (100 MHz, CDCl3): 167.50, 161.84, 132.03,

119.22, 114.35, 113.35, 101.43,55.73, 25.22, 16.88; HR-MS m/z:calcd for C11H10N2O3Na

[M+Na]: 241.0589; found: 241.0596.

7-Fluoro-4,5-dihydro-[1,2,3]oxadiazolo[3,4-a]quinolin-10-ium-3-olate (23): Brown

sticky solid; Rf : 0.5 (hexane/EtOAc, 5:5); Purified using flash column; Yield: 75%; IR

(neat) : 3085, 1739, 1495, 1249,767, 722cm-1; 1H NMR (400 MHz, CDCl3) δ ppm: 7.96-
13
7.99(s, 1H), 7.15-7.19(m, 2H), 3.07-3.11 (t, 2H), 2.95-2.98 (t, 2H); C NMR (100 MHz,

CDCl3):167.36, 165.11, 162.58, 133.05, 132.96, 127.64, 119.97, 119.88, 116.69, 116.46,

115.77, 115.53, 102.10, 24.96, 16.65;HR-MS m/z:calcd for C11H10N2O3Na [M+Na]:

229.0389; found: 229.0388.

7-Methyl-4,5-dihydro-[1,2,3]oxadiazolo[3,4-a]quinolin-10-ium-3-olate(24): Brown

sticky solid; Rf :0.6 (hexane/EtOAc, 5:5); Purified using flash column; Yield: 67%; IR

(neat) : 1743, 1739, 1493,1238, 767, 722cm-1; 1H NMR (400 MHz, CDCl3) δ ppm: 7.81-

7.83(d, 1H, J=8Hz), 7.23-7.27(m, 2H), 3.00-3.04 (t, 2H), 2.90-2.94 (t, 2H), 2.43 (s,

3H);13C NMR (100 MHz, CDCl3):167.57, 142.41, 129.97, 128.97, 128.92, 117.41,02.07,

24.77, 21.43, 16.89;HR-MS m/z:calcd for C11H10N2O2Na [M+Na]: 225.0640; found:

225.0641.

97
Results and Discussion

Figure 16: IR spectra of Compound 21.

Figure 17: MASS spectra of Compound 21.

98
Results and Discussion

Figure 18: 1H NMR spectra for Compound 21.

Figure 19: 13C NMR spectra for Compound 21.

99
Results and Discussion

Figure 20: IR spectra for Compound 22.

Figure 21: MASS spectra for Compound 22.

100
Results and Discussion

Figure 22: 1H NMR spectra for Compound 22.

Figure 23: 13C NMR spectra for Compound 22.

101
Results and Discussion

Figure 24: IR spectra of Compound 23.

Figure 25: MASS spectra for Compound 23.

102
Results and Discussion

Figure 26: 1H NMR spectra for Compound 23.

Figure 27: 13C NMR spectra for Compound 23.

103
Results and Discussion

Figure 28: IR spectra for Compound 24.

Figure 29: MASS spectra for Compound 24.

104
Results and Discussion

Figure 30: 1H NMR spectra for Compound 24.

Figure 31: 13C NMR spectra for Compound 24.

105
Results and Discussion

Scheme 50 gives the synthesis of Ethyl 3-phenyl-4,5-dihydropyrazolo[1,5-


a]quinoline-2-carboxylates:

The cycloaddition reaction of the sydnones was carried out with three different alkynes.

The reaction takes place via [3+2] cycloaddition. The elimination of CO 2 results in the

final pyrazole ring. The reaction of sydnone with ethylphenyl acetylene results in only

one product, ethyl 3-phenyl-4,5-dihydropyrazolo[1,5-a]quinoline-2-carboxylate.

Mechanism of Scheme 50.

This was confirmed using Mass, IR, 1H NMR, 13


C NMR, 2D-NMR. The mass spectra

shows the [M+H] peak of 319.1449 amu. The proton NMR shows, 3 methyl protons of

the terminal –CH3 at δ 1.22-1.26 ppm as a triplet due to the adjuscent –CH2. The –CH2

appears as a quartet at δ 4.22-4.27 ppm due to the –CH3. The –CH2 of the

106
Results and Discussion

tetrahydroquinoline ring appear as triplets at δ 3.42-3.38 ppm, δ 3.04-3.01 ppm,


13
respectively. The C NMR shows aromatic carbon at 163.71,

154.06,143.98,135.68,132.97,129.52,128.41,128.29,127.83,127.68,126.72,126.15,116.63,

109.18 and aliphatic carbons at 60.05,24.69, 21.55,14.11.

The 13C NMR shows all the aromatic protons appear between δ 7.70-7.18 ppm. The 2D-

NMR spectrum revealed the interaction between the proton of the tetrahydroquinoline

ring and the proton of the phenyl ring as shown below.

The following are the physical and analytical data for Ethyl 3-phenyl-4,5-dihydropyra-

-zolo[1,5-a]quinoline-2-carboxylates;

Ethyl 3-phenyl-4,5-dihydropyrazolo[1,5-a]quinoline-2-carboxylate (26): White solid;

Rf : 0.4 (hexane/EtOAc, 9:1); m.p.: 87-90°C; Yield:66 %; IR (neat) : 2931, 1719, 1708,

1418, 1288, 1155, 1130, 1065, 765, 696 cm-1;1H NMR (400 MHz,CDCl3) δ ppm: 7.70-

7.72(m, 1 H), 7.40-7.45(d, 2 H), 7.37-7.39(m, 3 H), 7.28-7.36(m, 1H), 7.25-7.26(d, 1H),

107
Results and Discussion

7.18-7.22(d, 1H, J=8Hz), 4.22-4.27 (q, 2H),3.42-3.38 (t, 2H),3.04-3.01(t, 2H),1.22-

1.26(t,3H);13C NMR(100 MHz, CDCl3):163.71, 154.06, 143.98, 135.68, 132.97, 129.52,

128.41,128.29,127.83,127.68, 126.72, 126.15, 116.63, 109.18, 60.05, 24.69, 21.55,14.11;

HR-MS m/z: calcd for C20H18N2O2H [M+H]: 319.1447; found: 319.1449.

7-Methoxy-3-phenyl-4,5-dihydropyrazolo[1,5-a]quinoline-2-carboxylate (27): White

solid; m.p.: 128-130°C; Rf : 0.5(hexane/EtOAc, 9:1); Yield:68 %; IR (neat) : 2951 ,1741,

1720, 1736, 1477, 1294, 1226, 1077, 760 cm-1;1H NMR (400 MHz,CDCl3) δ ppm: 7.89(d,

1H), 7.70-7.72(d,2H, J=8Hz), 7.40-7.42 (d, 2H, J=8Hz), 7.26(s, 2H), 6.82-6.87(t, 1H),

4.23-4.25(q, 2H), 3.83(s, 3H), 2.98-3.02(t, 2H),1.22-1.26(t, 3H);13C NMR(100

MHz,CDCl3):163.76,157.83,153.69,143.00,129.51, 128.29,127.64,117.79,113.94,112.54,

108.79,59.95,55.52,25.08,21.55,14.10;HR-MS m/z: calcd for C21H20N2O3Na[M+Na]:

317.1372; found: 371.1372.

Ethyl 7-fluoro-3-phenyl-4,5-dihydropyrazolo[1,5-a]qunoline-2-carboxylate (28):

White solid; m.p.: 20-121°C; Rf :0.5(hexane/EtOAc, 9:1); Yield:70 %; IR (neat) : 3372,

2018, 1701, 1158, 1018, 764 cm-1;1H NMR (400MHz,CDCl3) δ ppm: 7.91-8.00(m, 1H),

7.70-7.71(m, 2H), 7.26(m, 3H), 6.99-7.05(m, 2H), 4.22-4.27(q, 2H),3.38-3.42(t, 2H),3.00-

3.04(t, 2H),1.22-1.25(t, 3H);13C NMR(100 MHz,CDCl3):163.59,161.91,159.46,154.06,

143.38, 132.84, 132.01, 129.48, 128.86, 128.45, 127.69, 118.23, 115.31, 115.08, 114.59,

114.36, 109.22, 60.07, 24.84, 21.35, 14.09; HR-MS m/z: calcd for C20H17 FN2O2H[M+H]:

337.1352; found: 337.1354.

108
Results and Discussion

Ethyl 7-methyl-3-phenyl-4,5-dihydropyrazolo[1,5-a]quinoline-2-carboxylate (29):

White solid; m.p.:79-80°C, Rf : 0.54(hexane/EtOAc, 9:1); Yield:63 %; IR (neat) : 2980,

1715,1503,1147cm-1;1H NMR (400 MHz,CDCl3) δ ppm:7.86(m, 1H), 7.84(m,2H), 7.71-

7.72(m,3H), 7.12-7.14(d,1H,J=8Hz),7.07(s,1H),4.23-4.24(q,2H),3.35-3.39(t,2H),2.95-

2.99(t,2H),2.35(s,3H),1.24(q,3H);13C NMR(100 MHz,CDCl3):163.76, 153.84, 143.63,

135.93, 133.46, 133.06,129.53, 128.89, 128.32, 127.67, 116.47, 108.95, 24.70, 21.62,

21.01, 14.21;HR-MS m/z: calcd for C21H20N2O2H[M+H]: 333.1603; found: 333.1601.

Figure 32: MASS spectra for Compound 26.

109
Results and Discussion

Figure 33: 1H NMR spectra for Compound 26.

Figure 34: 13C NMR spectra for Compound 26.

110
Results and Discussion

Figure 35: NOESY spectra for Compound 26.

Figure 36: IR spectra for Compound 27.

111
Results and Discussion

Figure 37: MASS spectra for Compound 27.

Figure 39: 1H NMR for Compound 27.

112
Results and Discussion

Figure 40: 13C NMR for Compound 27.

Figure 41: IR spectra for Compound 28.

113
Results and Discussion

Figure 42: MASS spectra for Compound 28.

Figure 43: 1H NMR for Compound 28.

114
Results and Discussion

Figure 44: 13C NMR for Compound 28.

Figure 45: IR spectra for Compound 29.

115
Results and Discussion

Figure 46: MASS spectra for Compound 29.

Figure 47: 1H NMR for Compound 29.

116
Results and Discussion

Figure 48: 13C NMR for Compound 29.

Scheme 51 gives the synthesis of Ethyl dimethyl 4,5-dihydropyrazolo[1,5-


a]quinoline-2,3-dicarboxylates:

On reaction of the sydnoquinoline with dimethyl acetylene dicarboxylate only one single

product was formed and this was confirmed using 1H NMR, 13


C NMR and Mass. The

mass spectra shows an [M+Na] peak of 309.0854.The proton NMR shows methyl

protons of the –COOCH3 at 3.98 and 3.88 as singlets. The –CH2 of the

tetrahydroquinoline ring were split into triplets and appeared at δ 3.33-3.29 ppm and δ

117
Results and Discussion

13
3.02-2.98 ppm. The aromatic protons appear between δ 7.97-7.21 ppm. The CNMR

shows 9 aromatic carbon atoms at 144.53, 143.27, 135.17, 128.44, 127.98,127.03,

117.04, 111.25, 4 aliphatic carbon atoms at 52.66,51.80,24.46,20.94 and carboxylic

carbons at 162.89 and 162.80.

The following are the physical and analytical data for dimethyl 4,5-dihydropyrazolo[1,5-

a]quinoline-2,3-dicarboxylates;

Dimethyl 4,5-dihydropyrazolo[1,5-a]quinoline-2,3-dicarboxylate (31): White solid;

m.p.: 127-128°C, Rf : 0.5 (hexane/EtOAc, 8:2); Yield:80%; IR (neat) : 2951, 1720, 1736,

1477, 1294, 1226, 1077, 760 cm-1;1HNMR (400MHz, CDCl3) δ ppm: 7.95-7.97(d, 1 H,J

=8Hz),7.33-7.37(m, 1 H),7.21-7.28 (m, 2 H), 3.98(s, 1 H), 3.88(s, 1 H), 3.20-3.33(t, 2 H),

2.98-3.02 (t, 2 H);13C NMR(100 MHz,CDCl3):162.89,162.80,144.53,143.27,135.17,

128.44,127.98,127.03,117.04,111.25,52.66,51.80,24.46,20.94; HR-MS m/z: calcd for

C15H14N2O2Na [M+Na]:309.0851;found: 309.0854.

Dimethyl 7-methoxy-4,5-dihydropyrazolo[1,5-a]quinoline-2,3-dicarboxylate (32):

White solid;m.p.: 116-117°C, Rf: 0.48(hexane/EtOAc, 8:2);Yield: 80%;IR (neat) : 2916,

2847,2615,1717,1522,1223,1021, 772cm-1;1H NMR(400 MHz,CDCl3) δ ppm:7.87-7.89

118
Results and Discussion

(d,1H),6.85-6.89(d,1H,J=8Hz),6.80(s,1H),3.98(s,3H),3.87(s,3H),3.83(s,3H),3.27-3.31(t,

2H),2.95-2.99(t,2H);13CNMR(100MHz,CDCl3):162.91,158.44,143.99,142.36,128.96,

128.62,118.31,113.97,112.71,111.06,55.52,52.62,51.76,24.87,20.96;HR-MS m/z:calcd

for C16H16N2O5Na [M+Na]:339.0957; found: 339.0956.

Dimethyl 7-fluoro-4,5-dihydropyrazolo[1,5-a]quinoline-2,3-dicarboxylate(33): pale

brown solid; m.p.:110-111°C; Rf :0.5(hexane/EtOAc, 8:2); Yield: 80%; IR(neat) : 2918,

2954,1739,1731,1471,1225,1077cm-1;1H NMR(400 MHz,CDCl3)δppm:7.90-7.94(m,1H),

6.99-7.06(m,2H),3.98(s,3H),3.87(s,3H),3.29-3.33(t,2H),2.98-3.01(t,2H);13C NMR(100

MHz,CDCl3):162.76,162.65,162.32,159.86,144.49,142.69,131.43,129.29,129.21,118.79,

118.70,115.48,114.80,114.58,111.27, 52.64, 51.80,24.56,20.70;HR-MS m/z calcd for

C15H13FN2O4Na [M+Na]:327.0757; found: 327.0752.

Dimethyl 7-methyl-4,5-dihydropyrazolo[1,5-a]quinoline-2,3-dicarboxylate (34): pale

brown solid; m.p.: 82-83°;Rf : 0.46 (hexane/EtOAc, 8:2); Yield:80%; IR (neat) : 2954,

1742,1719,1476,1224,1079cm-1;1HNMR(400MHz,CDCl3)δppm:7.83-7.85(d,1H,J=8Hz),

7.14-7.16(d, 1H, J=8Hz),7.08(s,1H),3.98(s, 3H),3.87(s, 3H),3.27-3.31(t, 2H),2.94-2.97(t,

2H),2.36(s, 3H);13C NMR(100 MHz,CDCl3):162.88, 144.22, 142.96, 136.97, 132.95,

129.00, 128.49, 126.87, 116.90, 111.14, 52.63, 51.77, 24.48, 21.02.; HR-MS m/z:calcd

for C16H16N2O4Na [M+Na]:323.1008; found: 323.1012.

119
Results and Discussion

Figure 49: IR spectra for Compound 31.

Figure 50: MASS spectra for Compound 31.

120
Results and Discussion

Figure 51: 1H NMR for Compound 31.

Figure 52: 13C NMR for Compound 31.

121
Results and Discussion

Figure 53: IR for Compound 32.

Figure 54: MASS for Compound 32.

122
Results and Discussion

Figure 55: 1H NMR for Compound 32.

Figure 56: 13C NMR for Compound 32.

123
Results and Discussion

Figure 57: IR for Compound 33.

Figure 58: MASS for Compound 33.

124
Results and Discussion

Figure 59: 1H NMR for Compound 33.

Figure 60: 13C NMR for Compound 33.

125
Results and Discussion

Figure 61: IR for Compound 34.

Figure 62: MASS for Compound 34.

126
Results and Discussion

Figure 63: 1H NMR for Compound 34.

Figure 62: 13C NMR for Compound 34.

127
Results and Discussion

Scheme 52 gives the synthesis of benzyl 4,5-dihydropyrazolo[1,5-a]quinoline-3-


carboxylate:

The scheme shows the cycloaddition of sydnoquinoline with benzyl propiolate. The

reaction leads to two isomers A and B shown below. This was due to the less steric

hinderance on one side of the triple bond. Hence, the cycloaddition could result in both

isomers.

Both the isomers were isolated by column chromatograpy using PE:EtOAc as solvent

system. The products were confirmed using Mass, 1H NMR and 13


C NMR. The mass

spectra shows [M+Na] of 327.1107 for the isomer (A) and [M+H] of 305.1292 for the

isomer (B). The proton NMR shows the protons of the –CH2 in the tetrahydroquinoline

ring as triplets at δ 2.91 ppm and δ 3.01 ppm for isomer A and δ 2.96 ppm and δ 3.30

ppm for the isomer B. A single proton was present at δ 8.89 ppm for isomer A and δ 6.69

128
Results and Discussion

ppm for isomer B. This was used to differentiate between the isomers. The aromatic
13
protons are found between δ 7-8 ppm. The C NMR reveal aromatic carbons at

163.23,142.70,142.18,136.18,135.79,128.58,128.40,128.18,128.11,127.89,126.63,126.28,

116.50,111.27 and aliphatic carbons at 65.83,24.63,20.87 for isomer A and aromatic

carbons at 162.35,143.71, 139.78,135.97,128.51,128.33,128.18,127.90,127.12,126.37,

117.78,106.78 and aliphatic carbons at 66.52,25.39,21.26 for isomer B.

1
H NMR

13
C NMR

The orientation of the isomers were confirmed by 2D-NMR. Interactions between the

proton of the tetrahydroquinoline and H of the benzyl propiolate is observed for one

isomer (B) whereas no interaction is observed for the other isomer (A).

129
Results and Discussion

The following is the physical and analytical data of the compounds benzyl 4,5-dihydro

pyrazolo[1,5-a]quinolinecarboxylates;

Benzyl 4,5-dihydropyrazolo[1,5-a]quinoline-3-carboxylate (36): Light brown solid;

m.p.: 88-89°C; Rf : 0.62(hexane/EtOAc, 8:2); Yield:35%; IR (neat) :2928, 1719, 1577,

1253, 756cm-1;1H NMR (400 MHz, CDCl3) δ ppm:8.89(s,1H),8.61-8.64(d,1H,J=12Hz),

7.25-7.44(m,6H),7.18-7.21(m, 2H),5.32(s, 2H),3.33-3.36(t, 2H),2.99-2.96(t, 2H);13C

NMR(100 MHz,CDCl3):163.23, 142.70, 142.18, 136.18, 135.79, 128.58, 128.40, 128.18,

128.11,127.89,126.63,126.28,116.50,111.27,65.83,24.63,20.87;HR-MS m/z: calcd for

C19H16N2O2H [M+H]: 305.1290; found: 305.1292.

benzyl 4,5-dihydropyrazolo[1,5-a]quinoline-2-carboxylate (40): Light brown solid;

m.p.: 110-112°C;Rf : 0.58(hexane/EtOAc, 8:2); Yield:25%;1H NMR (400 MHz, CDCl3) δ

ppm: 8.05-8.03 (d,1H,J=8Hz), 7.46-7.48 (d, 2H, J=8Hz), 7.32-7.46 (m, 4 H), 7.17-7.25

(m, 2H), 6.69 (s,1H), 5.4(s, 2H), 3.01-3.04(t, 2H),2.95-2.98 (t, 2H);13C NMR(100

MHz,CDCl3): 162.35, 143.71,139.78, 135.97, 128.51, 128.33, 128.18, 127.90, 127.12,

126.37, 117.78, 106.78, 66.52,25.39,21.26; HR-MS m/z: calcd for C19H16N2O2Na

[M+Na]: 327.1109; found: 327.1107.

130
Results and Discussion

Benzyl 7-methoxy-4,5-dihydropyrazolo[1,5-a]quinoline-3-carboxylate (37): White

solid; m.p.: 89-90°C;Rf : 0.64 (hexane/EtOAc, 8:2); Yield:40%; IR (neat) : 2928, 1719,

1577,1253,756cm-1;1H NMR (400MHz,CDCl3)δ ppm: 8.02(s,1H),7.80-7.82(d,1H,J=8Hz),

7.33-7.44(m,5H),7.25(s,1H),6.84-6.87(d,1H,J=12Hz),5.32(s, 2H),3.82(s, 3H), 3.30-3.34

(t,2H),3.82(s,3H),3.30-3.34(t, 2H),2.92-2.96(t, 2H);13C NMR(100 MHz,CDCl3):163.31,

157.89, 141.78,136.24,129.65,128.57,128.15,128.08,117.64,114.03,112.60,110.93,65.77,

55.52,25.01,20.87; HR-MS m/z: calcd for C20H18N2O3Na[M+Na]:357.1215; found:

357.1216.

Benzyl 7-methoxy-4,5-dihydropyrazolo[1,5-a]quinoline-2-carboxylate (41): White

solid; m.p.: 106-107°C; Rf : 0.59(hexane/EtOAc, 8:2); Yield:30%;1H NMR (400 MHz,

CDCl3) δ ppm:7.95-7.97(m,1H),7.46-7.48(m,2H),7.32-7.39(m,3H),6.84-6.87(d,1H,

J=12),6.77-6.78(s,1H), 6.68(s,1H),5.40(s,2H),3.82(s,3H),2.00-3.02(t,2H),2.91-2.94

(t, 2H);13C NMR(100 MHz,CDCl3):162.42, 157.97,143.09,138.98,136.03,129.89,128.71,

128.49,128.31,128.15,118.25,113.95,112.60,106.64,66.44,55.50,25.73,21.29.; HR-MS

m/z: calcd for C20H18N2O3Na [M+Na]: 357.1215; found: 357.1216.

Benzyl 7-fluoro-4,5-dihydropyrazolo[1,5-a]quinoline-3-carboxylate (38): Pale brown

solid; m.p.:93-94°C; Rf : 0.60(hexane/EtOAc, 8:2); Yield:38%;1H NMR (400 MHz,

CDCl3) δ ppm: 8.03(s,1H),7.37-7.4(m,6H),7.25(m,2H),5.31(s,2H),3.32-3.34(t,2H),2.96-

2.98(t,2H);13C NMR(100 MHz,CDCl3):142.14, 136.12, 128.59, 128.21, 128.11, 118.20,

115.43,115.20,114.67,114.44,111.33, 65.88, 24.78, 20.67; HR-MS m/z: calcd for

C19H15FN2O2H [M+H]: 323.1196; found: 323.1197.

Benzyl 7-fluoro-4,5-dihydropyrazolo[1,5-a]quinoline-2-carboxylate (42): Pale brown

solid; m.p.: 112-113°C; Rf :0.55(hexane/EtOAc, 8:2); Yield:25%,IR (neat) :2918,1713,


131
Results and Discussion

1567,1503,1254,1239,1095,1055cm-1;1HNMR(400MHz,CDCl3)δppm:7.46-7.48(m,2H),

7.33-7.39(m,4H), 7.26(s, 1H),6.96-7.05(m, 2H),5.4(s, 2H),3.02-3.04(t, 2H),2.96-2.98(t,

2H);13C NMR(100Hz,CDCl3):162.25,162.01,143.69,139.25,135.92,128.51,128.33,118.80,

115.35,115.12,114.72,114.50,106.86,66.56,60.37; HR-MS m/z: calcd for C19H15FN2O2Na

[M+Na]: 345.0757; found: 345.1018.

Benzyl 7-methyl-4,5-dihydropyrazolo[1,5-a]quinoline-3-carboxylate (39): Pale brown

solid; m.p.:87-88 °C; Rf : 0.60(hexane/EtOAc, 8:2); Yield:40% IR (neat) : ,2952, 2921,

2119,1714,1561, 1507, 1256, 1237, 1096cm-1;1H NMR (400 MHz, CDCl3) δ ppm: 8.03

(s,1H), 7.76-7.78(d,1H, J=8), 7.34-7.44(m,5H), 7.25(s,1H), 7.13-7.15(d, 1H, J=8), 7.07

(s,1H),5.31(s,2H),3.30-3.32(t,2H),2.93-2.95(t,2H),2.35(s,3H);13C NMR(100MHz,CDCl3):

162.41,143.37,139.5,136.19,136.02,133.80,128.92,128.49,128.41,128.32,128.15,126.98,

116.89,106.67,66.96,25.39,21.34,20.99; HR-MS m/z: calcd for C20H18N2O2Na [M+Na]:

341.1266; found: 341.1265.

Benzyl 7-methyl-4,5-dihydropyrazolo[1,5-a]quinoline-2-carboxylate (43): Pale brown

solid; m.p.:126-127°C; Rf : 0.57(hexane/EtOAc, 8:2); Yield:30%;1H NMR (400 MHz,

CDCl3) δ ppm:7.91-7.93(d,1H, J=8Hz),7.46-7.48(m, 2H), 7.34-7.37(m,3H), 7.13-7.15

(d,1H,J=8Hz),7.05(s,1H),6.68(s,1H),5.41(s,2H), 2.99-3.01(t,2H),2.92-2.94(t,2H),2.34

(s,3H);13CNMR(100MHz,CDCl3):162.41,143.37,139.5,136.19,136.02,133.80,128.2,

128.49, 128.32,128.41,126.98,106.67,66.46,25.39,21.34,20.99; HR-MS m/z: calcd for

C20H18N2O2Na [M+Na]: 341.1266; found: 341.1262.

132
Results and Discussion

Figure 63: IR spectra for Compound 36.

Figure 64: MASS spectra for Compound 36.

133
Results and Discussion

Figure 65: MASS spectra for Compound 40.

Figure 66: 1H NMR for Compound 36.

134
Results and Discussion

Figure 67: 1H NMR for Compound 40.

Figure 68: 13C NMR for Compound 36.

135
Results and Discussion

Figure 69: 13C NMR for Compound 40.

Figure 70: NOESY for Compound 36.

136
Results and Discussion

Figure 71: NOESY for Compound 40.

Figure 72: IR spectra for Compound 37.


137
Results and Discussion

Figure 73: MASS spectra for Compound 37.

Figure 74: MASS spectra for Compound 41.

138
Results and Discussion

Figure 75: 1H NMR for Compound 37.

Figure 76: 1H NMR for Compound 41.

139
Results and Discussion

Figure 77: 13C NMR for Compound 37.

Figure 78: 13C NMR for Compound 41.

140
Results and Discussion

Figure 79: IR spectra for Compound 38.

Figure 80: MASS spectra for Compound 38.

141
Results and Discussion

Figure 81: MASS spectra for Compound 42.

Figure 82: 1H NMR for Compound 38.

142
Results and Discussion

Figure 83: 1H NMR for Compound 42.

Figure 84: 13C NMR for Compound 38.

143
Results and Discussion

Figure 85: 13C NMR for Compound 42.

Figure 86: IR spectra for Compound 39.

144
Results and Discussion

Figure 87: MASS spectra for Compound 39.

Figure 88: MASS spectra for Compound 43.

145
Results and Discussion

Figure 89: 1H NMR for Compound 39.

Figure 90: 1H NMR for Compound 43.

146
Results and Discussion

Figure 91: 13C NMR for Compound 39.

Figure 92: 13C NMR for Compound 43.

147
Results and Discussion

4.4 In vitro anticancer studies

The synthesized molecules were subjected to in vitro cytotoxicity studies against HEp-2,

MCF-7, HeLa and HBL-100 cell lines. 5-FU was used as the standard drug for

comparison. The cytotoxicity was performed by the MTT assay (Carmichael et al.,1987).

The results are given in Tables 6-9 and Figures 94-97.

Table 6 and Figure 93 gives the IC50 values of 4,5-dihydro[1,2,3]oxadiazolo[3.4-

a]quinolin-10-ium-3-olates. The IC50 value of 5-FU is 7.8 µg/mL, 11.7 µg/mL and 9.1

µg/mL for HEp-2, MCF-7 and HeLa cell lines, respectively. The IC50 values of the

sydnoquinolines range from 30 µg/mL to 100 µg/mL. The methoxy substituted

sydnoquinoline (22) is active against MCF-7 (30 µg/mL) and HeLa (30 µg/mL). The

fluoro substituted sydnoquinoline (23) is active against HeLa (30 µg/mL). Compounds 21

and 24 were found to be moderately active against all the cell lines.

Compound no. HEp-2 MCF-7 HeLa HBL-100 cells


µg/mL µg/mL µg/mL µg/mL
21 80 60 60 70
22 70 30 30 50
23 50 60 30 40
24 60 100 50 90

Table 6: IC50 (µg/mL) values for 4,5-dihydro[1,2,3]oxadiazolo[3,4-a]quinolin-10-ium-3-


olates against the selected human cancer and normal cell lines.

148
Results and Discussion

Figure 93: IC50 values of compounds 21-24

Table 7 and Figure 94 gives the IC50 values of phenyl-4,5-dihydropyrazolo[1,5-

a]quinoline-2-carboxylates. The IC50 values of the pyrazoloquinolines 26 to 29 range

from 20 µg/mL to 90 µg/mL. The fluoro derivative (28) is found to be active against

HEp-2 and HeLa with IC50 values of 30 µg/mL and 20 µg/mL, respectively. When the

cytotoxicity was checked against breast cancer cells (MCF-7), it is found to be less active

when compared with HEp-2 and HeLa. The other molecules are found to be moderately

active.

149
Results and Discussion

Compound no. HEp-2 MCF-7 HeLa HBL-100

µg/mL µg/mL µg/mL µg/mL

26 50 40 70 60

27 40 30 40 50

28 30 90 20 70

29 70 50 70 80

Table 7: IC50 (µg/mL) values for phenyl-4,5-dihydropyrazolo[1,5-a]quinoline-2-


carboxylates against the selected human cancer cell lines.

Figure 94: IC50 values of compounds 26 to 29

150
Results and Discussion

Table 8 and Figure 95 gives the IC50 values of 4,5-dihydropyrazolo[1,5-a]quinoline-2,3-

dicarboxylates. The IC50 values for compounds 31 to 34 range from 40 µg/mL to 80

µg/mL. All the compounds are found to be active against HEp-2 cells with an IC50 value

of 40 µg/mL except 32 (70 µg/mL).

Compound no. HEp-2 MCF-7 HeLa HBL-100


µg/mL µg/mL µg/mL µg/mL
31 40 50 70 80
32 70 40 70 80
33 40 70 70 80
34 40 50 40 60
Table 8: IC50 (µg/mL) values for dimethyl 4,5-dihydropyrazolo[1,5-a]quinoline-2,3-
dicarboxylates against the selected human cancer cell lines.

Figure 95: IC50 values of compounds 31 to 34

151
Results and Discussion

Table 9 and Figure 96 gives the IC50 values of benzyl 4,5-dihydropyrazolo[1,5-

a]quinolinecarboxylates. The synthesis of these molecules resulted in two isomers. This

was due to the less steric hinderance on one side of the alkyne used for the cycloaddition.

Both the isomers were evaluated for their anticancer activity. The IC50 values of the

evaluated molecules range from 20 µg/mL to 90 µg/mL. The methoxy substituted

pyrazoloquinoline, 41 shows good activity with an IC50 value of 20 µg/mL against MCF-

7 cells and 30 µg/mL against HEp-2 cells when compared to the standard, 5-FU with an

IC50 value of 11.7 µg/mL and 7.8 µg/mL, against MCF-7 and HEp-2 cells, respectively.

Compound MCF-7
HEp-2 cells HeLa HBL-100 cells
no. cells
ug/mL ug/mL ug/mL ug/mL
36 50 70 60 80
40 60 50 40 60
37 50 60 60 70
41 30 20 50 60
38 80 90 60 80
42 60 60 80 90
39 90 40 80 70
43 60 60 50 80
Table 9: IC50 (µg/mL) values for benzyl 4,5-dihydropyrazolo[1,5-
a]quinolinecarboxylates against the selected human cancer cell lines.

152
Results and Discussion

Figure 96: IC50 values of compounds 36 to 43

The in vitro results reveal that methoxy and fluoro derivatives show good activity when

compared to the compounds with other substituents. The pyrazoloquinolines show better

activity when compared to the sydnoquinolines. This could be due to the increased

functionalization of the pyrazole ring. Benzyl 7-methoxy-4,5-dihydropyrazolo[1,5-

a]quinoline-3-carboxylate is active against HEp-2 and MCF-7 cell lines with IC50 values

of 30 µg/mL and 20 µg/mL, respectively. In the methoxy substituted pyrazoloquinolines

with benzyl acetate group attached to the pyrazole nucleus seems to be essential for the

activity because an increase in the IC50 value is observed in the presence of these groups

in the basic scaffold.

A comparison between the in silico and in vitro studies reveal that the synthesized

compounds, with the phenyl and ethylcarboxylate groups exhibit good activity as

predicted by the docking scores.

The images of the cytotoxicity studies of compounds 23, 27, 28 and 41 are given in

Figure 97. These compounds show considerably good activity when compared to others.

153
Results and Discussion

The images show the morphological difference in the treated cells after 48h of exposure

to the synthesized molecules at their IC50 values. The images show a loss in morphology

and the complete detachment of the cells after treatment.

D
Figure 97 :(A)Images of HeLa cells: Control cells and cells treated with 23 (B) Images
of MCF-7 cells: Control and cells treated with 27 (C) Images of HeLa cells: Control cells
and cells treated with 28 (D) Images of MCF-7 cells: Control and cells treated with 41.

154
Results and Discussion

FUTURE PERSPECTIVES

In the present study all the synthesized compounds show moderate to good in

vitro cytotoxic activity. Compounds 22, 23, 28 and 41 possess potent in vitro

cytotoxic activity. It is worthwhile, therefore, to study the compounds synthesized

in the present investigation for their activity in vivo.

The synthesized molecules can be taken further to design and synthesize more

potent topo II inhibitors and subject them to detailed in vivo and toxicity studies.

A detailed topo II inhibiton studies may also be carried out. QSAR and CoMFA

studies on these molecules are under progress in order to identify the active

pharmacophore. This will be effective in designing more potent molecules.

155
Summary and Conclusions

5. SUMMARY AND CONCLUSIONS

Cancer is one of the most dreaded diseases. Multidisciplinary scientific investigations are

being made today to combat this disease, but perfect cure is yet to be brought into the

world of cancer medicine. The problems associated with the current cancer chemotherapy

are non-specific toxicity and multi drug resistance (MDR).

DNA topo II inhibitors are a major target for antineoplastic agents used in the treatment

of breast, lung, and prostate cancer, sarcomas, and hematological malignancies because

of the important role played by them in the survival of cells.

Sydnones and pyrazoles are known for their broad range of biological activities. 4-

Substituted sydnones and pyrazoloquinolines have been evaluated for their anticancer,

antiinflammatory, antihypertensive, antimicrobial, antidiabetic, DNA cleavage activity

and antitubercular activities.

Although pyrazoloquinolines have been widely studied, drugs based on this nucleus are

rare. Various synthetic routes for the synthesis of sydnones and pyrazoloquinolines have

been reported. A thorough review of literature, however, revealed that there are only a

few reports on the synthesis and biological evaluation of fused ring sydnones. Sydnones

can undergo cycloaddition reactions to give pyrazoles. Fused ring sydnoquinolines and

dihydropyrazolo[1,5-a]quinolines have not so far been explored thoroughly.

The objective of the present study was, therefore, to design, synthesize and evaluate some

novel sydnoquinolines and pyrazoloquinolines for their anticancer activity in vitro using

various cell lines.

The following are some of the important finding made from the present study;

156
Summary and Conclusions

· A total of 180 compounds including 4,5-dihydro[1,2,3]oxadiazolo[3,4-a]quinolin-

10-ium-3-olates and 4,5-dihydropyrazolo[1,5-a]quinolines were designed for the

target, topo II and docking studies were carried out for the designed molecules.

The scores were compared with that of salvicine, the known topo II inhibitor.

Molecules C49, C36 and C45 showed a glide score slightly higher than that of the

salvicine.

· Post docking analysis showed various interactions of the ligands with the protein.

The molecules C48, C37, C44, C41, C39, C40, C43, C47, C35, C34, C46 and

C42 showed good glide score. The scores of these molecules are in the range of

-7.3846 to -4.0409 as compared to salvicine which is -6.9092.

· The ADME properties of the designed molecules were analyzed in silico using the

Qikprop programme in Glide, Schrodinger. All the molecules showed good

ADME predictions for all the parameters although slight deviations were

observed for a few.

· Based on the glide scores and synthetic considerations, a total of 16 novel 4,5-

dihydro[1,2,3]oxadiazolo[3,4-a]quinolin-10-ium-3-olates and 4,5-

dihydropyrazolo[1,5-a]quinolines were selected and successfully synthesized

using various synthetic schemes. They were purified and characterized using IR,

MASS, 1H NMR and 13C NMR.

· A total of 4 novel sydnoquinolines were synthesized and characterized using IR,

MASS, 1H NMR and 13C NMR.

· The synthesized sydnoquinolines were subjected to cycloaddition reaction and

successfully converted to 4,5-dihydropyrazolo[1,5-a]quinolines. The reaction was


157
Summary and Conclusions

found to be a concerted reaction as observed by the formation of single products.

No intermediates were formed or observed. The synthesized 4,5-

dihydropyrazolo[1,5-a]quinolines were characterized using IR,MASS, 1H NMR


13
and C NMR. A total of 16 novel pyrazoloquinolines were synthesized and

characterized and being reported for the first time.

· In the case of cycloaddition of pyrazoloquinolines with benzyl propiolate two

isomers were formed. These isomers were confirmed using NOESY. This helped

in confirming the interactions between the protons and thus confirming the

structures of the isomers.

· All the synthesized molecules were subjected to in vitro cytotoxicity studies

against HEp-2 (Human larynx cancer) cells, MCF-7 (breast cancer) cells, HeLa

(Cervical cancer) cells and HBL-100 (Human breast normal epithelial) cells.

· Among all the compounds screened for cytotoxicity activity, the methoxy

substituted pyrazoloquinoline 41 shows good activity with an IC50 value of 20

µg/mL against MCF-7 cells and 30 µg/mL against HEp-2 cells when compared to

the standard, 5-FU which has an IC50 value of 11.7 µg/mL and 7.8 µg/mL, against

MCF-7 and HEp-2 cells, respectively.

· The in vitro results reveal that methoxy and fluoro derivatives have good activity

when compared to other compounds.

· The pyrazoloquinolines were found to have better activity when compared to the

sydnoquinolines. This could be due to the increased functionalization of the

pyrazole ring. Benzyl 7-methoxy-4,5-dihydropyrazolo[1,5-a]quinoline-3-

158
Summary and Conclusions

carboxylate was found to be active against HEp-2 and MCF-7 cell lines with IC50

value of 30 µg/mL and 20 µg/mL, respectively.

· The in vitro results and the in silico results correlate reasonably well. Compounds

22, 23, 26, 27, 28, 31, 33, 34, 40, 41 and 39 show good in vitro cytotoxicity. Most

of these compounds show good glide score as well.

· The in silico-in vitro correlation reveal that the substitution at the R1 and R2

position of the pyrazoloquinoline is crucial for the activity. These substituents

were also important for the increase in the binding affinity in silico.

In conclusion, in the present study, 20 compounds were synthesized, characterized

and are being reported for the first time. The compounds show moderate to good

anticancer activity both in silico and in vitro. Among these compounds, compound 41

shows good activity with an IC50 value of 20µg/mL against MCF-7 cells when

compared to 5-FU which shows an activity of 11.7µg/mL. Compounds 22, 23, 28 and

41 were also found to possess fairly good in vitro anticancer activity. With the

correlated outcome of in silico and in vitro results, it may be concluded that these

compounds possess anticancer activity via topo IIα inhibition at the ATP site. These

compounds can be further taken up for the in depth in vivo studies.

159
Bibliography

6. BIBLOIOGRAPHY

Avendano C and Menendez JC. Medicinal Chemistry of Anticancer drugs. Elsevier, 2008

Abu-Hashem AA and Aly AS. Synthesis of new pyrazole, triazole and thiazolidine-
pyrimido[4,5-b]quinolines derivatives with potential antitumor activity. Arch Pharm Res.
2012, 35(3): 437-445.

Ackermann, E. On the pharmacology of sydnone and sydnonimine: A survey. Pharmazie,


1967, 22: 537.

Alice Park. The hero scientist who defeats cancer will likely never exist. Time-Medicine.
Team-Cancer;April 1, 2013.

Antony DB, Mark SB. Natural Product Chemistry for Drug Discovery, RSC publication,
edition 2009, 141.

Azarova AM, Lyu YL, Lin CP, Tsai YC, Lau JY, Wang JC and Liu LF. Roles of DNA
topoisomerase II isozymes in chemotherapy and secondary malignancies. Proc Natl Acad
Sci U S A; 2007; 104(26):11014-11019.

Baker W, Ollis WD and Poole V.D. Cyclic Mesoionic Compounds. Part III. Further
properties of sydnones and the mechanism of their formation. Journal of Chemical
Society. 1950, 1542-1551. doi: 10.1039/JR9500001542.

Barrett D, Sasaki H, Kinoshita T, Fujikawa A and Sakane K. A novel synthesis of


Pyrazolo[1,5-a]quinolines ring system. New N1-C2 bridged DNA gyrase inhibitors via a
novel tandem 1,4-conjugate addition-michael [3+2] annulation process. Tetrahedron,
1996, 52(25): 8471-8488.

Baker W, Ollis WD, Poole VD. Cyclic Mesoionic Compounds. Part I. The structure of
sydnones and related compounds.RSC; 1949: doi:10.1039/JR9490000307.

Bekhit AA, El-Sayed OA, Aboul-Enein HY, Siddiqui YM, Al-Ahdal MN. Evaluation of
some pyrazoloquinolines as inhibitors of herpes simplex virus type 1 replication. Arch.
Pharm. (Weinheim), 2005, 338:74–77.

Bjork PA.; Fex T, Pettersson, G, Soreson. P, Thrige DG. Immunomodulating compounds.


US Patent 6 642 249 B2, Nov 4, 2003.

160
Bibliography

Bonjean, K., De Pauw-Gillet MC, Defresne MP, Colson P, Houssier C, Dassonneville L,


Bailly C, Greimers R, Wright C, Quetin-Leclercq J, Tits M and Angenot L. The DNA
intercalating alkaloid cryptolepine interferes with topoisomerase II and inhibits primarily
DNA synthesis in B16 melanoma cells. Biochemistry. 1998; 37:5136-5146.

Butković K, Marinić Z and Šindler-Kulyk M. Synthesis of 3-(o-stilbenyl)sydnone and 3-


(o-stilbenyl)-4-substitutedsydnone derivatives and their antitumor evaluation, Arkivoc.
2011; 1-15.

Campbell S F, Hardstone J D and Palmer M J2,4-Diamino-6,7-dimethoxyquinoline


derivatives as alpha 1-adrenoceptor antagonists and antihypertensive agents. J Med
Chem. 1988; 31(5):1031-1035.

http://www.cancerresearchuk.org Accessed on 08/02/2013.

Chandrasekhar R and Nanjan MJ. Sydnones: A Brief Review, Mini-Reviews in Medicinal


Chemistry. 2012; 12:1359-1365.

Chandrasekhar, R; Gopalan, B. and Nanjan, M.J. Synthesis and Characterisation of 3-


Hydroxy- 4, 5-dihydro[1,2,3] Oxadiazolo [3,4-a]Quinolin-10-ium and its Fluoro
derivative. International Journal of ChemTech Research. 2011; 3(3): 1125-1128.

Chang EC, Wen YL, Chang CH, Shen YH, Wen SB, Yeh MY and Wong FF. A novel
one-pot synthesis of 2-arylpyrazoloquinolinone derivatives. Tetrahedron. 2012; 68:5920-
5924.

Colotta V, Capelli F, Lenzi O, Catarzi D, Varano F, Poli D, Vincenzi F, Varani K, Borea


PA, Ben DD, Volpini R, Cristalli G and Filacchioni G. Novel potent and highly selective
human A3 adenosine receptor antagonists belonging to the 4-amido-2-arylpyrazolo[3,4-
c]quinoline series: Molecular docking analysis and pharmacological studies. Bioorg.
Med.Chem. 2009; 17:401–410.

Davis AM, Keeling DJ, Steele J, Tomkinson NP and Tinker AC. Components of
successful lead generation. Curr. Top. Med. Chem. 2005; 5:421–439.

Deshayes C and Gelin S. Photocyclization of 5-(1-alkenyl)-1-phenylpyrazoles: A


convenient synthesis of 4,5-dihydropyrazolo[1,5-a]quinolines. Tetrahedron Lett. 1983.
24(43):4679-4682.

161
Bibliography

Deshpande SR and Pai KV. Synthesis and biological activities of certain mesoionic
sydnone compounds containing chalcone moiety. Journal of Basic and Clinical
Pharmacy. 2010; 1(3):147-152.

Duggineni S, Sawant D, Saha B and Kundu B. Application of modified Pictet–Spengler


reaction for the synthesis of thiazolo- and pyrazolo-quinolines. Tetrahedron. 2006;
62:3228–3241.

Dunkley CS and Thoman CJ. Synthesis and biological evaluation of a novel phenyl
substituted sydnone series as potential antitumor agents. Bioorg Med Chem Lett. 2003;
13:2899-2901.

Faidallah HM. and Rostom SAF. Synthesis, in vitro antitumor evaluation and DNA-
binding study of novel tetrahydroquinolines and some derived tricyclic and tetracyclic
ring systems. Eur.J. Med. Chem.2013; 63:133-143.

Fliedner, L. Ph.D. Thesis, University of Massachusetts.(Amherst), 1964.

Foster RS, Jakobi H, Harrity JPA. Regioselectivity studies of sydnone cycloaddition


reactions of azine-substituted alkynes. Tet. Lett. 2011; 52:1506–1508.

Jeffery GA. An Introduction to Hydrogen Bonding, Oxford University Press, New York,
1997.

Godaginamath GS, Pujar SR and Kavali RR. Chemoselective reaction of 1-p-acetanilido-


3-acetyl-5-hydroxy-2-methylindole towards methyl chloroacetate: Synthesis and anti-
inflammatory activity of some new 5-pyrrolyl/oxadiazolyl/triazolyl/quinazolinylmetho-
xyindole derivatives. Indian J. Chem. 2003; 42[B]: 2023.

Grynberg N, Gomes R, Shinzato T, Echevarria A and Miller J. Some new aryl-sydnones:


effects on murine tumours. Anticancer Res. 1992; 12:1025.

Hang C, Li Q, Zhu Y and Katayama H. Copper(I)-Catalyzed Tandem


Cyclization/Condensation Reaction to Novel 4,5-Dihydropyrazolo[1,5-a]quinolines and
Pyrazolo[1,5-a]indoles, Syn.Comm. 2011; 41(22):3318-3324.

http://www.centerwatch.com/patient/drugs/Druglist.html. Accessed on 08/02/2013.

Collins I and Workman P. Newer approaches to molecular cancer therapeutics. Nature


Chem. Bio. 2006; 2: 689-700.

162
Bibliography

El-Deeb IM and Lee SH. Design and synthesis of new potent anticancer pyrazoles with
high FLT3 kinase inhibitory selectivity. Bio.Med. Chem. 2010;18(11):3961–3973.

Imbriglio JE, DiRocco D, Bodner R, Raghavan S, Chen W, Marley D, Esser C, Holt TG,
Wolff MS, Taggart AKP, Waters MG, Tata JR and Colletti SL. The discovery of high
affinity agonists of GPR109a with reduced serum shift and improved ADME properties.
Bio.Med. Chem.Lett. 2011; 21:2721–2724.

Spychala J. Selective cytostatic and cytotoxic anticancer effects of bisfunctional agents:


A strategy for the design of DNA binding agents. Cancer Lett. 2009; 281:203-212.

Jenekhe SA, Lu L and Alam MM. New conjugated polymers with donor-acceptor
architectures: Synthesis and photophysics of carbazole-quinoline and phenothiazine-
quinoline copolymers and oligomers exhibiting large intramolecular charge transfer.
Macromolecules.2001;34(21):7315-7324.

Agrawal AK and Jenekhe SA, Synthesis and processing of heterocyclic polymers as


electronic, optoelectronic, and nonlinear optical materials. 2. New series of conjugated
rigid-rod polyquinolines and polyanthrazolines. Macromolecules.1993;26(5):895-905.

Jegou G and Jenekhe SA. Highly fluorescent poly(arylene ethynylene)s containing


quinoline and 3-alkylthiophene. Macromolecules. 2001;34(23):7926-7928.

Kalayanov GD, Kang SK, Cheon HG, Lee SG, Yum EK, Kim SS and Choi JK Synthesis
of Pyrazoloquinolines as Gastric H+/K+-ATPase Inhibitors. Bull. Korean Chem. Soc.
1998; 19(6):667-671.

Kalluraya B, Rahiman MA and Banji D. Sydnone derivatives: part V – synthesis and


pharmacological properties of some novel triazolothiadiazepin. Indian J Chem. 2002;
41B:1712.

Kaplan AP, Gupta V and Wasley JWF. Therapeutic pyrazolonaphthyridine derivatives.


US Patent 7 858 614 B2, Dec 28, 2010.

Kaplan AP and Gupta V. Therapeutic pyrazoloquinoline urea derivatives. US Patent 7


863 266 B2, Jan 4 2011.

163
Bibliography

Kato JY, Aoyama H and Yokomatsu T. Development of a new cascade reaction for
convergent synthesis of pyrazolo[1,5-a]quinoline derivatives under transition-metal-free
conditions. Org. Biomol. Chem. 2013;11:1171-1178.

Kier LB and Roche EB. Medicinal chemistry of the mesoionic compounds. J. Pharm Sci.
1967;56:149.

Klebe G. Recent developments in structure-based drug design. J. Mol. Med 2000;


78(5):269–281.

Kuntz ID. Structure-Based Strategies for Drug Design and Discovery. Science
1992;257(5073):1078–1082. [PubMed: 1509259]

Lambordino JG, Otterness IG and Muren JF. Benzothiopyrano[4,3-c]pyraozoles as


immunoregulatory agents. US Patent 4 268 516, May 19, 1981.

Latthe PR and Badami BV. Tandem synthesis of novel 4-(azidocarbonyl)phenylazide


from 3-(4-hydrazinocarbonyl)phenylsydnone and its chemoselective 1,3 dipolar
cycloaddition reaction and Curtius rearrangement, Org. Commun. 2011;4(2):18-25.

Lenzi O, Colotta V, Catarzi D, Varano F, Squarcialupi L, Filacchioni G, Varani K,


Vincenzi F, Borea PA, Ben DD, Lambertucci C, Cristalli G. Synthesis, structure–affinity
relationships, and molecular modeling studies of novel pyrazolo[3,4-c]quinoline
derivatives as adenosine receptor antagonists. Bioorg.Med. Chem. 2011;19:3757–3768.

Magnus PD, Iliadis T, Eisenbeis SA and Fairhurst RA. Synthesis of tetrahydroquinoline


enediyne core analogues of dynemicin. US Patent 5 442 065,Aug 15, 1995.

Mallur SG, Tiwari, AK, Chinna Raju B, Suresh Babu K, Zehra Ali A, Sastry BS,
Madhusudana Rao J. Synthesis and evaluation of phenyl substituted sydnones as potential
DPPH scavengers. Indian J Chem. 2007; 46B:1686.

Hadden M, Nieuwenhuyzen M, Osborne D, Stevenson PJ and Thompson N. Synthesis of


the heterocyclic core of the alkaloids martinelline and martinellic acid. Tet.Lett. 2001;
42:6417.

Wentland MP, Aldous SC, Gruett MD, Perni RB, Powles RG, Danz DW, Klingbeil KM.,
Peverly AD, Robinson RG. Corbett TH, Rake JB and Coughlin SA. The antitumor
activity of novel pyrazoloquinoline derivatives. Bioorg.Med.Chem.Lett. 1995; 5(4):405-
410.
164
Bibliography

Kalluraya B and Sreenivasa S.Synthesis and pharmacological properties of some


quinoline derivatives. Farmaco. 1998;53(6):399-404.

Matthews IA, Coulter TS, Ghiron C, Brennan CJ, Kamal Uddin M, Pettersson LOG,
Thrige DG and Huxley P. .Immunomodulatory compounds.US Patent 7 081 456 B2, July
25, 2006.

Michael JP.Quinoline, quinazoline and acridone alkaloids. Natl. Prod. Rep. 1997, 14,
605.

Mishra AP. 2D-QSAR study of 2, 5-dihydropyrazolo [4, 3-c] quinoline-3-one a novel


series of PDE-4 inhibitors. Int.J.Res. Pharma. and Biomed.Sci. 2012;3(1):105-109.

Chaudhuri MK and Hussain S. An efficient synthesis of quinolines under solvent-free


conditions. J. Chem. Sci., 2006;118(2):199–202.

Michael JP.Quinoline, quinazoline and acridone alkaloids. Natl. Prod. Rep. 1997, 14,
605.

Nassoy AC, Raubo P and Harrity JPA. Synthesis and cycloaddition chemistry of
oxetanyl-substituted sydnones, Tet. Lett. 2013; doi: http://dx.doi.org/10.1016/

Nithinchandra B, Kalluraya S, Aamir and Shabaraya AR. Regioselective reaction:


Synthesis, characterization and pharmacological activity of some new Mannich and
Schiff bases containing sydnone. Eur.J. Med.Chem. 2012; 54:597-604.

Onkol T, Sahin MF, Yildirim E, Erol K and Ito S. Synthesis and antinociceptive activity
of (5-chloro-2(3H)-benzoxazolon-3-yl)propanamide derivatives. Arch. Pharm Res. 2004;
27:1068-1092.

Oprea TI, Davis AM, Teague SJ and Leeson PD. Is there a difference between leads and
drugs? A historical perspective. J. Chem. Inf. Comput. Sci. 2001;41:1308–1315.

Pandey VK, Mukesh, Tandon M. Synthesis and antiviral activity of quinazolinyl


syndnones. Indian J Heterocycl Chem, 2006; 15:399.

Pinar A, Yurdakul P, Yildiz I, Temiz-Arpaci O, Acan NL, Aki-Sener E and Yalcin I.


Some fused heterocyclic compounds as eukaryotic topoisomerase II inhibitors. Biochem
Biophys Res Commun. 2004;317(2):670-4.

165
Bibliography

Woodward RB, Hoffmann. Stereochemistry of Electrocyclic Reactions J. Am. Chem. Soc.


1965; 87:395.

Rai G, Puranik VG, Kalluraya B and Hegde JC. Exclusive Formation of


1ǦArylǦ3Ǧ(5ǦnitroǦ2Ǧfuryl)Ǧ4Ǧaroylpyrazoles via Regiospecific 1,3ǦDipolar Cycloaddition
of 3ǦArylsydnones with α,βǦAcetylenic Ketones, Syn.Comm. 2006; 36(9):1285-1290.

Rai NS, Kalluraya B, Lingappa B, Shenoy S and Puranic VG. Convenient access to 1,3,4-
trisubstituted pyrazoles carrying 5-nitrothiophene moiety via 1,3-dipolar cycloaddition of
sydnones with acetylenic ketones and their antimicrobial evaluation. Eur.J.Med.Chem.
2008; 43:1715-1720.

Siegel R, Naishadham D and Jemal A. Cancer Statistics.2012. Cancer J.Clin. 2012;


62:10-29.

Reis RR, Azecedo EC, De Souza MCBV. Ferreira VF, Montenegro RC, Araujo AJ,
Pessoa C, Costa-Lotufo LV, De Moraes MO, Filho JDBM, De Souza AMT, De Carvalho
NC, Castro HC, Rodrigues CR and Vasconcelos TRA. Synthesis and anticancer activities
of some novel 2-(benzo[d]thiazol-2-yl)-8-substituted-2H-pyrazolo[4,3-c]quinolin-3(5H)-
ones. Eur.J.Med.Chem. 2011; 46:1448-1452.

Shaikh SA, Jain T, Sandhu G, Latha N and Jayaram B. From drug target to leads-
sketching: a physicochemical pathway for lead molecule design in silico. Curr. Pharm.
Des. 2007;13:3454-3470.

Satyanarayana K and Rao MNA. Synthesis of 4-[5-(substituted aryl)-4,5-dihydro-1H-


pyrazol-3-yl]-3-phenylsydnones as antiinflammatory, antiarthritic and analgesic agents.
Eur.J.Med.Chem.1995; 30:641-645.

Satyanarayana K, Deshpande SR, Subbarao B, Rao MNA. Synthesis and nitric oxide
donor activity of phenylsydnones. Indian Drugs. 2002; 39:578.

Schoeffler AJ, Berger JM. DNA topoisomerases: Harnessing and constraining energy to
govern chromosome topology. Q Rev Biophys. 2008; 41:41-101.

Selvi TS, Nadaraj V, Mohan S, Sasia R and Hema M. Solvent free microwave synthesis
and evaluation of antimicrobial activity of pyrimido[4,5-b] and pyrazolo[3,4-
b]quinolines, Bioorg. Med.Chem. 2006; 14:3896–3903.

166
Bibliography

Senff-Ribeiro A, Echevarria A, Silva EF, Franco CRC, Veiga SS and Oliveira MBM.
Cytotoxic effect of a new 1,3,4-thiadiazolium Mesoionic compound (MI-D) on cell lines
of human melanoma. Brit. J. Cancer. 2004;91:297-304.

Yang S-W, Smotryski J, McElroy WT, Tan Z, Ho G, Tulshian D, Greenlee WJ, Guzzi M,
Zhang X, Mullins D, Xiao L, Hruza A, Chan T-M, Rindgen D, Bleickardt C and
Hodgson R. Discovery of orally active pyrazoloquinolines as potent PDE10 inhibitors for
the management of schizophrenia. Bioorg. Med.Chem Lett. 2012; 22: 235–239.

Rowinsky EK, Windle JJ and Von Hoff DD. Ras Protein Farnesyltransferase: A Strategic
Target for Anticancer Therapeutic Development. J Clin Oncol 1999;17(11):3631-3652.

Stewert FHC. The chemistry of the sydnones. Chem Rev. 1964; 64:129.

Taj T, Raikar SV and. Kamble RR. Synthetic utility of sydnones to couple


pharmacologically important heterocycles for antitubercular activity. Arabian J. Chem..
2011, doi:10.1016/j.arabjc.2011.01.029

Tanimoto KO, Oi MO, Tsuboi YO and Moritani YO. Pyrazolo[1,5-a]pyrimidine


compounds as CB1 receptor antagonists. EP Patent 2 520 577 A1, November 7, 2012.

Niwa T, Takeda N, Kaneda N, Hashizume Y and Nagatsu T. Presence of


tetrahydroisoquinoline and 2-methyl-tetrahydroquinoline in Parkinsonian and normal
human brains. Biochem.Biophy. Res. Comm. 1987;144: 1084.

Toyoda E, Kagaya S, Cowell IG, Kurosawa A, Kamoshita K, Nishikawa K, Iiizumi S,


Koyama H, Austin CA, Adachi N. NK314, a topoisomerase II inhibitor that specifically
targets the α isoform. J Biol Chem. 2008; M803936200.

Tandon VK, Yadav DB, Chaturvedi AK and Shukla PK. Synthesis of (1,4)-
naphthoquinono-[3,2-c]-1H-pyrazoles and their (1,4)-naphthohydroquinone derivatives
as antifungal, antibacterial, and anticancer agents. Bioorg Med Chem Lett.
2005;15(13):3288-91.

Vlahovic G and Crawford J.Activation of Tyrosine Kinase in Cancer. The


Oncologist.2008;8:531-538.

Vogt BR. Pyrazolo[1,5-c]quinazoline derivatives and related compounds. US Patent 4


076 818, Feb 28, 1978.

167
Bibliography

Wang JC. DNA topoisomerases. Annu Rev. Biochem 1985; 54: 665-97.

Wentland MP, Aldous SC, Gruett MD, Perni RB, Powles RG, Danz DW, Klingbeil KM,
Peverly AD, Robinson RG, Corbctt TH, Rake JB and Coughlin SA. The antitumor
activity of novel pyrazoloquinoline derivatives. Bioorg. Med.Chem. Lett. 1995;5(4):405-
410.

who.int/mediacentre/factsheets/fs297/en Accessed on 08/02/2013.

Baker W, Ollis WD and Poole VD. Cyclic Mesoionic Compounds. Part III. Further
properties of sydnones and the mechanism of their formation.J. Chem. Soc. 1950;1542-
1551.

Winters G, Odaso G, Galliani G and Lerner LJ. 2-Phenylpyrazolo[1,5-a]quinolines


compounds. US Patent 4 024 149, May 17, 1977.

Wojciechowskan JK, Lange J, Ksiazek W, Gniewosz M and Rump S. Structure-activity


relationship investigations of the modulating effect of core substituents on the affinity of
pyrazoloquinolinone congeners for the benzodiazepine receptor. Il Farmaco.
1998;53:579-585.

Wu C, Fang Y, Larock RC and Shi F. Synthesis of 2H-Indazoles by the [3+2]


Cycloaddition of Arynes and Sydnones. Org Lett. 2010;12(10):2234–2237.
doi:10.1021/ol100586r.

XU Jun, GU Qiong, LIU HaiBo, ZHOU JiaJu, BU XianZhang, HUANG ZhiShu, LU


Gui, LI Ding, WEI DongQing, WANG Ling and GU LianQuan. Chemomics and drug
innovation. Science China Chemistry. 2012, 55: 1-15.

Yamato M, Takeuchi Y, Chang MR, Hashigaki K, Tsuruo T, Tashiro T and Tsukagoshi


S. Synthesis and Antitumor Activity of Fused Quinoline Derivatives. Chem. Pharm. Bull.
(Tokyo). 1990; 38(11):3048-52.

Yokoyama, N. Heterocyclic fused pyrazolo[3,4-d]pyrdin-3-ones as benzodiazepine


receptor modulators. US Patent 4 560 689, December 24, 1985.

Yokoyama N. Pyrazoloquinolines. US Patent 4 312 870, Jan 26 1982.

Zhang ZB, Wu YR and Yin CL. The efficient synthesis of new sydnones containing
fused ring, Syn. Comm. 2002; 32(14):2203-2207.

168
Bibliography

Jorgensen WL. The Many Roles of Computation in Drug Discovery. Science 2004;
303:1813-1818.

Matsugi M, Tabusa F and Minamikawa J-I Doebner–Miller synthesis in a two-phase


system: practical preparation of quinolines. Tet. Lett. 2000; 41:8523–8525.

Riego E, Bayo N, Cuevas C, Albericio F and ´ lvarez MA. A new approach to 3-


hydroxyquinoline-2-carboxylic acid. Tetrahedron. 2005;61:1407–1411.

Kim H, Gim H, Yang M, Ryu J-H and Jeon R. Design, synthesis and evaluation of
Tetrahydroquinoline-linked thiazolidinedione Derivatives as PPAR-g selective activators.
Heterocycles. 2007; 71(10):2131-2154.

Banerjee S, Ferrence GM and Hitchcock SR. Catalytic asymmetric addition of


diethylzinc to aldehydes via chiral, non-racemic b-hydroxy and b-methoxy
salicylhydrazone catalysts. Tetrahedron: Asymmetry. 2010;21:837–845.

Vlahovic G and Crawford J. Activation of tyrosine kinase in cancer. The Oncologist.


2003;8(6):531-538.

Rowinsky EK, Windle JJ and Von Hoff DD. Ras Protein Farnesyltransferase:A Strategic
Target for Anticancer Therapeutic Development. J.Cli.Onco.1999;17: 3631-3652.

Ollis WD and Ramsden CA. Mesoionic compounds. Adv Heterocycl Chem Katritzky Ar
Boulton., Ed.Academic Press Newyork.1976;19:1.

169

You might also like