10 chapter 3shodhganga.inflibnet.ac.in/bitstream/10603/76197/10/10_chapter 3.p… · compounds used...

Preview:

Citation preview

Chapter – 3

Section-A Synthesis of Cyclobutane and Quinoline Derivatives

63

Introduction

A series of N-substituted-quinolinone-3-aminocycloamides derivatives were

synthesized and evaluated for antiviral, antifungal and antibacterial activities. In general,

the derivatives were found to be potent Antifungal agents and Antibacterial agents the

results are discussed in terms of structure-activity relationships and an attempt is made to

define the structural features required for activity.

AIDS is essentially a viral disease and should be treated by antiretroviral agents.1

Although a combination of antiretroviral therapy has made it possible to suppress the

replication of HIV-1 in infected individuals to such an extent that it becomes

undetectable in the plasma, the virus persists in reservoirs such as peripheral blood

mononuclear cells or resting T lymphocytes which went back to a latent state after an

early replicative stage.2-4

Thus, after more than 3 years of multidrug therapy, it turned out

that the HIV-1 infection can be controlled but not eradicated with current treatments.

Besides, the emergence of resistance against both reverse transcriptase and protease

inhibitors, which was a frequent issue of the single-agent regimen, was not completely

suppressed by the use of combination therapy. It is therefore important to identify new

agents that could block the virus at a step of its replicative cycle which is not yet affected

by current treatments. For these reasons, the integrase (IN), the third viral enzyme which

is strictly required for the establishment of a stable infection, 5

is an attractive target for

new antiviral agents and potential synergy with currently available HIV reverse

transcriptase and protease inhibitors.6

Recently the fragment-based approach (F-BA) to drug discovery has gained

stronger attention than ever before. During the last 10 years of this methodology has been

intensively developing and now it can be successfully used as an alternative for poorly

effective.7-8

However , it should be remembered that the molecules used as fragments do

not possess high activity, but they are much more druggable than huge, sophisticated

structures usually pointed out in HTS. Thus in F-BA we are looking rather for possibility

of creating a new drug than for activity. Successful molecular scaffold should be easily

accessible and transferable into real drug. Probably the best way to achieve this aim is to

64

explore the so called privileged structures as a scaffold.9 unfortunately there are no single

rule that specify which structure is really privileged.

On the other hand quinoline and its derivatives such as quinoline antibiotic can be

the most spectacular example of the potential efficiency of this system.10

Quinine

molecule, which also contain quinoline moiety, proves the nature preference for the

system. Quinoline scaffold can be also found in many classes of other biologically active

compounds used as antifungal, antibacterial and antiprotozoic drug 11-14

as well as

antituberculotic agents.15

some quinoline analogues shows antineoplastic activity.

Through knowledge of all interaction which occur in solutions or crystals of

molecules proposed as scaffolds is very important for better understanding its fate in

biological system and for SAR studies Certain specific interactions are able to change

crucial parameters of molecules or even to change the structure via shift of tautomeric

equilibrium.16

Quinoline-5, 8-dione is substantial molecular fragment of lavndomycin

and related compounds. Lavendomycin was originally isolated from the fermented broth

of Streptomyces, lavenduale and the compounds of this class were identified in the 1970s

as anti-tumour agents. Although the toxicity of Lavendomycin makes it unsuitable for

clinical use, its activity has inspired several investigations.17-20

Quinolinone- based antibiotics

N

O

O

N

N

H2NCOOH

CH3

N

O

O

N

H2N

H2NCOOH

CH3

MeO

OH

OMe

OMe

Lavendamycin Streptonigrin

N

O

O

HN

H2N

H2N

CH3

MeO

OH

OMe

OMe

O

Streptonigrone

Styrylquinoline as HIV Integrase inhibitor

65

Styrylquinoline have been recognized as potent HIV-1 IN inhibitors that block

HIV-1 replication in cell based assays.21

with the aim to explore their mechanism of

action, various novel Styrylquinoline derivatives were synthesized and their biological

properties were evaluated. Since Styrylquinoline fall within the general category of

inhibitors characterized by two aryl groups linked through a central spacer, we have

examined the molecular determinants of each of these subunits, through a progressive

series of analogues. These results clearly demonstrate that the presence of a free carboxyl

group at C-7 and a hydroxyl at C-8 in the quinoline half is required for the biological

activity of the drug. The other prerequisite for in vitro potency was the presence of an

ancillary aromatic ring.

However the in vitro inhibitory profile tolerates the introduction of various

substituent’s in this ring or its replacement by heteroatomic nuclei. The structural

requirements for ex vivo activity are more stringent than for in vitro IN inhibition. In

addition to the presence of an o-hydroxyl acid group in the quinoline, the anti-HIV

activity indeed requires the presence of one ortho pair of substituent’s in the ancillary

phenyl ring: a free hydroxyl group at C-4 and a hydroxyl or methoxy group at C-3.

Examination of SARs developed for our compounds, interpreted in the light of literature

data, led to a putative binding mode of Styrylquinoline to HIV-1 IN. Since the Mg2+

ion

is a cofactor required for the enzymatic activity of HIV-1 IN, we hypothesized that the o-

hydroxyl acid group lies in a possible coordination to this metal cation. The additional

binding force which plays a significant role in the recognition of the drug by the protein

apparently involves the ancillary aromatic ring.

However, since the exact mechanism by which Styrylquinoline inhibited

productive infection is unknown, the definitive mapping of the binding sites of these

inhibitors awaits further investigation. In summarizing the above findings, it can be

concluded that the insights so gained are likely to aid the design and development of new

useful HIV-1 IN inhibitors. HIV-1 IN has indeed become an attractive target for

intervention by chemotherapeutics, because of the rapid emergence of resistance against

66

both reverse transcriptase and protease inhibitors, currently used in combination regimens

for the treatment of AIDS.22

Binding mechanism of Styrylquinoline related compound to active site of HIV

integrase inhibitor

Most of HIV integrase inhibitors are be lived to interact with divalent metal

cations such as magnesium or manganese, located at the protein active site.23

Crystal

structures of the core domain of HIV-1 IN mutants complexes with Mg2+

have been

reported.24

The magnesium is coordinated in the active site by Asp- 64 and Asp-116, whereas

the third residue of the catalytic amino acid triad, Glu-152, does not directly participate in

metal binding. Such a binding site was unequivocally identified by X-ray structure

determination of a complex between a naphthalene disulfonic inhibitor (Y-3) and ASV

IN, a protein which exhibits great similarity with HIV-1 IN.25

The Y-3 molecule in the

crystal is located in close proximity to the enzyme active site. Recently, the X-ray

structure of the HIV-1 IN catalytic domain complexes with an inhibitor (5CITEP) has

been determined. The inhibitor binds centrally in the active site of the IN and makes a

number of close contacts with the protein. Only minor changes in the protein accompany

inhibitor binding.26

Since it was recognized that HIV-1 IN requires a divalent cation as cofactor for its

enzymatic activity (presumably Mg2+

), and assuming that an o-hydroxyl acid group in the

quinoline subunit is crucially involved in both in vitro and ex vivo inhibitory activities,

they hypothesized that this group lies in a possible coordination to the metal cation. Such

an assumption has been recently reinforced by computational studies. Indeed,

minimization of the energy of interaction between Styrylquinoline drugs on the whole

surface of the catalytic core domain of RSV IN revealed that the best fits were found for

binding of the drugs on Mg2+

in the vicinity of the active site.27

Recently retroviral has been elucidated by mean crystal structure of IN from PFV

virus.28-29

Designing drugs is a complex issue that still lacks general approach. Although

we usually do not realize that, the lack of the appropriate wide spectrum biological data is

one of the important problems. In fact, this contributes to the low efficiency of the current

67

molecular design. Thus, molecular modeling provides us with molecular data describing

small molecule effectors. However, we cannot model so efficiently biological records. On

the other side, only very few experimental information are available on such compounds

with the exception of the certain activities under investigations. It has been suggested that

QSAR efficiency could be significantly improved by the incorporation into the respective

models of not only structural, physical or chemical parameters but also a spectrum of

biological activities. From the medicinal chemist point of view this strategy suggests that

we should investigate and report the data for a variety of biological effects. Quinoline

moiety is present in many classes of biologically active compounds.

A number of them have been clinically used as antifungal, antibacterial, and

antiprotozoic drugs as well as antineoplastics.30

Styrylquinoline derivatives have gained

strong attention recently due to their activity as perspective HIV integrase inhibitors.31–34

Quinoline Salicylic Acids derivatives and SAR as P-selectin activity

Letter on Neelu Kaila and coworkers defined SAR of Quinoline Salicylic Acids

and show that both hydroxyl and carboxylic acid must require for P-selectin activity.

N

O OH

OH

Boths groups are required for Activity.

R1

R8

R7

R6

small groups are prefferdeg.R= Cl,Br, I-Pr, N-(Et)2, CF2,OCF3Polar groups are inactiveAryl groups are not activeAmide functionality is not tolerated.

R8= hydrophobic groups areprefferdsmall steric bulk is toleratedhydrophelic groups have generally negative impact on activityAryl substitution result in increase in activity.

R7 = Substitution at seven position does not lead to potent compound.

R6 = Substitution at six position does not lead to potent compound

Figure 3.132

68

4-Hydroxy 3-quinolinecarboxamide derivatives

Roquinimex-related 3-quinolinecarboxamide derivatives were prepared and

evaluated for treatment of autoimmune disorders.36

For a long time Katarzyna and

coworkers had been interested in quinoline chemistry 7 and this situation prompted us to

study 4-hydroxy-3-quinolinecarboxamides, which could actually be regarded as possible

bioisosteres of oxicams I.37

Katarzyna and coworkers selected three basic quinoline

scaffolds as a fragment of HIV integrase inhibitors. Their crystal structures and in vitro

activities were collected and molecular docking were performed.38

O

O

NH

O

1

N

OH

O

OH

2

N

OHO

HO

3

Figure 3.2

Some differences in the molecular interaction between inhibitors and enzyme

seem to elucidate the activity pattern. Most active scaffold of 5-hydroxy-quinaldine -6-

carboxylic acids (Scaffold2) acids was found to be more efficient in hydrogen bonding

interaction with enzyme active site other than two compounds.

69

Present work

Our conclusion from literature survey

1) Quinoline pharmacocore is important for biological activity.

2) Both acid and hydroxyl groups must be ortho required for HIV Integrase activity.

3) In case of aryl substituent on basic quinoline pharmacocore has less impact on Anti

HIV activity.

4) Aryl substituent at 8 position of quinoline salicylic acid must be tolerated.

5) Substituent on any other does not give potent compound.

6) According to literature survey and best of our knowledge nobody had tried. Synthesis

of compound having hydroxyl and amide groups ortho to each other and does not test

against Anti HIV activity.

7) We had changed position of hydroxyl and carboxylic acid group.

Keeping these things in mind we have designed novel series of compounds with

different variations. In summery we have done

1) Basic quinoline Scaffold was kept constant.

2) Put cycloamide and hydroxyl groups are put on quinoline ring at 3&4 position

hypothetical mechanism of these groups with magnesium cation has derived using

literature.

3) Aromatic substitution on 8 positions may lead to increase in activity.

General Structure of Proposed compound

N

NH

OOH

Basic quinoline pharmacocore

Both groups required for activity

newly praposed novel amide group.

novel substitutent at 8 position.

R

Figure 3.3 General Structure of Proposed compound

70

Hypothetical mechanism of designed quinoline cyclobutyl carboxyamide

OH

O

HN

Mg2+

O

O

N

OH

OH

O

N

H

OASP64ASP116

Ar

Figure 3.4 Hypothetical mechanism of designed quinoline cyclobutyl carboxyamide derivative –

magnesium cation interaction at the active sites of HIV integrase.

Retro synthetic approach for synthesis of proposed compound

N

NH

OOH

R

N

NH

OOH

BrN

Br

OH

OOH

NH2

Br

DEMM

Figure 3.5 Retro synthetic approaches for synthesis of designed compound

71

GENERAL APPROACHES TO 4-HYDROXY QUINOLINE

OH

OH

O

NH

H

O

COOR

NH

COOR

COOR

X

O

COOR

NR2

NH2

O

COOR

R2

R1

O

R1 = OCH3; R2 = NHCOArR1= NEt2; R2 -N=NR2

Based- catalysedcyclisation

Cyclization usingsubstituted o-N-vinylamino derivaties

FriedelCraft enaminoester cyclization.

Intramolecular SNAr using halogenated aminovinyl phenyl ketones.

Closure by an intramolecular michel reaction

Figure 3.6 Current approaches to the synthesis of 4-hydroxy quinoline.

72

Results and Discussion

The 4-hydroxyl-3-carboxy cyclobutyl amide analogues were synthesized 14 as

shown in Scheme 2. 1. Starting from ortho bromo aniline. O-Bromo Aniline was reacted

with diethoxyethylmethlene malonate to give Michel type of adduct (Compound14). This

on refluxing in biphenyl ether gives compound 15. Further compound 15 was reacted

with different boronic acid under Suzuki reaction conditions gives compound (15A-15G).

Br

NH2

Br

NH

COOEt

COOEt

N

Br

OH

O

O

N

Br

OH

OOH

N

O

N

OH

Br

N

O

N

OH

a b c

d

e

13 1415 16

1718A-18GR

N

O

OEt

OH

R

N

O

OH

OH

R

15A-15G 16A-16G

b1 c1

Scheme 3.1 Synthesis of 8-bromo- N-cyclobutyl-4-hydroxyquinoline-3- carboxamide

Reactions and conditions: a) Diethoxy methyl ethylene malonate, o-bromo aniline heat

at 90 0C, 2 h, 92%; (b) Biphenyl ether, 270

0C, 3 h, 88%; Toluene, (b1) Phenyl Boronic

Acid, Tetrakis, CS2CO3, 111 0C, 2 h, 78%; (c) 10%Aq. NaOH, THF, 70

0C, 2.5 h, 74.5%;

(c1)Toluene, Phenyl Boronic Acid, Tetrakis, CS2CO3, 111 0C, 2 h, 65%; (d) DMF, EDCI,

HOBT, NMM, rt, 4 h, 90%; (e) Toluene, Phenyl Boronic Acid, Tetrakis, CS2CO3, 111

0C, 2 h, 60%.

73

Compound 15A-15G may acts as prodrugs in biological system. Compound 15

was hydrolyzed to give 4-hydroxy 3- carboxylic acid quinoline derivatives (compound

16). Compound 16 was reacted with different boronic acid to give Compound (16A-

16G). Compound 16 was converted in to cyclobutyl amide derivatives compound (17)

using EDCI, HOBT, NMM, in DMF. Compound 17 was reacted with different boronic

acid under Suzuki reaction condition gives Compound (18A-18G).

All synthesized compounds (15A-15G, 16A-16G, 18A-18G) were characterized

by NMR, ms and HPLC. Few of selected compounds were submitted for Anti HIV

activity. Biological activity result and SAR of compounds were discussed in chapter 5.

74

Synthesis of diethyl 2-((2-bromophenylamino)methylene)malonate (14)

Br

NH

COOEt

COOEt

A suspension of 2-bromo aniline (1)5gm (31mmol) diethyl ethoxymethylene malonate

(8g, 37.2mol) was heated to 110 0C for 4h. The reaction mixture was cooled to rt. The

solid thus formed was taken in pet ether and stir for 15 min and filtered to get Compound

2 as a white crystalline solid 9.7g (95% yield.

Spectral data

IR (KBr, Cm-1) v: 3467(NH), 1709(C=O), 1765(C=C); 1H NMR (300MHz,CDCL3)

δppm: 8.45 (d, J = 9.6Hz, 1H),7.62 (m, 2H), 7.36 (d, J = 6.1Hz, 1H), 7.25 (m, 1H), 4.34

(q, 2H), 1.38 (t, J = 5.3Hz, 3H), 1.35 (t, J = 5.3Hz, 3H); MS (ESI+) (342.3) Calcd for

C14H16BrNO4 : 341.03.

Synthesis of ethyl 4-hydroxy-8-phenylquinoline-3-carboxylate (15)

N

OH

OEt

O

Br

Compound 2 (9g, 27mmol) was heated at 250 0C for 4hr in dry medium, the reaction

mixture was cooled to rt and added hexane (200ml) and stired for 15 min, the precipited

solid was filtered and dried to get compound 3 as white solid. 8.8gm (88% yield.

Spectral data

IR (KBr, Cm-1) v: 3519(OH), 1709(C=O), 1H NMR (300MHz ,CDCL3) δppm: 11.68

(br s, 1H), 9.68 (s, 1H), 8.47 (m, 2H), 7.59 (t, J = 7.8Hz, 1H), 4.24 (q, 2H), 1.29 (t, J =

7.2Hz, 3H); MS(ESI+) Calcd. For C10H12BrNO3; 294.98.

75

General process for synthesis of compound (15A-15G)

To a solution of 3 (2.99 g, 9.83 mmol), CS2CO3 (4.076 g, 29.49 mmol), and

Tetrakis (0.80 g, 0.98 mmol) in dry Toluene (15 mL) was added phenyl boronic acid

(1.2334 gm, 20994 mmol).

The reaction mixture was heated to reflux at 111 0C for 6 h, then cooled to

ambient temperature, poured into a water (50 mL), and extracted with EtOAc (3 _ 40

mL). The combined organic layer was washed with brine (50 mL), dried over MgSO4,

and evaporated. The crude solid was purified over silica gel-60 via MPLC (EtOAc/pet

ether) to afford 2.64 g (91% yields) of a solid.

76

Spectral data

Ethyl 4-hydroxy-8-(4-methoxyphenyl) quinoline-3-carboxylate (15A)

N

OH

OEt

O

OMe

Nature: Solid; MP: 172-174 oC; Yield: 75%;

1H NMR (300MHz, DMSO-d6) δppm;

9.68 (s, 1H), 8.01- 7.90(m, 2H), 7.74 (d, J = 7.5Hz, 2H) 7.59 (t, J = 7.8Hz, 1H), 7.42 (d, J

= 7.5Hz, 2H), 4.24 (q, 2H), 3.95 (s, 3H), δ1.29 (t, J = 7.2Hz, 3H); MS (ESI+) m/z: 324.3

(M+1) Calcd. For C19H17NO4: 323.3.

Ethyl-8-(4-fluorophenyl)-4-hydroxyquinoline-3-carboxylate (15C)

N

OH

OEt

O

F

Nature: Solid; MP: 170-171 oC; Yield: 77%;

1H NMR (300MHz, DMSO-d6) δppm;

9.68 ( s, 1H), 8.01-7.91 (m, 2H), 7.70 (d, J = 7.5Hz, 2H) 7.54 (t, J = 7.8Hz, 1H), 7.36 (d,

J = 7.5Hz, 2H), 4.24 (q, 2H), δ1.29 (t, J = 7.2Hz, 3H); MS(ESI+)m/z: 312.3 (M+1) Calcd.

For C18H14FNO3 : 311.3.

Ethyl 4-hydroxy-8-(4-(methyl sulfonyl) phenyl) quinoline-3-carboxylate (15D)

N

OH

OEt

O

SO

O

77

Nature: Solid; MP: 178-179 oC; Yield: 80%;

1H NMR (300MHz, DMSO-d6) δppm;

9.68 ( s, 1H), 8.01- 7.88(m, 2H), 7.70 (d, J = 7.5Hz, 2H) 7.54 (t, J = 7.8Hz, 1H), 7.58 (d,

J = 7.5Hz, 2H), 2.68 ( s, 3H), 4.24 (q, 2H), δ1.29 (t, J = 7.2Hz, 3H); MS(ESI+) m/z: 372.3

(M+1) Calcd. For C19H17NO5S: 371.4.

Ethyl 4-hydroxy-8-phenylquinoline-3-carboxylate (15E)

N

OH

OEt

O

Nature: Solid; MP: 176-178 oC; Yield: 82%;

1H NMR (300MHz, DMSO-d6) δppm;

9.68 ( s, 1H), 8.01(d, J = 7.5Hz, 1H), 7.70 – 7.68 (m, 2H), 7.54 – 7.48 (m, 3H), 7.40 (d, J

= 7.5Hz, 2H), 4.24 (q, 2H), δ1.29 (t, J = 7.2Hz, 3H); MS(ESI+) m/z: 294.3 (M+1); Calcd.

For C18H15NO3: 293.2.

Ethyl 4-hydroxy-8-P-tolylquinoline-3-carboxylate (15F)

N

OH

OEt

O

Me

Nature: Solid; MP: 178-179 oC; Yield: 78%;

1H NMR (300MHz, DMSO-d6) δppm;

9.68 ( s, 1H),7.98-7.88(m, 2H), 7.70 (d, J = 7.5Hz, 2H) 7.54 (t, J = 7.8Hz, 1H), 7.37 (d, J

= 7.5Hz, 2H), 2.31 ( s, 3H), 4.24 (q, 2H), δ1.29 (t, J = 7.2Hz, 3H); MS(ESI+) m/z: 308.3

(M+1) Calcd. For C19H17NO3: 307.3.

Ethyl 8-(4-cyanophenyl)-4-hydroxyquinoline-3-carboxylate (15G)

78

N

OH

OEt

O

CN

Nature: Solid; MP: 189-190 oC; Yield: 70%;

1H NMR (300MHz, DMSO-d6) δppm;

9.68 ( s, 1H),7.98-7.88(m, 2H), 7.70 (d, J = 7.5Hz, 2H) 7.54 (t, J = 7.8Hz, 1H), 7.48 (d, J

= 7.5Hz, 2H), 4.24 (q, 2H), δ1.29 (t, J = 7.2Hz, 3H); MS(ESI+) m/z: 319.3 (M+1) Calcd.

For C19H17NO3: 318.3.

79

Synthesis of 8 – bromo-4-hydroxyquinoline-3-carboxylic acid (16)

N

Br

OH

OH

O

To A solution of 3 (4gm, 1.222mmol) in THF (50ml) 10 % Aq. NaOH (20mL)

was added. The reaction mixture was stirred at rt for 4 hr and reaction was monitored by

TLC.

After complete disappearance of starting material from TLC RM was concentrate

to remove THF on rotavapour. Residue was diluted with water and neutral ph was

maintained by using dil. HCl solution and compound was extracted in DCM (2 × 25 mL).

Combined DCM layer was dried over anhydrous sodium sulphate and concentrate on

rotaevapo. To get compound (4)

white solid. 1H NMR (300MHz, CDCl3) δppm: 11.68 (br s, 1H, OH), 10.02 (s, 1H), 8.47

(m, 2H), 7.59 (t, J = 7.8Hz, 1H), MS(ESI+) m/z: 269.1 (M+1) Calcd. For C10H6BrNO3;

268.1

General Procedure for synthesis of compound (16A-16G)

To a solution of 16 (2.99 g, 9.83 mmol), CS2CO3 (4.076 g, 29.49 mmol), and

Tetrakis (0.80 g, 0.98 mmol) in dry Toluene (15 ml) was added phenyl boronic acid

(1.2334gm, 20994mmol).

The reaction mixture was heated to reflux at 111 0C for 6 h, then cooled to

ambient temperature, poured into water (50 mL), and extracted with EtOAc (3x40 mL).

The combined organic layer was washed with brine (50 mL), dried over Na2SO4, and

evaporated. The crude solid was purified over silica gel-60 via MPLC (EtOAc/hexane) to

afford 2.64 g (91% yield) of a solid.

80

Ethyl 4-hydroxy-8-(4-methoxyphenyl) quinoline-3-carboxylic Acid (16A)

N

OH

OH

O

OMe

Nature: Solid; MP: 272-274 oC; Yield: 82%;

1H NMR (300MHz, DMSO-d6) δppm;

10.47 (s, 1H), 8.01- 7.90(m, 2H), 7.74 (d, J = 7.5Hz, 2H) 7.59 (t, J = 7.8Hz, 1H), 7.42 (d,

J = 7.5Hz, 2H), 4.10 (t, 3H); MS(ESI+) m/z: 296.3 (M+1) Calcd. For C17H13NO4: 295.3.

Ethyl-8-(4-fluorophenyl)-4-hydroxyquinoline-3-carboxylic Acid (16C)

N

OH

OH

O

F

Nature: Solid; MP: 260-262 oC; Yield: 80%;

1H NMR (300MHz, DMSO-d6) δppm;

10.47 (s, 1H), 8.01- 7.90(m, 2H), 7.74 (d, J = 7.5Hz, 2H) 7.59 (t, J = 7.8Hz, 1H), 7.38 (d,

J = 7.5Hz, 2H); MS (ESI+) m/z: 284.3 (M+1) Calcd. For C16H10FNO3 : 283.3.

Ethyl 4-hydroxy-8-(4-(methyl sulfonyl) phenyl) quinoline-3-carboxylic acid(16D)

N

OH

OH

O

SO

O

81

Nature: Solid; MP: 273-275 oC; Yield: 82%;

1H NMR (300MHz, DMSO-d6) δppm;

10.47 (s, 1H), 8.01- 7.90(m, 2H), 7.74 (d, J = 7.5Hz, 2H) 7.59 (t, J = 7.8Hz, 1H), 7.44 (d,

J = 7.5Hz, 2H); 2.68 (s, 3H); MS (ESI+) m/z: 344.4 (M+1) Calcd. For C19H17NO5S:

343.1.

Ethyl 4-hydroxy-8-phenylquinoline-3-carboxylic Acid (16E)

N

OH

OH

O

Nature: Solid; MP: 272-274 oC; Yield: 82%;

1H NMR (300MHz, DMSO-d6) δppm;

10.47 (s, 1H), 8.22 (s, 1H) 8.01(d, J = 7.5Hz, 1H), 7.70 – 7.68 (m, 2H), 7.54 – 7.48 (m,

3H), 7.42 (d, J = 7.5Hz, 2H); MS(ESI+) m/z: 266.3 (M+1); Calcd. For C18H15NO3: 265.1.

Ethyl 4-hydroxy-8-P-tolylquinoline-3-carboxylic Acid (16F)

N

OH

OH

O

Me

Nature: Solid; MP: 271-272 oC; Yield: 82%;

1H NMR (300MHz, DMSO-d6) δppm;

10.47 (s, 1H), 8.01- 7.90(m, 2H), 7.74 (d, J = 7.5Hz, 2H) 7.59 (t, J = 7.8Hz, 1H), 7.40 (d,

J = 7.5Hz, 2H), 2.31 (s, 3H), 4.24 (q, 2H), δ1.29 (t, J = 7.2Hz, 3H); MS(ESI+) m/z: 280.3

(M+1) Calcd. For C17H13NO3: 279.3.

Ethyl 8-(4-cyanophenyl)-4-hydroxyquinoline-3-carboxylic Acid (16G)

82

N

OH

OH

O

CN

Nature: Solid; MP: 275-276 oC; Yield: 82%;

1H NMR (300MHz, DMSO-d6) δppm;

10.47 (s, 1H), 8.01- 7.90(m, 2H), 7.74 (d, J = 7.5Hz, 2H) 7.59 (t, J = 7.8Hz, 1H), 7.48 (d,

J = 7.5Hz, 2H); MS (ESI+) m/z: 319.3 (M+1) Calcd. For C19H17NO3: 318.3.

83

Synthesis of 8-bromo-N-cyclobutyl-4-hydroxyquinoline-3-carboxamide (17)

N

O

N

OH

Br

N-methylmorpholine (12ml,4.43mmol),cyclobutyl amine(13 mg,0.18mmol), 1-

hydroxybenzotriazole(30mg, 0.22mmol), 1-3(3-dimethylamino-propyl)-3-

ethylcarbodiimide hydrochloride(32mg,0.17mmol)were added to a solution of compound

4 in DMF(Dimethyl Formamide). After stirring at room temperature for 6h reaction was

monitored by TLC. The rm was evaporated under reduced pressure to remove DMF and

DCM was added. The DCM layer was washed with an aqueous solution of sodium

bicarbonate solution, with brine (2x3mL) and dried over sodium sulphate and evaporated

under reduced pressure. The crude residue was purified by column chromatography,

Ethyl Acetate: Pet ether as a solvent on combi flash to get compound 5 (46mg,

81%Yield).

1H NMR (300MHz, DMSO-d6)

δppm; 11.68 (br s, 1H,OH), 10.47(s,1H), 8.47 (m, 2H)

7.59 (t, J = 7.8Hz, 1H), 3.73(m, 1H), 2.32- 2.28 (m 4H) 2.1(m, 2H); MS(ESI+) m/z:

222.2 (M+1) Calcd. For C14H13BrN2O3; 221.3.

84

General procedure for Synthesis of Compound (18A-18G)

To a solution of 17 (2.99 g, 9.83 mmol), CS2CO3 (4.076 g, 29.49 mmol), and Tetrakis

(0.80 g, 0.98 mmol) in dry Toluene (15 mL) was added phenyl boronic acid (1.2334gm,

20994mmol). The reaction mixture was heated to reflux at 111 0C for 6 h, then cooled to

ambient temperature, poured into a water (50 mL), and extracted with EtOAc (3 x 40

mL). The combined organic layer was washed with brine (50 mL), dried over Na2SO4,

and evaporated. The crude solid was purified over silica gel-60 (EtOAc/hexane) to afford

2.64 g (91% yield) of a solid.

85

Spectral data

N-cyclobutyl-4-hydroxy-8-(4-methoxyphenyl) quinoline-3-carboxamide (18A)

N

O

NH

OH

OMe

Nature: Solid; MP: 162-164 oC; Yield: 77%;

1H NMR (300MHz, DMSO-d6) δppm;

10.47 (s, 1H), 7.98-7.88(m, 2H), 7.70 (d, J = 7.5Hz, 2H) 7.54 (t, J = 7.8Hz, 1H), 7.42 (d,

J = 7.5Hz, 2H), 3.95(s, 3H),3.73(m, 1H), 2.32- 2.28 (m, 4H), and 1.9(m, 2H).

N- cyclobutly-4-hydroxy-8-p-tolylquinoline-3-carboxamide (18B)

N

O

NH

OH

Me

Nature: Solid; MP: 168-170 oC; Yield: 70%;

1H NMR (300MHz, DMSO-d6) δppm;

10.47 (s, 1H), 7.98-7.88(m, 2H), 7.70 (d, J = 7.5Hz, 2H) 7.54 (t, J = 7.8Hz, 1H), 7.42 (d,

J = 7.5Hz, 2H), 3.95(s, 3H),3.73(m, 1H), 2.32- 2.28 (m, 4H), 1.9(m, 2H); MS(ESI+) m/z:

333.4 (M+1) Calcd. For C21H20N2O2: 332.4

N- cyclobutyl -8-(4-fluorophenyl)-4hydroxyquinoline-3-carboxamide (18D)

N

O

NH

OH

F

86

Nature: Solid; MP: 172-174 oC; Yield: 73%;

1H NMR (300MHz, DMSO-d6)

δppm;10.47 (s, 1H), 7.98-7.88(m, 2H), 7.70 (d, J = 7.5Hz, 2H) 7.54 (t, J = 7.8Hz, 1H),

7.37 (d, J = 7.5Hz, 2H), 3.95(s, 3H),3.73(m, 1H), 2.32- 2.28 (m, 4H), 1.9(m, 2H);

MS(ESI+) m/z: 337.3 (M+1) Calcd. For C20H17FN2O2: 336.4

8-(4-cyanophenyl)-N-cyclobutyl-4-hydroxyquinoline-3-carboxamide (18F)

N

O

NH

OH

CN

Nature: Solid; MP: 172-174 oC; Yield: 82%;

1H NMR (300MHz, DMSO-d6) δppm;

10.47 (s, 1H), 7.98-7.88(m, 2H), 7.70 (d, J = 7.5Hz, 2H) 7.54 (t, J = 7.8Hz, 1H), 7.48 (d,

J = 7.5Hz, 2H), 3.95(s, 3H),3.73(m, 1H), 2.32- 2.28 (m, 4H), 1.9(m, 2H); MS(ESI+) m/z:

344.4 (M+1) Calcd. For C21H17N3O2: 343.3.

87

Representative Spectra

1H NMR OF COMPOUND 16A

MS OF COMPOUND 16A

88

HPLC OF COMPOUND16A

1H NMR OF COMPOUND 18A

89

HPLC OF COMPOUND 18A

90

References

1. Ho, D. D.; Neumann, A. U.; Perelson, A. S.; Chen, W.; Leonard,J. M.;

Markowitz, M.; Nature 1995, 373, 123.

2. Zhang, L.; Ramratnam, B.; Tenner-Racz, K.; He Y.; Vesanen, M.; Lewin, S.;

Talal, A.; Racz, P.; Perelson, A.; Korber, B.; Markowitw, M.; Ho, D.; N. Engl. J.

Med. 1999, 340, 1605.

3. Furtado, M.; Callaway, D.; Phair, J.; Kunstman, K.; Stanton, J.; Macken, C.;

Perelson, A.; Wolinsky, S.; N.; Engl. J. Med. 1999,340, 1614.

4. Finzi, D.; Silliciano, R. F.; Cell 1998, 93, 665.

5. Lafemina, R. L.; Schneider, C. L.; Robbins, H. L.; Callahan, P. L.; Legrow, K.;

Roth, E.; Schleif, W. A.; Emini, E. A.; J. Virol.1992, 66, 7414.

6. De Clerq, E.; J. Med.Chem. 1995, 38, 2491.

7. Rees, D. C.; Congreve, M.; Murray, C. W.; Carr, R.; Nat. Rev. Drug Disc. 2004,

3, 660.

8. Hajduk, P. J.; Greer, J.; Nat. Rev. Drug Disc. 2007, 6, 211.

9. Kubinyi, H. Fischer, J., Ganellin, C. R., Eds.; Wiley- VCH: Weinheim, 2006, 53.

10. Petersen, U. Quinolone Antibiotics: The Development of Moxifloxacin.

InAnalogue Based Drug Discovery; Fischer, J., Ganellin, C. R., Eds.; VCH-Wiley

Verlag GmbH & Co., 2006; pp 315–370.

11. Fostel, J. M.; Lartey, P. A.; Drug Discovery Today, 2000, 5, 25.

12. Zainaba, D.; Meryem, L.; Abdelmejid, B.; Abdelfatah, A.; Mohammed, H.; Said,

K.; Mohammed, B.; Mohammed, B.; Farmaco, 2004, 59, 195.

13. Musiol, R.; Serda, M.; Hensel-Bielowka, S.; Polanski, J.; Curr. Med. Chem.,

2010, 17, 1960.

14. Majerz-Maniecka, K.; Oleksyn, B.; Musiol, R.; Podeszwa, B.; Polanski, J.; In

Sci. Pharm. 2005; 73, 194.

15. Urbina, J.; Cortes, J.; Palma, A.; Lopez, S.; Kouznetsov, V. V.; Zacchino, S.

A.;Enriz, R. D.; Ribas, J. C.; Bioorg. Med. Chem. 2000, 8, 691.

16. Majerz-Maniecka, K.; Musiol, R.; Nitek, W.; Oleksyn, B. J.; Mouscadet, J.; Le

Bret,M.; Polanski, J.; Bioorg. Med. Chem. Lett, 2006, 16, 1005.

91

17. Doyle, T. W.; Balitz, D. M.; Grulich, R. E.; Nettleton, D. E.; Gould, S. J.; Tann,

C.-H.; Meows, A. E.; Tetrahedron Lett, 1981, 22, 4595.

18. Hackethal, C. A.; Golbey, R. B.; Tan, C. T. C.; Karnofsky, D. A.; Burchenal, J.

H.; Antibiot. Chemother, 1961, 11, 178.

19. Boger, D. L.; Mitscher, Y. M.; Drake, S. D.; Kitos, P. A.; Thompson, S. C.; J.

Med.Chem., 1987, 30, 1918.

20. Wilson, W. L.; Labra, C.; Barrist, E.; Antibiot. Chemother. 1961, 11, 147.

21. Mekouar, K.; Mouscadet, J.-F.; Desmae¨le, D.; Subra, F.; Savoure´, D.; Auclair,

C.; d'Angelo, J.; J. Med. Chem. 1998, 41, 2846.

22. Mekouar, K.; Mouscadet, J.-F.; Desmae¨le, D.; J. Med. Chem. 2000, 43, 1533.

23. Lubrowski, J.; Yang, F.; Alexandratos, J.; Wlodawer, A.; Zhao, H.; Burke, Jr., T.

R.; Neamati, N.; Pommier, Y.; Merkel, G.; Skalka, A. M.; Proc. Natl. Acad. Sci.

U.S.A. 1998, 95, 4831.

24. Goldgur, Y.; Dyda, F.; Hickman, A. B.; Jenkins, T. M.; Craigie, R.; Davies, D.

R.; Proc. Natl. Acad. Sci. U.S.A. 1998, 95, 9150.

25. Lubrowski, J.; Yang, F.; Alexandratos, J.; Wlodawer, A.; Zhao,H.; Burke, Jr., T.

R.; Neamati, N.; Pommier, Y.; Merkel, G.; Skalka, A. M.; Proc. Natl. Acad. Sci.

U.S.A. 1998, 95, 4831.

26. Goldgur, Y.; Craigie, R.; Cohen, G. H.; Fujiwara, T.; Yoshinaga, T.; Fujishita, T.;

Sugimoto, H.; Endo, T.; Murai, H.; Davies, D. R.; Proc.Natl. Acad. Sci. U.S.A.

1999, 96, 13040.

27. Ouali, M.; Laboulais, C.; Leh, H.; Gill, D.; Desmae¨le, D.; Mekouar, K.; Zouhiri,

F.; d'Angelo, J.; Auclair, C.; Mouscadet, J.-F.; Le Bret, M. J. Med. Chem. 2000,

43, 1789.

28. Hare, S.; Gupta, S. S.; Valkov, E.; Engelman, A.; Cherepanov, P.; Nature, 2010,

464, 232.

29. Maertens, G. N.; Hare, S.; Cherepanov, P. Nature 2010, 468, 326.

30. Neamati, N.; Sunder, S.; Pommier, Y. Drug Discov. Today 1997, 2, 487.

92

31. Artico, M.; Di Santo, R.; Costi, R.; Novellino, E.; Greco, G.; Massa, S.;

Tramontano, E.; Marongiu, M. E.; De Montis, A.; La Colla,P. J. Med. Chem.

1998, 41, 3948.

32. Weinberg, B.; Shugars, D. C.; Sherman, P. A.; Sauls, D. L.; Fyfe, J. A.; Biochem.

Biophys. Res.Commun. 1998, 246, 393.

33. Jing, N.; Hogan, M. E. J. Biol. Chem. 1998, 273, 34992.

34. Hong, H.; Neamati, N.; Wang, S.; Nicklaus, M. C.; Mazumder, A.; Zhao, H.;

Burke, Jr., T. R.; Pommier, Y.; Milne, G. W. A.; J. Med. Chem. 1997, 40, 930.

35. Journal of Medicinal Chemistry, 2007, Vol. 50, No. 1

36. Weinberg, B.; Shugars, D. C.; Sherman, P. A.; J. Med. Chem. 2004, 47, 2075.

37. Artico, M.; Di Santo, R.; Costi, R.; Novellino, E.; Greco, G.; Journal of

Medicinal Chemistry, 1988, Vol. 31, No. 7)

38. K. Majerz-Maniecka et al. / Bioorg. Med. Chem. 2011, 19, 1606–1612

Section-B

Greener Methodology for Synthesis of Urea Derivatives

93

Introduction

A simple and efficient route for the synthesis of unsymmetrical N,N`-Diphenyl

urea have been developed in aqueous medium under base and catalyst free condition

from corresponding substituted isocynate and amines. The remarkable key feature of the

reaction include the use of water as an inexpensive and environmentally benign reaction

medium, absence of base and any additional catalyst, and easy isolation of the product.

We report a simple, economical, efficient, high yielding, one-pot synthesis of

unsymmetrical disubstituted ureas, from the corresponding isocynates under aqueous

medium. The remarkable key feature of the reaction include the use of water as an

inexpensive and environmentally benign reaction medium ,absence of base and any

additional catalyst, and easy isolation of the product. We believed that this synthesis

protocol offer a more general method for the formation of C–N, C-O & C-S bonds

essential to numerous organic syntheses. Various unsymmetrical disubstituted urea’s,

thiourea derivatives were prepared in high yields and their spectroscopic confirmation

was achieved.

Recent focus on urea’s stems from their wide range of application in

petrochemicals, agrochemicals, and pharmaceuticals' used as dyes for cellulose fibres,

antioxidants in gasoline or as plant growth regulators, pesticides and herbicides. The

unsymmetrical urea functional group is also encountered in several biologically active

synthetic targets. In particular, potent urea containing HIV-1 protease inhibitors2 and p38

kinase inhibitors3 have recently been disclosed. Substituted urea’s are very important

class of compounds that display a wide range of interesting applications6. They have

extensively been used as agrochemicals7, pharmaceuticals

8, intermediates in organic

synthesis9, for protection of amino groups

10, and as linkers in combinatorial chemistry

11.

These require their preparation by convenient and safe methodology. Traditionally, their

synthesis involves a reaction of amines with phosgene12

, its derivatives13

, carbonyl-

imidazoles14

, or carbon monoxide15

using various kinds of metal and non-metal catalysts.

Despite the growing number of synthetic methodologies, ureas are most

commonly synthesized by reaction of an amine with phosgene or carbamates. This

approach is particularly efficient for symmetrical urea’s. In the case of unsymmetrical

94

urea’s, the synthetic efficiency is limited by the formation of symmetrical urea side

products. In the last few years, toxic and unstable reagents such as phosgene and isolated

isocyanides have been increasingly substituted for cleaner and inherently safer

alternatives4.

These include use of carbonates or carbonyl imidazole or taking advantage of the

reactivity of carbonates’ with amines to produce urea. Unfortunately, production and use

of phosgene opens many worrying toxicological and environmental problems.

Nevertheless, about 2 million tons per year of phosgene are produced and utilised

worldwide.5 Under the new environmental legalization of the developed countries,

industrial and academic research groups have performed methodologies for preparation

of urea based on the use of reagents which are less toxic and less hazardous than

phosgene5.

Method for preparing 1, 3-disubstituted urea through catalytic process by reacting

a cyclic carbonic acid ester with an amine was disclosed16

. This method is too expensive

to use on a large scale. The transformation of amines to disubstituted urea through

catalytic carbonylation provides an alternative environmental benign method and has

been investigated over many years using various kinds of metallic catalysts17

. However,

these methods failed due to the problems of regenerating the catalysts from the products.

Moreover, their formation using CO2 employed harsh reaction conditions, such as long

reaction times, use of expensive strongly basic reagents, tedious work-up, and low

yields18

. Consequently, there is continued interest in developing new and convenient

methods for the synthesis of substituted urea using mild reaction conditions.

95

Results and Discussion

Most of the synthetic approaches to produce urea utilize phosgene or its tamed

analogs. Commercially available reagents such as Benzyl Isocynate & Phenyl Isocynate

also effectively convert into corresponding disubstituted urea under dry reaction

condition. Reaction of Phenyl Isocynate in 1, 4-dioxane/water or pyridine/water unable to

gives desired 1, 3-diphenylurea in required yield. 19

Reaction was very slow and takes

about 12 to 16 h to complete reaction. Reagents like Isocynate might not withstand in

aqueous condition or react efficiently with complex starting materials. According to our

knowledge there was no method for synthesis of urea in water from isocynate and amine.

We have therefore soughed to develop a methodology which should be Simple, scalable

& ecofriendly method to produced disubstituted urea, Use of reagent that electrophonic

enough and effectively react with amines of various structures, yet reasonably stable in

aqueous environments.

Reaction of 1-fluoro-4-isocyanatobenzene with anisole in aq. Medium gives

desired urea in very low yield (10%) and it takes 10-15 hr to complete the reaction. This

could be due to instability of isocynate in water. When we repeated same reaction in

water at 4-50C, excellent yield of desired urea was observed in 30min.From above

observation we conclude that reagent like isocynate may be stable in aq. Medium at

lower temperature. Although this was our observation in this particular case, however we

do not have any proof for the stability of such reagent in water at lower temperature. To

test the feasibility and practical applicability, a reaction on large scale was conducted in

water at 4-50C; 85% of conversion was observed after 30 min. The mixture was just filter

out to get pure biphenyl urea.

Anisole dissolved in water (complete or partial) and the mixture was cooled to

50C. After 5 min 1mol. eq. of 1-fluoro-4-isocyanatobenzene was slowly added in above

reaction mixture in such way that temperature of reaction mixture doesn’t rise above

100C. As reaction proceeds solid falls out. RM was stirred for 30 min at 5

0C & reaction

monitor by TLC. Solid was filter out & residue washed with water. Obtained product

does not required further purification. Solid was collected to report yield and analysis.

96

NCO

F

O

H2N

H2O, 4-5 0C

30 min

HN

HN

F OO

Scheme3.2

Under optimized reaction condition, the scope of reaction was explored with

structurally and electronically diverse amine, thiols, alcohol and isocynate to get

respective urea, thiourea & carbamate. Mosubstituted aryl amine such as n methyl phenyl

amine treated with 1-fluoro-4-isocyanatobenzene under similar condition gives 80%

yield. Also good yield was obtained in case of amine which was partially soluble in aq.

Medium at lower temperature. Reaction of p-nitro aniline with 1-fluoro-4-

isocyanatobenzene failed to give desired product under similar reaction condition. This

may be decrease in Nucleophilicity of amine due to p-nitro group.

When Thiol treated with 1-fluoro-4-isocyanatobenzene gives corresponding

thiourea. Similarly when phenol treated with 1-fluoro-4-isocyanatobenzene gives

corresponding carbamate. We found slight lower yields of Thiourea & Carbamate as

compare to urea. We also observed low yields when n methyl phenyl amine was treated

with 1-fluoro-4-isocyanatobenzene. From above observation we conclude that low yield

was due to decrease in nucleophilicity of Phenol, thiols & mosubstituted aryl amine in aq.

Medium.

97

Table 2.B.1 Synthesis of N-N` Biphenyl urea

Sr.No. Product Time (h) Yielda (%) M.P. (°C)

3.B.1

0.5 85 233-237

3.B.2

1 80 120-125

3.B.3

0.5 91 145-150

3.B.4

2 82 135-140

3.B.5

3 73 ---

3.B.6

2 77 155-157

3.B.7

2 80 157-160

3.B.8

2 75 125-127

3.B.9

1.5 80 77-80

3.B.10

0.5 87 132-134

3.B.11

1 75 140-142

3.B.12

1.5 80 130-132

3.B.13

0.5 94 196-198

3.B.14

3 72 120

aIsolated yield

98

Experimental

General procedure: Anisole (Amine) (10mmol) was dissolved in water and the mixture

cooled to 50C. After 5 min 1-fluoro-4-isocyanatobenzene (10mmol) was slowly added in

above reaction mixture in such way that temperature of reaction mixture doesn’t increase

above 50C. As reaction proceeds solid falls out. RM was stirred for 30 min at 5

0C &

reaction was monitor by TLC. After completion of reaction Solid was filter out & residue

washed with water. Solid was collected to report yield & analysis of respective urea. The

product was confirmed by Melting points and spectral analysis such as MS, NMR (Table

1, entry 1)

1H NMR (300MHz ,DMSO-d6) δppm; 8.60 (s, 1 H),8.45 (s, 1 H), 7.45 (dd, J = 9.1, 4.9

Hz, 2H), 7.35 (d, J = 9.1 Hz, 2 H), δ7.10 (t, J = 8.9 Hz, 2 H),δ6.87 (d, J = 8.7Hz, 2 H),

δ3.71 (s, 3 H); MS (ESI+) Calcd for C14H13FN2O2 : 260.26, Found: 261.11 [M+1]. White

solid.

99

General Methods

All Reagents were commercially purchased from Aldrich and used without further

purification. Commercial reagents were used as received. Reaction were monitored by

thin-layer chromatography (TLC) on 0.25mm precoated Merck Silica Gel 60 F254,

visualizing with ultraviolet light or Ninhydrine. 1H NMR spectra were recorded on a

Bruker DPX-400 with standard pulse sequences, operating at 300 MHz. Chemical shifts

were reported in parts per million (ppm) downfield from tetramethylsilane (TMS), which

was used as internal standard. HPLC-MS analyses were performed with an Agilent

Technologies 1100 series consisted of a quaternary pump with degasser, auto sampler,

column oven and DAD detector.

1-(4- fluorophenyl)-3-(4- methoxyphenyl) urea (3.B.1)

HN

HN

OF O

1H NMR (300MHz ,DMSO-d6) δppm: 8.60 (s, 1 H),8.45 (s, 1H), 7.45 (dd, J = 9.1, 4.9

Hz, 2 H), 7.35 (d, J = 9.1 Hz, 2H), 7.10 (t, J = 8.9 Hz, 2H), 6.87 (d, J = 8.7Hz, 2 H), 3.91

(s, 3 H); MS(ESI+) Calcd for C14H13FN2O2 : 260.26, Found: 261.11 [M+1].

3-(4- fluorophenyl)-1methyl-1-phenyl urea (3.B.2)

HN N

OF

1H NMR ( 300MHz, DMSO-d6) δppm; 8.21 (s, 1 H), 7.38 - 7.46 (m, 4H), 7.30 - 7.35 (m,

2H), 7.21 - δ7.28 (m, 1H), 7.02 - 7.10 (m, 2H), 3.26 (s,3H); MS(ESI+) Calcd for

C14H13FN2O :224.12, Found:225.21 [M+1].

N-(4 – fluorophenyl)-3, 4-dihydroisoquinoline-2(1H)-carboxamide (3.B.3)

100

HN N

OF

1H NMR (300MHz, DMSO-d6)δppm; 8.61 (s, 1 H), 7.45 - δ7.55 (m, 2H), 7.19 (s, 4H),

7.08 (t, 2H), 4.63 (s, 2H), 3.69 (t, J = 5.9 Hz, 2H), 2.85 (t, J = 6.0Hz, 2H); MS (ESI+)

Calcd for C16H15FN2O :270.12 ,Found: 271.11 [M+1].

1-(4-fluorophenyl)-3-(2-methoxyethyl) urea (3.B.4)

HN

HN

OF

O

1H NMR (300MHz ,DMSO-d6)δppm; 8.54 (s, 1H), 7.37 (dd, J = 9.3, 5.1 Hz, 2H), 7.05

(t, J = 9.1 Hz, 2H), 6.16 (t, J = 5.5 Hz, 1H), 3.32 - 3.40 (m, 5H), 3.21 -δ 3.25 (m, 2H);

MS (ESI+) Calcd For C10H13FN2O2: 212.22; Found:213.23 [M+1].

S-phenyl N-4- fluorophenylcarbamothioate (3.B.5)

HN S

OF

1H NMR (300MHz ,DMSO-d6) δppm; 10.57 ( s,1H),7.48 - 7.56 (m, 4H), 7.44 - 7.48 (m,

3H), 7.12 - 7.19 (m, 2H); MS (ESI+) Calcd for C13H10FNOS : 247.29 ,Found:248.29

[M+1].

4-fluorophenyl 4- fluorophenyl carbamate (3.B.6)

OHN

OFF

1H NMR (300MHz ,DMSO-d6) δppm; 10.29 (br. s, 1H), 7.51 (dd, J = 9.1, 4.9 Hz, 2H),

7.24 - 7.29\(m, 4H),7.18 (t, J = 8.9 Hz, 2H); MS (ESI+) Calcd for C13H9F2NO2 :

249.21,Found: 250.01 [M+1].

4-methoxyphenyl 4- fluorophenyl carbamate (3.B.7)

101

OHN

OOF

1H NMR (300MHz ,DMSO-d6) δppm; 10.20 (br. s., 1H), 7.51 (dd, J = 9.1, 4.9 Hz,

2H),7.22 – 7.09 (m, 3H), 6.95 (d, J = 9.1Hz, 2H), 3.76 (s, 3H); MS (ESI+) Calcd for

C14H12FNO3 : 261.25; Found:262.15[M+1].

Phenyl 4- fluorophenyl carbamate (3.B.8)

OHN

OF

1H NMR (300MHz ,DMSO-d6) δppm; 10.27 (br. s, 1H), 7.52 (dd, J = 9.1, 4.9Hz, 2H),

7.39 - 7.47(m, 2H), 7.13 - 7.30(m, 5H); MS (ESI+) Calcd for C13H10FNO2 : 231.22;

Found:232.54[M+1].

102

Representative Spectra

1HNMR of Compound 3.B.1

MS of Compound 3.B.1

103

HPLC of Compound 3.B.1

104

References

1. Sartori, G. Green Chemistry, 2000,2, 140.

2. L. Cassar, Chem. Ind. (Milan), 1990, 72, 18.

3. Lam, P. Y. S.; Jadhav, P. K.; Eyermann, C. J.; Hodge, C. N.; Ru, Y.; Bacheler, L. T.;

Meek, J. L.; Otto, M. J.; Rayner, M. M.; Wong, Y. N.; Chang, C. N.; Weber, P. C.;

Jackson, D. A.; Sharp, T. R.; Erickson- Viitanen, S. Science1994, 263, 380;

4. Chauhan, Y. S; Apphun, A.; Singh, V. K.; Dwivedi, B. S. Field Crops Res, 2004, 89,

17.

5. Frezza, M.; Castang, S.; Estephanem, J.; Soule`re, L.; Deshayes, C.; Chantegrel, B.;

Nasser, W.; Queneau, Y.; Reverchon, S.; Doutheau, A. Bioorg Med Chem, 2006, 14,

4781.

6. Li, G.; Hasvold, L. A.; Tao, Z. F.; Wang, G. T.; Waltney, S. L.; Patel, J.; Kovar, P.;

Credo, R. B.; Chen, Z.; Zhang, H. Bioorg Med Chem Lett, 2006, 16, 2293.

7. Hegarty, A. F.; Drennan, L. J. In: Katrizky AR, Methocohn, O, Rees CW (eds)

Comprehensive Organic Functional Group Transformations. Pergamon, Oxford,

1995, 6, 499.

8. Greene, T. W.; Wuts, P. G. M. Protective Groups in Organic Synthesis, 3rd edn. John

Wiley and Sons T. P. 1999. Vishnyakova, 1. A. Golubeva and E. V. Glebova, Russ.

Chem. Rev. (Engl. Transl.), 1985, 54, 249.

9. Han, Q.; Chang, C.-H.; Li, R.; Ru, Y.; Jadhav, P. K.; Lam, P. Y. S. J. Med.

Chem.1998, 41, 2019.

10. Regan, J.; Breitfelder, S.; Cirillo, P.; Gilmore, T.; Graham, A. G.; Hickey, E.; Klaus,

B.; Madwed, J.; Moriak, M.; Moss, N.; Torcellini, C. J. Med. Chem.,2002, 45, 2994.

11. Mensal, M.; Gutschow, M. Tetrahedron Lett 2005, 46, 2231.

12. Smith, M. B.; March, J. Advanced Organic Chemistry. Reactions, Mechanisms, and

Structure, 5th edn. Wiley Interscience, New York, Chichester, Brisbane, Toronto,

Singapore. 2001.

13. Guichard, G.; Semety, V.; Didierjean, C.; Asubry, A.; Briand, J. P.; Rodriguez, M., J

Org Chem.1999, 64, 8702.

105

14. Batey, R. A.; Santhakumar, V.; Yoshina-Ishii, C.; Taylor, S. D. Tetrahedron Lett,

1998, 39, 6267.

15. McCusker, J. E.; Main, A. D.; Johnson, K. S.; Grasso, C. A.; McElwee-White, L. J

Org Chem. 2000,65, 5216.

16. US Patent. 5902899, May 11, 1999

17. Mulla, S. A. R.; Rode, C. V.; Kelkar, A. A.; Gupte, S. P. J Mol Catal A Chem. 1997,

122, 103.

18. Tai, C. C.; Huck, M. J.; McKoon, E. P.; Woo, T.; Jessop, P. G. J Org Chem 2002, 67,

9070.

19. Liu, Q.; Luedtke, N. W.; Yitzhak, T. Tetrahedron Lett. 2001, 42, 1445.

Section-C

Green Methodology for Synthesis of Carbonyl

Compounds from Alkyl

106

3.C.1 Introduction

P-amino benzyl derivatives were found to react with 2, 3-dichloro-5,6-dicyno-1,4-

benzoquinone(DDQ) in 5% aq. HCl to give p-amino benzyl derivatives in good yields.

Thus, p- amino benzaldehyde was easily isolated from the reaction with p-toludine at

ambient temperature. Similarly hindered p-amino benzyl derivatives were

regiospecificaly oxidized to corresponding carbonyl compound in high yield in Aq.

Medium.

The discovery of new environmentally friendly methods for selective catalytic

oxidation of alkyl substrates to aldehyde and ketones in presence of amine is an important

goal in the development of modern methods for chemical synthesis.1

With the

considerable development of selective organic chemistry in the last few decades, specific

reagents have seen their potential increase dramatically. The spectrum of their

applications in modern organic chemistry has been enlarged to offer new challenging

synthesis opportunities.2 Not an exception, DDQ has been investigated as a powerful

oxidizing agent by several research groups, and was proved to be useful for a wide range

of reactions. Although several uses of DDQ have been mentioned in many patents from

the pharmaceutical and chemical specialties industries, we will restrict this article to

regiospecific oxidation by DDQ of an unhindered alkyl groups in sterically hindered

aromatic amine.

Amino benzaldehydes are the important key intermediate for synthesis of many

biologically active compounds. They are use for synthesis of inosine monophosphate

dehydrogenase (IMPDH) inhibitors 3ab

in development of anticancer drugs,4Chk1 kinase

inhibitors5 and also in the preparation of an intermediate of one of the major metabolites

of the antihypertensive Trequisin,6 Novel thyroid hormone receptor antagonists.

7

The oxidation of the sterically hindered aromatic amine has been reported using

enzymatic, 8

electrochemical9 and chemical reactions

10, 11a, b

. In the reaction the nitrogen

of the molecule is envold in the oxidation and not the alkyl group.

107

2, 3- Dichloro- 5,6- dicyno-1,4 benzoquinone (DDQ) and related high potential

oxidant interact with aliphatic amino groups and doubts have been raised about the nature

of reaction with aromatic amines. One of the earlier reports13

was successful to do

regiospecific oxidation of mesidine to 4-amino-3, 5-dimethylaldehyde by using DDQ.

They have isolated above product in solid crystalline from by using 1, 4-dioxane as

solvent. This method has certain drawbacks such as 1) as low yield, long reaction time

and tedious workup, 2)failed to give p-amino benzaldehyde from p- toludine.3) 2,6-

dimethyl-4-propylaniline to corresponding carbonyl compound gives very less yield. An

aerobic oxidations by using molybdovanadophosphate as a catalyst Convert 2,4,6-

trimethylaniline to 4-amino-2,6-dimethylbenzaldehyde in good yield (64%) which is

difficult to prepare by conventional methods however this reaction was catalyzed by

metal and required high temperature with long reaction time.12

This method is also not

suitable for oxidation of p- toludine. To overcome these drawbacks we have decided to

develop methodology which is high yielding, short reaction time, and use of greener

protocol.

There are no previous reports, to the best of our Knowledge, of the use of water as

a solvent and method for the regiospecific oxidation of p-toludine to p-amino

benzaldehyde.

108

3.C.2 Results and Discussion

To continue our interest in conversion of aromatic methyl group into

corresponding carbonyl compound we have selected mesidine as model substrate for

further studies. As mention earlier studies when mesidine treated with 3eq DDQ in 1, 4 -

Dioxane in 6 hr give corresponding aldehyde in 49 % yield.13

When above reaction was

done with 1 eq. and 2 eq. gives only 12% and 20% yield of product respectively. To

improve yield of reaction we have decided to use different solvent keeping rest of thing

constant. Similar reaction was repeated in solvent such as THF, Acetonitrile and DMF

gives almost similar yield. Drastic change in yield was observed when reaction was done

in water, this method gives 70% desired product of aldehyde from mesidine. Although

the amines are partially soluble in water the advantages of the employment of water as a

solvent are numerous: not only is it cheap, nontoxic, and inflammable, but it also has the

feature that the stability of intermediate (A, B, C) in water which leads to high yield of

desired product.

NH2

DDQ

NH2

CHO

DDQH2

23 24

a

Scheme-3.3

When mesidine treated with DDQ in 5% aq. HCl the 4- methyl group was

regiospecificaly oxidized to corresponding aldehyde with 94% yield. It has isolated in

pure state and can be store for longer time. For consistency of result we have repeated

same protocol 3 times for mesidine and we have got similar result as we done previously.

Mesidine (1gm) was taken in 5% aq. HCl (20ml) to which DDQ (3eq) was added in it

and mixture was allow to stir for 4 hr. reaction was monitor by TLC. After complete

disappearance of starting material from TLC, reaction mixture was basified with 10% aq.

NaOH and compound was extracted in Ethyl Acetate. Ethyl Acetate layer was dried over

anhydrous sodium sulphate and concentrated to get white crystalline solid. Under

109

optimized reaction condition, the scope of reaction was explored with structurally and

electronically diverse amine.

Steric hindrance certainly plays role in the success of this reaction. Based on

earlier observation amino group is not a favorable substituent in DDQ oxidation13,15

still

gives good yield, as long as it is crowded by other groups. Less hindered anilines are not

suitable for the DDQ oxidation such as P-Toludine failed to give P-amino

benzaldehyde13

and 2, 4 dimethyl aniline gave only 10% of respective aldehyde.13

Reaction of p-Toludine with DDQ in H2O gives only 35% yield of p- amino

benzaldehyde. Whereas 58% yield was achieved when similar reaction was carried out in

5% Aq HCl with DDQ. We believed that this is novel method for oxidation of P-

toludine to P-aminobenzaldehyde in water at ambient temperature.

A considerable increase in the yield was observed when the substituent in ortho

position to the amine was more bulky. More the steric bulk at ortho position of amine

will give more yield of respective aldehyde. Similarly, 2- chloro-4-methylaniline gives 4-

amino-3-chlorobenzaldehyde in 80%.and 2 bromo-4methyl Aniline were successfully

oxidized to 4-amino-3-bromobenzaldehyde the corresponding aldehyde with very good

yield. While oxidation of 2-methyl-4- methyl aniline to 4-amino-3-methyl benzaldehyde

gives 72% yield. Reactions of 4- benzyl-2-methylbenzenamine (Table3.C.1 entry 9) with

DDQ under optimize condition gives 73% of (4-amino-3-methylphenyl1) (phenyl)

methanone.

DDQ oxidation of 2, 6-dimethyl1-4propaniline in to respective ketone gives only 10 %

yield. The poor yield might be due to a further dehydrogenation of the – CO-CH2CH3

group there by generating very active species – CO-CH=CH2, although this is only our

speculation, since we do not have evidence on this matter.

110

Table- 3.C.1

Sr. No.

Final

product

Starting

Material

Final Producta Physical constant

(M.P) 0C

Yield (%)b

3.C.1

101-102

94

3.C.2

98-100

82

3.C.3

109-110

80

3.C.4

104-106

75

3.C.5

161-163

80

Me

Me Me

NH2

C

Me Me

NH2

HO

Me

Me

NH2

C

Me

NH2

HO

Me

Br

NH2

C

Br

NH2

HO

Me

Cl

NH2

C

Cl

NH2

HO

C

Br

NH2

HO

Br

Me

Br

NH2

Br

111

3.C.6

42-44

58

3.C.7

169-170

80

3.C.8

81-82

10

3.C.9

121-122

73

3.C.10

169-170

95

aAll the products are characterised by 1H-NMR and purity checked by HPLC and the melting point and

spectroscopic data are in good agreement with those in reported method 13; bIsolated Yield.

Me

NH2

C

NH2

HO

NH2

O

Me

NH2

Me

NH2

MeNH2

Me

O

NH2

Me

Br

Br

Me

NH2

OH

Br

Br

Me

NH2

Br

Br Me

HO

NH2

Me

Br

Br Me

112

Abstraction of a hydride ion at the activated para-benzylic carbon as a possible

first Step could ultimately result in the formation either of an iminoquinone Methide

intermediate (A).12,16

Then 1, 6 addition of Aq. HCl to A gives compound (B) And final

Aminobenzaldehyde may be produced By the hydrolysis of dihydroxy intermediate (C).

C was derived from (B) and Aq. HCl via similar reaction path as the formation of (B).

Above mechanism was proposed on the basis of oxidation of phenolic compound. Use of

more polar protic solvent such as MeOH, Water stabilize intermediate A and B which

will automatically result in high yield of respective amino benzaldehyde.15

Similarly we

are getting high yield of aminobenzaldehyde due to stability of A and B in aq. HCl.

Me Me

CH2

NH

OHH

NH2

Me

OHHO

NH2

Me MeMe

HO

NH2

Me Me

10%Aa NaOH1

(A) (B) (C)

In section we report a simple, economical, efficient, high yielding, method for

regiospecific oxidation of aromatic alkyl group in to corrourpounding carbonyl

compound in presence of amine depending on nature of substrate. Various Amino

Benzaldehyde (Table 2.C.1) were prepared in high yields under optimize reaction

protocol and their spectroscopic confirmation was achieved.

113

3.C.3 Experimental

General procedure

Mesidine (15.0 g; 0.111 moles) was dissolved in 5% Aq. HCl (300 ml) and DDQ

(15.0 g; 0.225 moles) was added. A green colour complex was generated. This changed

its colour to dark brown, within five minutes of stirring. Stirring was further continued

for 4 hrs and the residue was treated with dilute sodium hydroxide and extracted with

ethyl acetate and then worked up in the usual manner. Final purification by column

chromatography on silica gel, Using hexane as eluent, gave, the main fraction 2,6-

dimethyl-4-formylaniline.

114

Spectral data

4- amino-3, 5-dimethylbenzaldehyde

NH2

Me Me

OH

Crystalline solid ; M.p.78 - 800C;

1H NMR (CDCl3, 300 MHz) ppm: 9.71 (s, 1H), 7.47

(s, 2H), 2.23 (s, 6H), 4.22 (br.s, 1H)

p- aminobenzaldehyde.

NH2

OH

p-Aminobenzaldehyde) Crystalline solid; Mp 41-43 o C;

1H NMR (CDCl3, 400 MHz)

ppm; 9.74 (s, 1H), 7.68 (d, J = 8.5Hz, 2H), 6.77 (d, J = 8.5 Hz, 2H), 4.37 (br.s, 2H) 4-

amino-3-bromobenzaldehyde

NH2

OH

Br

Solid; Mp 109-110 0C;

1H NMR (CDCl3, 300 MHz) ppm; 9.71 (s, 1H), 7.95 (d, J = 8.2

Hz, 2H), 7.64 (d, 1H, J = 8.2 Hz), 6.80 (s, 1H), 4.73 (br.s, 2H, NH2)

115

4-amino-3-Chlorobenzaldehyde

NH2

OH

Cl

Solid; Mp 104-106 0C;

1H NMR (CDCl3, 300 MHz) ppm; 9.71 (s, 1H), 7.95 (d, J = 8.2

Hz, 2H), 7.64 (d, 1H, J = 8.2 Hz) 6.80 (s, 1H), 4.73 (br.s, 2H) and 9.71 (s, 1H)

4-amino-2, 3-dibromo-6- methyl benzaldehyde

NH2

OH

Br

Br Me

Mp 169-170 0C;

1H NMR (CDCl3, 300 MHz) ppm; 10.33 (s, 1H) 6.54 (s, 1H), 4.76 (br.

s, 2H), 2.51 (s, 3H).

4-amino-2, 3-dibromo-5- methyl benzaldehyde

NH2

OH

Br

Br

Me

Mp 169-170 0C;

1H NMR (CDCl3, 300 MHz) ppm; 10.33 (s, 1H) 6.54 (s, 1H), 4.76 (br.

s, 2H), 2.51 (s, 3H)

1-(4amino-3, 5-dimethylphenyl) propan-1-one

116

NH2

Me Me

O

Mp 81-82 oC;

1H NMR (CDCl3, 400 MHz) ppm; 7.47 (s, 2H), 2.23 (s, 6H), 2.122 (q,

2H), 1.21 (t, J = 6.9, 3H).

4-amino-3-methylphenyl1) (phenyl) methanone

NH2

Me

O

Mp 121- 122 oC;

1H NMR (CDCl3, 300 MHz) ppm; 7.47 - 7.71 (m, 4H), 7.57 (m, 1H),

6.86 (d, J = 7.6,1H), 7.51 (d, J = 7.6,1H), 7.41 (s,1H, Ar H)2.21 (s, 3H), 4.22 (br.s, 2H)

4- amino-3, methylbenzaldehyde.

NH2

Me

HO

Mp 98-100 0C; IR (neat) 1677 cm

-1;

1H NMR (CDCl3, 400 MHz) ppm; 9.87 (s, 1H),

6.72 (s, 1H), 7.57 (d, J = 7.6,1H), 7.67 (d, J = 7.6,1H), 2.23(s, 3H), 4.22 (br. s, 2H), and

9.87 (s, 1H).

117

3.C.4 Representative Spectra

1HNMR of Compound 3.C.1

HPLC of Compound 3.C.1

118

References

1. Chiellini, G.; Nguyen, N.-H.; Yoshihara, H. A.; Scanlan, T. S.; Bioorg. Med.

Chem. Lett. 2000, 10, 2607

2. Elke Fritz-Langhals; Johannes Freudenreich; US Patent no. US006023000A.

3. Iwanowicz, E.J.; Watterson, S.H.; Guo, J.; Pitts, W.J.; Muralidhar, T.G.; Shen,

Z.; Chen, P.; Gu, H.H.; Fleener, C.A.; Rouleau, K.A.; Cheney, D.L.; Townsend,

R.M.; Hollenbaugh, D.L;. Bioorganic & Medicinal Chemistry Letters 2003,13,

2059.

4. Rangarajan, M.; Kim, J.S.; Sim, S.P.; Liu, A.; Liu, L.F.; LaVoie, E.J.; Bioorganic

& Medicinal Chemistry 2000,8, 2591.

5. Gaoquan, Li,Lisa.; Hasvold, A.; Bioorganic & Medicinal Chemistry Letters 2006,

16 ,2293.

6. Bansi Lal; Bhattacharya, B. K.; Dadkar, N. K.; Dohadwalla,A. N.; H.; Dornauer,

N. J.; Weithmann, U.; IRCS Med. Sci., 1981,2, 325.

7. Komatsu, T.; Hirano, T.; Songkram, C.; Kawachi, E.; Kagechika, H.; Bioorganic

& Medicinal Chemistry, 2007 15,3115.

8. Bartling, G. J.; and L. J. Forrester, Trans. M. O.; Acad. Sci., 1973 ,78 , 220

9. Tiedemann, W.; and Newmann, J.; Electroanal, J.; Chem. Interfacial

Electrochem., 1975, 63, 187

10. Hedayatullah, M,; Dechatre, J. P.; and Denivelle, E. L.; Tetrahedron Lett.1975,

2039; Konand, E.; and MC Nelis, E, Chem. Comm. 1973, 562.

11. (a) Goldstein, S. L.; and McNelis, E,; J.Org. Chem., 1973, 38, 183; (b) Holmer-

Siedle, A. G.; and Saunders B. C.; Chem. Ind., 1959, 164.

12. Bartling, G. J.; and L. J. Forrester, Trans. M. O.; Journal of Molecular Catalysis

A: Chemical 1996,110, 105.

13. Bansilal, Genial disuza; Tet.Lett., 1984, 29, 2901.

14. LEE-RUFF, E.; and ABLENAS F. J.; Canedian. J. Chem.1989, 67, 699.

15. Becker H. D.; "The Chemistry of Quinonoids compounds". (Ed. S. Patai), Wiley

Interscience, New York (1974). P. 398.

16. Becker H. D.; J. Org. Chem., 1980, 45, 1596.

Recommended