SYNTHESIS OF NUCLEOSIDE ANALOGUES WITH NEW TYPE OF

Ministry of Higher Education
And Scientific Research
University of Baghdad
College of Science
Department of Chemistry
SYNTHESIS OF NUCLEOSIDE ANALOGUES
WITH NEW TYPE OF NITROGEN
BASES
A thesis
Submitted to the College of Science-University of Baghdad
In Partial Fulfillment of Requirement for the Degree
of Master of Science in Organic
Chemistry
By
Asmaa Mazin Abdul Hamid Kadhim Al-Samaraey
B.Sc. (Baghdad University, 1999)
Supervised by
Prof. Dr.Yousif Ali Al- Fattahi and Prof. Suad Al- Araji
2005
i
We certify that this thesis was prepared under our supervision at the
University of Baghdad, College of Science, as a partial requirement for the
degree of Master of Science in Chemistry.
Signature:
Signature:
Advisor: Prof. Dr.Yousif Ali Al- Fattahi
Date:
Advisor: Prof. Suad Al- Araji
Date:
In view of the available recommendation, I forward this thesis for debate by
the examining committee.
Signature:
Name: Prof.Suad Al-Araji
Chairman of the Committee of
Graduated Studies in Chemistry.
Date
ii
We the examining committee, certify that we have read this thesis and
examined the student in its contents and that, in our opinion, it is adequate with
"
" standing as a thesis for the degree of Master of Science in Chemistry.
Signature:
Signature:
Name
:
Name
:
Date
:
Date
:
Member
Chairman
Signature:
Signature:
Name
:
Name
: Prof. Dr.Yousif Ali Al- Fattahi
Date
:
Date
:
Member
Supervisor
Signature:
Name
: Prof. Suad Al- Araji
Date
:
Supervisor
Approved by the University Committee on Graduate Studies.
Signature:
Name
: Prof.Dr. A. M. Taleb
Dean of the College of Science.
Date
:
iii
Acknowledgment
The work described in this thesis was performed at the department of
chemistry, college of science, university of Baghdad.
I have the honor to express my sincere thanks and gratitude to my Prof. Dr.
Yousif Ali- Al-Fattahi and Prof. Suaad M Al-Araji for suggesting the problem,
and for there valuable guidance and continuous encouragement during the
progress of this work.
Special thanks are due to the Department of Chemistry, College of Science,
University of Al-Nahrian, specially Dr. Shahbas A. Maki, Head of Department
of Chemistry for his help during my work.
My sincere thank and appreciation to Dr. Yaseen Al-Sood, Dr. Nadheer AlAnsary and Muhanad Masad, College of Scince, Al al-bayt University, Jordan,
for providing 1H-NMR and 13C-NMR spectra.
I wish to express my thanks to: Miss Hasseba, Dr. Suaad M. Hussin, Miss
Zainab, Mrs Nasreen for their help and encerigment.
Sincere thanks also to my colleagues from the research group, and all
member of the department of chemistry for their helps during the course of
study.
Asmaa M. Kadhim
iv
CONTENTS
LIST OF SCHEMES...................................................................................vii
LIST OF TABLES ………………………………………………………….vii
LIST OF FIGURES .………………………………………………………...viii
LIST OF ABBREVIATIONS…………………………………………………..x
ABSTRACT…………………………………………………………………..xii
CHAPTER ONE: INTRODUCTION
1.1. Nucleosides, nucleotides and nucleic Acids……………………………….1
1.2. Nucleoside analogues and biological activity...……………………………6
1.2.1. Based-modified nucleoside analogues…………………………...…8
1.2.2. Sugar-modified nucleoside analogues…………………………...….9
1.2.3. Nucleoside analogues as anti cancer chemotherapy………….........11
1.2.4. Nucleoside analogues as anti viral chemotherapy……………...….14
1.2.4.1. Nucleoside analogues active against DNA viruses….....…14
1.2.4.2. Nucleoside analogues active against RNA viruses….........14
1.2.4.3. Nucleoside analogues active against retroviruses…….......16
1.2.5. Nucleoside analogues as antibiotherapy…………………………..16
1.2.6. C-Nucleoside analogues and biological activity…………………..19
1.3. Nucleosides synthesis………………………………………………….…21
1.3.1. Synthesis of Nucleosides………………………………………….21
1.3.2. Synthesis of C- nucleosides……………………………………….23
CHAPTER TWO: EXPERIMENTAL
2.1. Apparatus and materials…………………………………………………..26
2.1.1. Apparatus………………………………………………………….26
2.1.2. Materials…………………………………………………………..27
2.2. Synthesis of nucleoside analogues………………………………………..27
v
2.2.1. Synthesis of Ribopyranonucleoside analogues…………………………27
1, 2, 3, 4-Tetra-O-benzoyl-β-D-ribopyranose [41]………………………27
2, 3, 4-tribenzoyl-β-D-ribopyranosyl bromide [42]……………………..28
Phenothiazine [43]…………………………………………………….…29
Phenoxazine [44]………………………………………………………...30
1, 2, 3, 4-Tetrahydro-carbazole [45]…………...………………………...30
Bis(theophylline-7-yl) mercury (II) [46]………………………………...30
Bis(phenothiazine-10-yl) mercury (II) [47]……………………………...31
Bis(phenoxazine-10-yl) mercury (II) [48]……………………………….32
Bis(indol-1-yl) mercury (II) [49]………………………………………..32
Bis(1, 2, 3, 4-tetrahydro-carbazole-9-yl) mercury [50]………………....32
7-(2/, 3/, 4/-Tri-O-benzoyl-β-D-ribopyranosyl) thiophylline [51]……….33
10-(2/, 3/, 4/-Tri-O-benzoyl-β-D-ribopyranosyl) phenothiazine [52]……34
10-(2/, 3/, 4/-Tri-O-benzoyl-β-D-ribopyranosyl) phenoxazine [53]……..35
1-(2/, 3/, 4/-Tri-O-benzoyl-β-D-ribopyranosyl) indol [54]………….…...35
9-(2/, 3/, 4/-Tri-O-benzoyl-β-D-ribopyranosyl)tetrahydrocarbazole[55]...35
7-β-D-Ribopyranosyl thiophylline [56]………………………………....36
10-β-D-Ribopyranosyl phenothiazine [57]……………………………...36
10-β-D-Ribopyranosyl phenoxazine [58]……………………………….37
1-β-D-Ribopyranosyl indol [59]………………………………………...37
9-β-D-Ribopyranosyl thiophylline [60]…………………………………37
2.2.2. Synthesis of Ribofuranonucleoside analogues………………………….38
1, 2, 3, 5-Tetra-O-benzoyl-β-D-ribofuranose [61]…...………………....38
1-O-Acetyl-2, 3, 5-tri-O-benzoyl-D-ribose [62]…..……………………39
1, 2, 3, 5-tetra-O-acetyl-β-D-ribofuranose [63]………...……………….39
Attempt
for
the
synthesis
of
7-(2/,
3 /,
4/-Tri-0-acetyl-β-D-
ribofuranosyl) thiophylline [64]…………………………………….......40
vi
CHAPTER THREE: RESULTS & DISCUSSION
Results and discussion...….……………………………………………………42
3.1 Syntheses of Ribopyranonucleoside analogues……………………………42
3.1.1. Synthesis of 2, 3, 4-tri-O-benzoyl-β-D-ribopyranosyl bromide [42].42
3.1.2. Synthesis of nitrogen bases and their mercury salts (II)……………49
3.1.3. Synthesis of benzoylated nucleoside analogues……………………57
3.1.4. Hydrolysis of the benzoate groups…………………………………75
3.2 Syntheses of Ribofuranonucleoside analogues……………………………93
3.2.1. Synthesis of 1-O-acetyl-2, 3, 5-tri-O-benzoyl-β-D-ribose [62]……93
3.2.2. Synthesis of 1, 2, 3, 5-tetra-O-acetyl-β-D-ribofuranose [63]……
99
3.2.3. Attempting for the synthesis of 7-(2/, 3/, 4/-Tri-0-acetyl-β-Dribofuranosyl) thiophylline [64]……………………………………………
99
References…………………………………………………………………..104
LIST OF SCHEMS
Scheme (1): Mechanism of nucleic acid chain elongation……………………...5
Scheme (2): Oversimplified virus replicative cycle………………………..….15
Scheme (3): Synthesis of nucleoside analogues [51], [52], [53], [54], [55], [56],
[57], [58], [59] and [60]………………………………………………………43
Scheme (4): The mechanism of the bromination reaction……………………..47
Scheme (5): Mechanism of β and α-nucleoside synthesis………..……………73
Scheme (6): Synthetic sreatigy………………………………………………...94
Scheme (7): The action of SnCl4 in activating poly acetyl ribose ……….……95
LIST OF TABLES
Table (1): Nucleoside and nucleotide analogues licensed for clinical anticancer
use……………………………………………………………………………...13
vii
Table (2): Structural-defined drugs licensed for clinical antiviral use………...17
Table (3): Naturally occurring C-nucleosides…………………………………20
LIST OF FIGURES
Figure (1): Partial chemical structures of the strands of DNA and RNA. The
sequence of nucleosides differs for each naturally occurring type of DNA or
RNA……………………………………………………………………………..3
Figure (2): The structure of ATP indicating its relationship to ADP, AMP and
adenosine. The phosphoryl groups, starting with that on AMP, are referred to as
the α, β, γ and phosphates……………………………………………………….4
Figure (3): Main structure modification susceptible to transform a natural
nucleoside (R=H or OH) into one of its analogues……………………………..7
Figure (4): Illustrative simple nucleoside analogues endowed with antibacterial
and antifungal activities………………………………………………………..18
Figure (5): IR Spectrum of compound [41]……………………………………45
Figure (6): IR Spectrum of compound [42]……………………………………48
Figure (7): IR Spectrum of compound [43]……………………………………50
Figure (8): IR Spectrum of compound [44]……………………………………52
Figure (9): IR Spectrum of compound [45]……………………………………54
Figure (10): IR Spectrum of compound [47]…………………………………..55
Figure (11): IR Spectrum of compound [48]…………………………………..56
Figure (12): IR Spectrum of compound [49]…………………………………..59
Figure (13): IR Spectrum of compound [50]…………………………………..60
Figure (14): IR Spectrum of compound [51]…………………………………..51
Figure (15): UV Spectrum of compound [51]…………………………………78
Figure (16): 1H-NMR Spectrum of compound [51]…………………………...62
Figure (17): The expansion of 1H-NMR Spectrum of compound [51]………..63
Figure (18): 13C-NMR Spectrum of compound [51]…………………………..64
Figure (19): IR Spectrum of compound [52]…………………………………..66
viii
Figure (20): IR Spectrum of compound [53]…………………………………..68
Figure (21): IR Spectrum of compound [54]………………………………….70
Figure (22): IR Spectrum of compound [55]…………………………………..74
Figure (23): IR Spectrum of compound [56]…………………………………..77
Figure (24): UV Spectrum of compound [56]…………………………………78
Figure (25): 1H-NMR Spectrum of compound [56]…………………………...79
Figure (26): The expansion of 1H-NMR Spectrum of compound [56]………..80
Figure (27): 13C-NMR Spectrum of compound [56]…………………………..81
Figure (28): IR Spectrum of compound [57]…………………………………..84
Figure (29): IR Spectrum of compound [58]………………………………….85
Figure (30): 1H-NMR Spectrum of compound [58]…………………………...86
Figure (31): IR Spectrum of compound [59]…………………………………..87
Figure (32): 1H-NMR Spectrum of compound [59]…………………………...88
Figure (33): The expansion of 1H-NMR Spectrum of compound [59]………..89
Figure (34): 13C-NMR Spectrum of compound [59]…………………………..90
Figure (35): The expansion of 13C-NMR Spectrum of compound [59]……….91
Figure (36): IR Spectrum of compound [60]…………………………………..92
Figure (37): IR Spectrum of compound [61]…………………………………..97
Figure (38): IR Spectrum of compound [62]…………………………………..98
Figure (39): IR Spectrum of compound [63]…………………………………101
Figure (40): IR Spectrum of compound [63]…………………………………102
ix
LIST OF ABBREVIATION
Ac
= Acetyl (CH3CO-)
ALP
= Alkaline phosphataes
Ar
= Aryl
Bz
= Benzoyl
CHCL3 = Chloroform
CH2Cl2 = Dichloromethan
13
o
C-NMR = Carbon thirteen Nuclear Magnetic Resonance
C
= Centigrade
dd
= Doublet of doublets
dec.
= Decomposition
d
= Doublet
DNA
= Deoxyribo Nucleic Acid
EtOH
= Ethanol
FTIR
= Fourier Transform Infra-Red
g.
1
= Gram
H-NMR = Proton Nuclear Magnetic Resonance
MeOH
= Methanol
MHz
= Megahertz
mL.
= Milliliter
m.mol.
= Millimole
mp
= Melting point
M
= Molar
m
= multiplet
Py
= Pyridine
x
Rf
= Factor of retention
R
= Alkyl
s
= Singlet
T.L.C
= Thin Layer Chromatography
t
= triplet
UV
= Ultra Violet in nm.
δ
= Chemical shifts in ppm., in (delta values)
%
= Percentage
xi
ABSTRACT
The work in this is divided into three parts. The first part is the introduction
which is concerned with the chemistry of nucleosides including their
configuration; classification; broad screening of their biological activity as
antitumor, antiviral and antibacterial agents; and their synthesis were reviewed.
The second part deals with the synthesis of new type of nucleoside
analogues of D-ribose. Started with preparing 1, 2, 3, 4-tetra-O-benzoyl-β-Dribopyranose [41] which was obtained from the reaction of anhydrous D-ribose
with benzoyl chloride in pyridine. When [41] was treated with 45% hydrogen
bromide solution in glacial acetic acid it gave 2, 3, 4-tri-O-benzoyl-β-Dribopyranosyl bromide [42]. The bromoribobenzoate [42] was then reacted with
different kinds of nitrogen bases ( theophylline, phenothiazine, phenoxazine,
indol and carbazol ) after being converted to the mercury salts [46], [47], [48],
[49] and [50] respectively. Applying Keonigs-Knorr method the nucleoside
analogues derivatives [51], [52], [53], [54] and [55] were obtained.
Debenzoylation of the benzoate groups of [51], [52], [53], [54] and [55]
using sodium methoxide as base afforded the free nucleoside analogues [56],
[57], [58], [59] and [60].
The third part deals with the discussion of FTIR, UV, H1-NMR and C13NMR spectral data, measurements. These results were discussed in this part to
give the final conclusion for the preparation of the above nucleoside analogues.
xii
CHAPTER ONE
INTRODUCTION
Chapter one
Introduction
1.1 Nucleosides, Nucleotides And Nucleic Acids
The term "nucleoside" was originally coined by Leven and Jacobs1. For
some time referred to compounds isolated from nucleic acids that contained a
carbohydrate attached through nitrogen to either a purine or pyrimidine base.
The evolution of the subject has brought to the point that considered a
nucleoside to be a compound of natural or synthetic origin that has a
carbohydrate attached to a nitrogen heterocyclic through either a carbonnitrogen bond as is commonly found, or a carbon-carbon bond, as found in Cnucleosides, a class of compounds discovered and developed relatively
recently2a.
Esterfication of their /5-hydroxyl group with phosphoric acid leads to
nucleotides which are the building blocks of the nucleic acids (DNA and RNA)
3a
(Fig. 1).
Natural nucleosides and nucleotides play a key role in many biosynthetic and
regulatory processes:
1- They form the monomeric units of nucleic acids. Indeed, nucleic acids are
synthesized directly from nucleoside triphosphates, the activated form of
nucleotides.
2- Nucleoside triphosphates, most conspicuously adenosine triphosphate
(ATP) are the "energy-rich" end products of most energy releasing pathways,
whose utilization drives most energy-requiring processes. Several activated
intermediates, such as Uridine diphosphate UDP-glucose penetrate in glycogen
synthesis, contain nucleotide components.
3- Most metabolic pathways are regulated, at least in part, by the levels of
nucleotides such as ATP, adenosine diphosphate (ADP), and adenosine
monophosphate (AMP) 4a (Fig. 2).
1
Chapter one
Introduction
4- Many hormonal signals, such as those controlling glycogen metabolism
are mediated intracellular by cyclic adenosine mono phosphate cAMP. or its
guanine analogue, cyclic guanine monophosphate (cGMP).
5- Adenine nucleotides are components of the co-enzymes nicotinamide
adenine dinucleotide (NAD+ ), nicotinamide adenine dinucleotide phosphate
NADP+, flavin mononucleotide FMN, flavin adenine dinucleotide FAD and coenzyme A4b.
As structural units of nucleic acids, nucleosides take part in the molecular
mechanisms of conservation, replication and transcription of the genetic
information.
In each cell, DNA is the support of this information which depends only on
the sequence of nucleosides in their chain. Thus DNA (in the nucleus) is
transcripted into messenger RNAs (mRNAs) which are transported into the
cytoplasm, where they are translated by ribosomes into proteins with the
involvement of the transfer RNAs (tRNAs). The mechanism of replication
(DNAàDNA) and of transcription (DNAàRNA) involves the polymerization
of nucleosides (in the form of their triphosphates precursors) (Scheme 1). Chain
elongation is affected by polymerases which catalyze the covalent binding at
the free-3-hydroxylated end of the chain with elimination of pyrophosphate.
The importance of nucleotides in cellular metabolism is indicated by the
observation that nearly all cells can synthesize them both de novo (a new) and
from salvage pathway (the degradation products of nucleic acids).
2
Chapter one
Introduction
NH2
N
N
N
O
P
O
N
O
O-
Adenine(A)
NH2
H
H
O
H
H
Cytosine(C)
N
H
O
P
O
DNA
O
N
O-
O
O
NH2
H
H
O
H
N
N
N
O
P
O
N
O
NH2
P
O
O
H
H
O
H
O
H
Cytosine(C)
O
O
O
O
H
O-
O
H
NH
OH
N
O
P
O
N
O
O
O
H
Thymine(T)
H
H
O
H
Guanine(G)
NH2
N
O
O-
NH
H
H
O
P
O-
H
N
3',5'phosphodiester
linkage
H3C
H
Deoxyribose
OH
N
Guanine(G)
NH2
N
O
ON
H
P
NH
H
H
H
O
H
3',5'phosphodiester
linkage
N
O
H
O-
N
Adenine(A)
O
H
H
O
OH
H
NH
H
Ribose
N
O
RNA
P
O
O
O
H
O-
Uracil(U)
H
H
H
O
OH
Figure 1: Part of chemical structures of the strands of DNA and RNA. The sequence
Of nucleosides differs for each naturally occurring type of DNA or RNA.
3
Chapter one
Introduction
phosphoester
NH2
bonds
N
N
phosphoanhydride
bonds
N
N
O-
O
P
O
O-
OO
P
O
O
P
O
O
O
H
H
OH
OH
H
H
Adenosine
AMP
ADP
ATP
Figure 2: The structure of ATP indicating its relationship to ADP, AMP, and adenosine
the phosphoryl groups, starting with that on AMP, are referred to as theα, β, γ and
phosphates.
4
Chapter one
Introduction
O
O
5end
O
Bn
O
O
+
OH
-
O
P
P
O-
O-
O
O
P
O
B
O
O-
R
OH
R
Polymerase
5end
O
Bn
O
O
O
O
O
P
+
R
O
O
-
P
O
O-
-
O
Bn+1
O
OH
P
O-
pyrophosphate
R
Scheme (1): Mechanism of nucleic acid chain elongation.
(R=OHàRNA; R=HàDNA)
5
O-
Chapter one
Introduction
1.2 Nucleoside Analogues and Biological Activity
Nucleosides, both of natural and synthetic origin have at least some
biological activity. A much smaller, but nevertheless significant number of
nucleosides are either in use as or have the potential based upon extensive
biological evaluation to be employed as chemotherapeutic agents2b. Such as
potential anti-viral5, fungicidal, and anti cancer agents6, 7. More recently, they
have been incorporated into oligonucleotides for application in the "antisense"
field, where oligonucleotides complementary to mRNA are sought as inhibitors
of gene expretion8.
So the major purpose for the syntheses of nucleosides is, of course, the
development of new compounds of chemotherapeutic interest.
Chemical modifications of naturally occurring nucleosides have been of
interest for over 50 years and numerous nucleoside analogues were synthesized
in order to selectively interfere with DNA and RNA. These structural
modifications involve either the heterocyclic ring or the sugar moiety (Fig.3)
resulting in analogues that act as anti-metabolites through the induction of one
of the following effects9:
(i) Inhibition of certain enzymes which are important for nucleic acids
biosyntheses.
(ii) Incorporation of analogues metabolites into nucleic acids which later
block their biosyntheses.
In many cases the nucleoside is not the actual active agent, but rather the
nucleoside is metabolized in cells to a mono-, di-, or triphosphate derivatives
before it is able to manifest its effect. These phosphate derivatives may be
active themselves, or they may be further metabolized. The inability of
phosphate derivatives of nucleosides to penetrate cells in significant amounts
has routinely led to the use of the nucleosides themselves as agents, thus
6
Chapter one
Introduction
Heterocyclic base
modification
Substitution of one(or
several) sugar atom (s)
Furanose ring breaking
(==>acyclonucleosides)
Base
HO
Nucleosidic linkage
displacement on the
sugar
N
O
inversion of configuration
(D==>L)
Modification of the ring
size (==> pyranose,..)
Anomeric inversion
( ==> )
OH
Addition of various
functions
Inversion, substitution or elimination
of hydroxyl groups
Figure (3): Main structure modifications susceptible to transform a natural nucleoside
(R=H or OH) into one of its analogues.
7
Chapter one
Introduction
requiring intracellular activation2c. Nucleoside analogues show importance in
several established chemotherapies (anticancer, antiviral and antibacterial ) and
other attractive fields like immunomodulation or regulation of gene expression
which could constitute new therapeutic approaches.10-12
1.2.1 BASE-MODIFIED NUCLEOSIDE ANALOGUES
1) 2/-DEOXY-2/-METHYLIDENE CYTIDENE (DMDS)13 [1] has been used as
anticancer drug but the 5-floro-2/-deoxy uridine (Fdurd) 14 [2] has been used
clinically as cancer ostatic agent, especially in the treatment of breast cancer
and of gastro intestinal tract.
NH2
O
F
N
N
OH
O
CH2
OH
N
OH
O
NH
O
O
H
OH
DMDS [1]
Fdurd [2]
2) 5-IODO-2/-DEOXYURIDINE (Idurd)15 [3] has been used as antiviral drug,
BRIVUDIN (BVDU)16 [4] represents highly potent and selective inhibitors of
Varicella-Zoster virus(VZV) replication in cell culture.
O
O
I
H
C
HN
NH
Br
C
H
OH
O
N
O
O
HO
OH
OH
Idurd [3]
N
O
OH
BVDU [4]
8
Chapter one
Introduction
3) DIDANOSINE (ddi)17 [5] and ZALCITABINE (ddg)18 [6] have been used as
antacids drugs.
NH2
O
N
N
OH
N
NH
OH
N
O
ddI [5]
O
N
O
ddG [6]
4) STAVUDINE (d4t)19, 20 [7] has been used as anti HIV-activity drugs
O
NH
OH
O
N
O
d4T [7]
1.2.2 SUGAR-MODIFIED NUCLEOSIDE ANALOGUES
1) VIDARABINE (AreA)21, 22 [8] has been used as antiviral drug, 5-(trifluoro
ethoxy methyl)-2/,3/-dideoxy uridine23 [9] and 5-[bis(trifluoro ethoxy)- methyl]2/,3/-dideoxy uridines23 [10] have been prepared and screened for their antiviral
activities.
N
O
O
NH2
F3CCH2OCH2
N
(F3CCH2O)2CH
NH
NH
HO
N
O
N
N
OH
O
HO
OH
AreA [8]
O
HO
N
O
Cl
[9]
9
[10]
O
Chapter one
Introduction
2) 4/-C-CYANO-2/-DEOXY PURINE24[11] have anti-HIV activity.
NH2
N
N
HO
N
N
O
N
C
OH
[11]
3) GEMCITABINE25 [12] and FLUDARABINE26 [13] have been used as
anticancer agents.
NH2
NH2
N
N
HO
N
O
O
HO
P
N
O
OH
F
OH
F
[13]
[12]
4)
N
O
HO
F
OH
N
O
3-(4-N-Acetylcytosine-1-yl)-7-hydroxy-1-hydroxymethyl-2,5-dioxabicyclo
heptan27[14] and 3-(4-N-Benzoyl cytosine-1-yl)-7-hydroxy-1-hydroxy methyl2,5-dioxabicyclo heptan27[15] have been prepared as Locked nucleic acid
(LNA) monomers which increased stabilities of duplexes with complementary
nucleic acids, as compared to the corresponding unmodified duplexes, showing
unprecedented thermal stabilities toward complementary DNA and RNA with
excellent mismatch discrimination and have application in molecular
diagnostics including gene array analyses.
10
Chapter one
Introduction
NHBz
NHAc
N
N
HO
HO
N
N
O
OH
O
O
O
O
OH
O
[14]
[15]
1.2.3. Nucleoside analogues as anticancer chemotherapy
Chemotherapy has been used in the treatment of cancer for several decades.
Established agents, alone or in combination with surgery or irradiation have
been increasingly used in the treatment of a range of common cancer types28.
The explanation of the selectivity of nucleoside analogues is that most of the
cancer cells are continuously undergo mitoses (in cycle). Consequently, theses
cells in cycle are more sensitive to nucleoside analogues (and to their nucleotide
metabolites) than resting cells.
Cancer chemotherapy has the potential to produce acute and chronic damage
in any organ system. Because, it has proved difficult to find general,
exploitable, biochemical differences between cancer cells and normally body
cells, an effective anticancer drugs must present some selective toxicity towards
malignant cells29,
30
. For example the fluorinated pyrimidines and their
nucleosides, constitute a very important class of antitumour agents31, among
them 5-fluorouracil (FUra, table 1)and 5-fluorodeoxyuridine (FdUrd,
floxuridine, table 1) are the most representative. 5-fluoro-2/-deoxyuridine and 5fluorouracil are used regularly for the treatment of breast cancer, tumors of the
gastrointestinal tract and other solid tumors32-33.
AraC (1-β-D-arabinofuranosylcytosine, cytosine arabinoside, cytarabine,
cytosar) (table 1) was synthesized in 1959. Acute leukemia's and lymphomas,
11
Chapter one
Introduction
especially acute nonlymphocytic leukemia, often respond successfully to
araC34.
Today, chemotherapy remains one of the hopes to control the mechanisms
involved in the growth of cancer cells. Cytotoxic drugs have reduced mortality
and restored many cancer patients to normal, and the emergence of new
antineoplastic nucleosides analogues like successful clinically tried 2/,2/difluorodeoxycytidine (dFdCyd, gemcitabine)35,36 (table 1) and 2/-deoxy-2/methylidenecytidine ((DMDC, table 1)37 appears as a promising prospect in the
treatment of cancer.
12
Chapter one
Introduction
Table 1: Nucleoside and nucleotide analogues licensed for clinical
anticancer use
Name
Structure
References
O
5-flurouracil
31
F
HN
O
N
H
O
5-fluorodeoxyuridine
32
F
NH
HO
O
N
O
OH
NH2
1,β-D-arabinofuranos-
35
N
yl cytosine
HO
O
N
O
HO
OH
2/,2/-difluorodeoxycyt-
NH2
36, 37
N
idine
HO
O
N
O
F
OH
F
2/-deoxy-2/-methylide-
NH2
38
N
necytidine
HO
N
O
OH
13
CH2
O
Chapter one
Introduction
1.2.4. Nucleoside analogues as antiviral chemotherapy
Over the past twenty six years, however, accumulating knowledge of viral
replication38 has made it possible to define specific targets which could be
affected by antiviral agents39. The virus replicative cycle (scheme 2) consists in:
(1) adsorption of the virion to the cell membrane, (2) penetration and uncoating,
(3) replication of the viral genome and protein synthesis, (4) assembly of
macromolecules into a virion, and finally (5), release of virions from the cell.
Any drug interfering selectively with one of these events is a candidate for
clinical use. In fact, the main targets of antiviral nucleoside analogues are
intracellular biosynthetic events, and their selectivity is generally due to the
inhibition of virus-associated or induced enzymes involved in nucleoside and
nucleotide metabolism.
1.2.4.1. Nucleoside analogues active against DNA viruses
5-Iododeoxyuridine (IdUrd, IDU, idoxuridine, iduviran, table 2) synthesized
in 1959, was the first clinically effective antiviral nucleoside analogue. IdUrd is
used in the topical treatment of herpetic keratitis and ocular herpes virus
infections40, 41.
5-Ethyldeoxyuridine (EDU, table 2) has been marketed (as Acedurid) for the
topical treatment of herpetic keratitis and clinical trials has been initiated in the
topical treatment of genital herpes42. 9-b-D-Arabinofuranosyladenine (araA,
vidarabine, vira-A, table 2) was first synthesized in 1960 and is used in the
treatment of herpetic keratitis and encephalitis43.
1.2.4.2 Nucleoside analogues active against RNA viruses
The major respiratory diseases in humans are caused by RNA viruses like
orthomyxovirus (influenza virus), paramyxovirus (respiratory syncytial virus,
parainfluenza virus), and picornavirus (rhinovirus).
14
Chapter one
Introduction
Scheme 2: virus replicative cycle.
15
Chapter one
Introduction
Ribavirine or 1-B-ribofuranosyl-1,2,4-triazole-3-carboxamide (virazole,
table 2) is a broad-spectrum antiviral agent active against both DNA and RNA
viruses. Its major clinical potentials lie in the treatment of respiratory syncytial
virus (RSV) and influenza A or B virus infection, where it can be administered
as a small-particle aerosol. Ribavirine is also effective in the therapy of Lassa
fever and other hemorrhagic fever virus infections40,42,44.
1.2.4.3. Nucleoside analogues active against retroviruses
Today, two important pathogenic human retrovirus have been discovered,
namely human T-cell leukemia virus (HTLV) 45 and human immunodeficiency
virus (HIV) which is the etiologic agent of acquired immune deficiency
syndrome (ADS)46, 47.
3/-azido-2/,3/-dideoxythymidine (azidothymidine, AZT, Azddthd, BW
A509U, retrovir, zidovudine, table 2) is the only drug approved for the
treatment of patients with AIDS49,50.
1.2.5. nucleoside analogues as antibiotherapy
Nucleoside antibiotics are by definition produced by microorganism
fermentation, and most of them have many biologic activities including
anticancer, antiviral and antiparasitic activity. Simple nucleoside antibiotics
(Fig.4) often act as inhibitors of nucleic acid syntheses50, 51.
16
Chapter one
Introduction
Table 2: Structural-defined drugs licensed for clinical antiviral use
Name
Structure
References
O
5-Iododeoxyuridine
I
OH
41,42
NH
N
O
O
OH
O
5-Ethyldeoxyuridine
43
C2H5
NH
HO
O
N
O
OH
NH2
9-β-D-ArabinofuranoN
44
N
osyl adinine
HO
N
N
O
HO
OH
O
1-β-ribofuranosyl-1,2-
41,43,45
N
NH2
,4-triazole-3-carboxaHO
mide
N
O
OH
3/-azido-2/,3/-dideoxy-
N
OH
O
49,50
H3C
NH
thymidine
HO
N
O
N3
17
O
Chapter one
Introduction
NH2
CH2CONH2
N
N
HO
N
HOOCCH2
N
O
O
N
OH
OH
OH
OH
OH
Neosidomycin
Psicofuranine
NH2
OH
Y
N
N
N
HO
HO
X
N
O
OH
O
N
OH
OH
N
OH
Tubercidin (X=N, Y=CH)
Pyrrolopyrimidine
Coformycin
Tetrahydroimidazodiazepine
Formycin A (X=CH, Y=N)
C-Nucleoside
Figure 4: Illustrative simple nucleoside analogues endowed with
antibacterial and antifungal activities.
18
Chapter one
1.2.6.
Introduction
C-NUCLEOSIDE
ANALOGUES
AND
BIOLOGICAL
ACTIVITY
By convention, a "c-nucleoside" is an analogue of nucleoside where the
heterocyclic nucleobase is an appended to the sugar via a carbon-carbon bond.
Thus, it is an analogue of the more common "N-nucleoside", where the
heterocyclic nucleobase is appended to the sugar via a carbon-nitrogen bond.
Although comparatively rare, C-nucleosides (table 3) exist in nature, where
they are found, free as natural products, and part of modified RNA molecules52 .
For many years, natural C-nucleosides and their synthetic analogues have
attracted wide interest in view of the importance of their biological activities53
as (antibacterial, antitumour and antiviral) agents.
Recently a renewed interest in these compounds arose as a consequence of
their potential applications in nucleic acid chemistry54. So the biological activity
exhibited by the naturally occurring C-nucleosides has given rise to research
directed toward their synthesis, as well as, toward the synthesis of many
analogues containing various heterocyclic and carbohydrates.
examples of the C-nucleoside analogues is the synthesized of the highly
functionalized imidazole55 (16), benzimidazole55 (17), 6-iodobenzimidazole55
(18) and indole55 (19) c-nucleosides.
I
N
HO
H
O
OH
OH
N
HO
N
[16]
N
H
O
OH
OH
[17]
[18]
N
N
HO
H
O
OH
OH
N
HO
H
O
OH
OH
[19]
Because the nucleobase is attached to the furanose by a carbon-carbon bond,
these are, nevertheless stable to phosphorylases and other enzymes that degrade
natural nucleosides.
19
Chapter one
Introduction
Table 3: Naturally occurring C-nucleosides
Name
Structure
References
CONH2
tiazofurin
HO
S
53
N
O
OH
OH
CONH2
pyrazofurin
53
HN
N
HO
OH
O
OH
OH
O
showdomycin
53
NH
HO
O
O
OH
OH
O
oxazinomycin
O
53
NH
HO
O
O
OH
OH
NH2
formycin
H
N
53
N
N
HO
O
OH
N
OH
O
formycine B
H
N
HO
N
O
OH
OH
20
53
NH
Chapter one
Introduction
1.3. Nucleosides Synthesis
1.3.1. Synthesis of nucleosides
Only 42 years ago, the synthesis of nucleosides with a C-N glycosidic
linkage have been reviewed56-59 with various representative examples of all of
the main type of chemical coupling procedures.
The overwhelming majority of nucleoside synthesis have been carried out
with the carbohydrate hydroxyls protected as explained below:
1) Condensation with heavy-metal salts of heterocycles (KeonigsKnorr method)
The initial syntheses of the naturally occurring purine nucleosides employed
the silver derivatine of the heterocycles60, but it was soon found that
chloromercuri derivatives provided greatly improved yields61.
Treatment of the chloromercuri derivative of 6-benzamidopurine [20] with
1-chloro-2,3,5-tri-O-acetyl-D-ribofuranose [21] followed by deacylation
afforded adenosine62[22].
O
NHCC6H5
CH2OAc
N
O
N
CL
+
adenosine
N
N
OAc
OAc
Hgcl
[20]
[21]
[22]
2) The Hilbert-Johnson method
a) With alkoxy pyrimidines
This method has been widely used for the synthesis of pyrimidine
nucleosides, and has been considerably refined since the initial research.
21
Chapter one
Introduction
A typical example of this method is the condensation of 2,4-di-alkoxy
pyrimidine [23] with the halosugar [24] to give the n-1-pyrimidine nucleoside62
[25].
OR
C
N
OR
CH2OBz
O
N
RO
N
N
CH2OBz
CL
+
OBz
OBz
OBz
OBz
[23]
O
O
[24]
[25]
b) Modified Hilbert-Johnson procedure
An extremely useful discovery was that the trimethylsilyl derivatives of
various pyrimidines could be useful in a modified Hilbert-Johnson procedure6365
. For example, heating 2,4-bis-tri-methyl silyloxy pyrimidine [26] with 2,3,5-
tri-O-benzoyl-D-ribofuranosyl chloride [27] in an inert dry solvent produced the
blocked uridine nucleoside [28].
O
C
OSiMe3
H
N
CH2OBz
N
O
+
N
BzOH2C
CL
O
O
Lewis
Me3SiO
N
acid
OBz
OBz
OBz
OBz
[26]
[27]
[28]
Other modification involved the use of various Lewis acids (AgClO4,
HgBr2) to improve the yields65- 67.
3) The fusion method
In this method acidic heterocyclic systems such as 2,6-dichloropurine [29]
react with peracylated sugar such as [30] at 150-155OC in a melt to give [31]
and the volatile acetic acid67,68. This fusion reaction is usually performed in the
22
Chapter one
Introduction
presence of catalytic amounts of Lewis acids, the reaction works with acidic
systems such as substituted or annulated imidazoles, purines, triazoles, or
pyrazoles69.
CL
N
N
CL
AcO
AcO
N
O
N
H
N
[29]
N
CL
O
+
CL
N
OAc
N
-AcOH
OAc
AcO
OAc
AcO
[31]
[30]
1.3.2 Synthesis of C-nucleosides
The biological activity exhibited by the naturally occurring c-nucleosides has
given rise to research directed toward their synthesis as well as toward the
synthesis
of
many
analogues
containing
various
heterocycles
and
carbohydrates. Two basic approaches have been employed for synthesis of cnucleosides. In the first a protected and activated heterocycle is condensed with
a suitably protected carbohydrate derivative in a carbon-carbon bond-forming
reaction.
In the second approach, which has been used in most examples, a
carbohydrate is constructed that contains both the carbon-carbon bond to
become the glycosidic linkage, and the functionality that will be elaborated to
the desired heterocycle.2d For example:
1- condensation of 2,4-di-tert-butoxy-5-lithiopyrimidine(32) with 2,4:3,5-diO-bezylidene-aldehydo-D-ribose(33) gave a mixture of alcohols70,
71
(34).
Removal of the t-butyl and benzylidene groups with aqueous acetic acid
followed by cyclization with HCl afforded pseudo uridine plus its -isomer. The
23
Chapter one
Introduction
altro isomer of (34) produced predominantly pseudo uridine (35), while the allo
isomer of (34) gave initially the -isomer of (35) as a major product.
CHO
Ot Bu
N
H
O
H
O
H
O
CHPh
N
+
CHPh
Ot Bu
CH2O
Li
[33]
[32]
O
HN
Ot Bu
N
N
NH
Ot Bu
H
H
OH
O
H
O
H
O
O
HOH2C
CHPh
O
CHPh
+
HO
isomer
HO
CH2O
[35]
[34]
2- Treatment of 2,3-O-isopropylidene-5-O-Trityl-D-ribofuranose(36) with
carbomethoxy methylene triphenyl phosphoran(37a)or cyano methylene
triphenyl phosphoran(37b) produced (38a)or (38b)72 (3:1, β/α) by a Wittig
reaction followed by Michael-type ring closure.
TrO
R
R1O
O
O
OH
+
XHC
pph3
O
O
H3C
OR2
OR2
CH3
[36]
[38]
[37]
a) X=CO2CH3
a) R=CH2CO2CH3
R1=trityl
b) X=Cn
R2=C(CH3)2
b) R= CH2CN
R1=trityl
R2=C(CH3)2
24
Chapter one
Introduction
3- treatment of bromoaldehyde [39] with 2-phenyl thio acetamide followed by
deblocking afforded the thiazole C-nucleoside [40]73.
CH2C6H5
N
S
HO
R1O
R
O
S
)C
H
CH
1 6 5
2CNH2
O
2)Deblock
OH
OR3
OR2
[40]
[39]
R=CHBrCHO, R1=trityl
R2=R3=C(CH3)2
25
OH
CHAPTER TWO
EXPERIMENTAL
Chapter two
Experimental
2.1 Apparatus and Materials
2.1.1 Apparatus
1) Infrared spectrophotometer:
IR
spectra
were
recorded
on
a
(SHIMADZU)
FTIR-8400
S
spectrophotometer. Solid samples were run in KBr disc, liquids were run as
smears.
2) Ultraviolet spectrophotometer:
UV spectra were recorded on UV-visible spectrophotometer, (SHIMADZU)
UV-1650PC.
3) Nuclear magnetic resonance spectrophotometer:
1
H-NMR and 13C-NMR spectra were taken with ultrashield (Bru Origen)
FT-NMR: 300MHz.
4) Melting point apparatus:
Melting points were determined in a (Gallen Kamp) m.p. apparatus with
sample contained in open capillary glass tube in an electrically heated metal
block apparatus, and are uncorrected.
5) Evaporation of solvent:
Using BUCHI vacuum rotary evaporator type 120 evaporated the solvent.
6) Thin layer chromatography:
TLC was performed on pre-coated plastic sheets with 0.25 mm layer of
silica Gel G/UV254, supplied by (CAMLAB). Column chromatography was
carried out with silica-gel 60 (Fluka). Compounds were detected by Iodine
vapour.
26
Chapter two
Experimental
2.1.2 Materials
All chemicals used were supplied from BDH, Merk, Fluka AG, Bushs SG and
Chem-Supply and were used without further purification, solvents were used
after distillation and drying.
2.2 Synthesis of nucleoside analogues
2.2.1. Synthesis of Ribopyranonucleoside analogues
1, 2, 3, 4-Tetra-0-benzoyl-β-D-ribopyranose74[41]
H
O
H
H
H
OBz
H
H
OBz
OBz
OBz
Pure D-ribose (5 g., 33.33 m.mol.) was added gradually to a well-stirred
mixture of dry pyridine (60 mL.) and benzoyl chloride (20 mL.), after agitation
for approximately forty-five minutes at (0oC) the reaction mixture was left in
the refrigerator overnight and then at room temperature for five hours, the
reaction was monitored by TLC (dichloromethane : ethanol, 8:2). When the
reaction was complete, it was poured on finely chipped ice and agitated
vigorously. The non-aqueous layer was augmented with ethylene dichloride (40
mL.) and then washed successively twice with ice-cold 3N sulfuric acid or until
the litmus paper becomes red, twice with saturated sodium bicarbonate solution
or until the solution becomes neutral to litmus paper and finally, thrice with
water. The organic layer was then dried with sodium sulfate, filtered and
concentrated in vacuo (40-45oC bath). The last traces of ethylene dichloride
were removed from the resulting syrup by evaporation in vacuo of absolute
alcohol (60 mL.). The resulting syrup was kneaded with a small quantity of
27
Chapter two
Experimental
alcohol, seeded and left at room temperature. (Seeded crystals were obtained
from dissolving (0.5 g.) of a syrup which obtained from another experiment in
(5 mL.) chloroform and then added to a column of silica gel 60, the column was
eluted with (CH2Cl2:C6H6, 8:2). The major fraction was evaporated to give a
pale yellow syrup which crystallized within three months in the refrigerator and
used as seeded crystals). As crystallization progressed over the course of three
days more alcohol was stirred into the mixture in order to improve its
filterability. The solid was then washed with (1:1) ether-alcohol mixture (9mL.)
and, finally with much absolute alcohol. And then recrystallized from (75%)
aqueous acetic acid. further recrystallization from 15 parts absolute ethanol and
from a mixture of 10 parts ether with 1 part pentane yielded 6.6 g. (35%) of
[41] as prismatic needles: mp 125-128 oC (literature mp 131oC); Rf 0.9; FTIR
(KBr disc), 3050 cm-1of (C-H) aromatic, 1728.1 cm-1of (C=O).
2, 3, 4-Tribenzoyl-β-D-ribopyranosyl bromide74, 75 [42]
H
O
H
H
H
Br
H
H
OBz
OBz
OBz
β-D-Ribopyranose tetrabenzoate [41] (3g., 5.3 m.mol.) was dissolved in
ethylene dichloride (3mL.). acetic anhydride (3 drops) and hydrogen bromide
in glacial acetic acid (6 mL.) (45% W./W.)were added and the mixture left at
room temperature for 2.5 hours. TLC (CH2CL2 : EtOH, 9:1) showed that the
reaction was complete, and the mixture was diluted with dry toluene (30 mL.)
and concentrated in vacuo (40-45oC bath). The resulting crystalline magma was
dissolved in a (3:1) mixture of ether and ethylene dichloride (100mL.) and
washed twice with ice-cold aqueous sodium bicarbonate solution and three
times with ice-water. After desiccation with anhydrous sodium sulfate, the
28
Chapter two
Experimental
solution was filtered and concentrated in vacuo to (10 mL.) and then diluted
with a mixture of ether (20 mL.) and pentane (50 mL.). On standing for several
hours in the refrigerator, elongated prisms was removed by filtration, washed
with ether and then with pentane. Recrystallization from the same mixture of
ethylene dichloride-ether-pentane yielded 1.56 g. (56%) of [42]; mp 148-150 oC
( literature m.p. 151-153oC); Rf 0.63; FTIR (KBr disc), 3058.89 cm-1 (C-H)
aromatic, 1720.39 cm-1 (C=O), 651.89 cm-1 (C-Br).
Phenothiazene76 [43]
S
N
H
A mixture of diphenyl amine (1.69 g., 0.01 mol.), sulfur (0.64 g., 0.02
mol.) and traces of iodine was heated in a sand bath maintained at (250-260oC)
for 6 hours. The reaction mixture was cooled and diluted in hot ethanol, the
solution was then added to water. The formed precipitate was filtered and
recrystallized from ethanol to yield 1.69 gm. (85%) of [43] as yellow crystals:
mp 175-178 oC (literature m.p. 180-181oC); purity of phenothiazene was
checked by TLC (chloroform:ethanol, 2:0.5); FTIR(KBr disc), 3340.48 cm-1
(NH), 1303.79 cm-1 (C=S).
Phenoxazine77 [44]
O
N
H
A mixture of o-aminophenol (109g, 1 mol.), ZnCl2 (2g.), and conc. H3PO4
(5 mL.) in (500 mL.) clasien flask was heated in a sand bath maintained at
(270-275oC) for 6 hours. The reaction mixture was cooled and extracted with
29
Chapter two
Experimental
cyclohexane in soxhlet extraction apparatus, the solvent was removed and the
formed precipitate recrystallized from ethanol to yield 54 gm. (50%) of [44] as
colorless needles: mp 150-152oC (literature m.p. 152-154oC); purity of
phenoxazine was checked by TLC (CHCL3:EtOH, 2:0.5); FTIR (KBr disc),
3404.1 cm-1 (NH), 1500.5 cm-1 phenoxazine ring.
1, 2, 3, 4-Tetrahydro-carbazole78 [45]
N
H
Phenylhydrazine (8mL., 89.79 m.mol.) was added to a mixture of
cyclohexanone (9 mL., 89.79 m.mol.) and glacial acetic acid (50 mL.). The
solution boiled under reflux for 5 minutes and then cooled, when the tetra
hydrocarbazole recrystallized, it was filtered at the pump, drained well, and
crystallized from aqueous ethanol to yield 11 gm. (70.8%) of [45] as colorless
crystals: mp 112-115oC(literature m.p. 118oC); FTIR(KBr disc), 3400.27 cm-1
(NH).
Bis(theophylline-7-yl) mercury(II)79 [46]
O
N
N
N
N
CH3
CH3
O
2
Hg
Theophylline hydrate (1g., 5 m.mol.) was dissolved in hot water (30 mL.)
and sodium hydroxide (0.2 g., 5.2 m.mol.) was added. A hot solution of
mercuric chloride (0.7g., 2.6 m.mol.) in ethanol (10 mL.) was added to the
vigorously stirred solution. The resulting suspension was cooled down and the
30
Chapter two
Experimental
product was filtered off and washed with distilled water until the filtrate was
neutral to litmus. After filtration by section a [46] was obtained and dried at
110oC to yield 1.34 g. (75%) as a white solid: mp > 347oC. It was stored over
phosphorus pentoxide.
Bis(phenothiazine-10-yl) mecury(II) [47]
S
N
2
Hg
This compound was prepared following a method described by Daroll and
Lowy80 and also by Yung and Fox81 for the synthesis of mercury salts.
Phenothiazine (1g., 5.02 m.mol.) was added to a boiling mixture of water (20
mL.) with ethanol (80 mL.) containing sodium hydroxide (0.2g., 5.02 m.mol.).
To the clear solution, 5mL. of hot ethanol containing mercuric chloride (0.68g.,
2.51 m.mol.) is added. A fine yellow precipitate formed immediately and after
continuous boiling for 10 minutes the precipitate color turned to dark gray.
After cooling for several hours at 5-10oC, the amorphous precipitate was
collected on a filter paper and washed repeatedly with water until the filtrate
was free from chloride ion by the (silver nitrate test). The solid was washed
successively with ethanol and ether and dried at 110oC to yield 0.33 g.
(23.16%) of [47]: m.p. > 250oC dec.; FTIR(KBr disc), disappearance of (NH) at
3340.48cm-1. It was stored over phosphorus pentoxide.
31
Chapter two
Experimental
Bis (phenoxazine-10-yl) mercury (II) [48]
O
N
2
Hg
This compound was prepared under similar conditions as for [47] to yield
0.35 gm (22.68%) of [48] as dark gray precipitate; mp >250oC, FTIR (KBr
disc), disappearance of (NH) at 3404.1 cm-1.
Bis (indol-1-yl) mercury (II) [49]
N
2
Hg
This compound was prepared under similar conditions as for [47] to yield
1.2 g. (64.93%) of [49] as fine white solid: mp > 300oC dec.; FTIR (KBr disc),
disappearance of (NH) at 3404.13cm-1.
Bis(1, 2, 3, 4-tetrahydro-carbazole-9-yl) mercury (II) [50]
N
2
Hg
1, 2, 3, 4-Tetrahydro-carbazole (1g., 5.78m.mol.) was added to a boiling
mixture of 1N-NaOH (30 mL.) and ethanol (30 mL.). To the stirred clear
solution, (5mL.) of hot ethanol containing (0.78 g., 2.89 m.mol.) of mercuric
32
Chapter two
Experimental
chloride was added. A voluminous yellow precipitate began to separate
immediately and turned gradually to a fine white precipitate after 10 minutes of
boiling. After cooling for several hours at 5-10oC, the fine precipitate was
collected on a filter paper and washed repeatedly with water until the filtrate
was free from chloride ion. The solid was washed once with ethanol and ether
and dried at 110oC for 2 hours to yield 0.88 gm. (56.1%) of [50] as a fine white
solid: mp 190oC; FTIR (KBr disc), disappearance of (NH) at 3400.27 cm-1. It
was stored over phosphorus pentoxide.
7-(2/, 3/, 4/-Tri-0-benzoyl-β-D-ribopyranosyl) thiophylline [51]
O
CH3
N
H
N
N
N
O
O
H
H
H
CH3
H
H
OBz
OBz
OBz
Dried bis(thiophylline-7-yl) mercury [46] (0.53g., 0.95m.mol.) was added
to a stirred solution of dried xylene (50 mL.) and celite (1 g.). Xylene (10 mL.)
was removed from the stirred refluxing mixture by means of the take-off
adapter, and 2, 3, 4-tribenzoyl-β-D-ribopyranosyl bromide [42], (1g., 1.9
m.mol.) was added to the vigorously-stirred refluxing suspension, the stopcock
of the take-off adapter was closed, and the refluxing was continued for further
3.5 hours at which time TLC (CH2CL2:EtOH, 99:1) showed the reaction was
complete. The hot turbid solution was filtrated; the filter cake was washed with
hot chloroform (3×10 mL.). The filtrate and chloroform washings were
combined and evaporated in vacuo and the residue was extracted with CHCl3
(20 mL.). The extract was washed with 30% aqueous potassium iodide (2×10
mL. portions) and water (2×10 mL. portions), then dried with anhydrous
33
Chapter two
Experimental
sodium sulphate and filtered. The filtrate was evaporated to dryness in vacuo to
give a yellow syrup which was treated with petroleum ether (40-60oC) (20 mL.)
to yield 0.68 g. (63%) of [51] as a white amorphous solid: mp 90-92 oC; Rf 0.52
;FTIR(KBr disc), 1467.73 cm-1(C-N); UV (chloroform), λmax at 276.5 nm and
at 246.5 nm; 1H-NMR (CDCl3): δ 8.25 (d, 1H), 8.3-7.2 (H Ar.), 6.78 (d, 1H),
6.34 (t, 1H), 5.88 (dd, 1H), 5.67 (m, 1H), 4.5-4.2 (complex m, 2H), 3.55 (s,
3H), 3.38 (s, 3H). 13C-NMR (CDCl3): δ 165.41, 165, 164.36, 154.77, 151.35,
148.73, 139.61, 133.77, 133.73, 133.57, 133.34, 130.11, 129.98, 129.83,
129.32, 128.86, 128.80, 128.48, 128.40, 128.08, 80.89, 69.91, 69.20,
66.77,64.31, 29.91, 28.07.
10-(2/, 3/, 4/-Tri-0-benzoyl-β-D-ribopyranosyl)phenothiazine [52]
S
N
H
O
H
H
H
H
H
OBz
OBz
OBz
This compound was prepared under the same conditions as [51] except the
reflux time was 2 hours to yield 0.45 g. (35%) of [52] as a green syrup: Rf 0.52;
FTIR (film), 1463.87 cm-1 (C-N).
10-(2/, 3/, 4/-Tri-0-benzoyl-β-D-ribopyranosyl)phenoxazine [53]
O
N
H
O
H
H
H
H
H
OBz
OBz
OBz
34
Chapter two
Experimental
This compound was prepared under the same condition as [51] except the
reflux time was (1.5) hours to yield 0.1 gm. (8.37%) of [53] as a dark brown
amorphous precipitate: Rf 0.3; mp 150oC dec.; FTIR (film), 1460 cm-1(C-N).
1-(2/,3/,4/-Tri-0-benzoyl-β-D-ribopyranosyl) indole [54]
H
N
O
H
H
H
H
H
OBz
OBz
OBz
This compound was prepared under the same conditions as [51] except the
reflux time was (3) hours to yield 0.63g. (59.84%) of [54] as Brown syrup: Rf
0.64; FTIR (film), 1421.4 cm-1 (C-N).
9-(2/,3/,4/-Tri-0-benzoyl-β-D-ribopyranosyl) tetrahydrocarbazol [55]
H
N
O
H
H
H
H
H
OBz
OBz
OBz
This compound was prepared under the same condition as [51] except the
reflux time was (3) hours to yield 0.21 g. (18.38%) of [55] as a Brown syrup; Rf
0.8; FTIR (film), 1465.8 cm-1 (C-N).
35
Chapter two
Experimental
7-β-D-Ribopyranosyl theophylline [56]
O
N
H
N
N
N
CH3
O
O
H
H
H
CH3
H
H
OH
OH
OH
7-(2/,3/,4/-Tri-0-benzoyl-β-D-ribopyranosyl) theophylline [51] (1 g.,
1.6 m. mol.) in 0.1 M methanolic sodium methoxide (32 mL.). the mixture
which
was
refluxed
with
stirring
for
1.5
hours
T.L.C
(
dichloromethane:ethanol, 99:1) showed that the reaction was complete, the
mixture was neutralized with glacial acetic acid and evaporated to dryness, the
residue was partitioned between water and chloroform and the aqueous phase
was evaporated to dryness in vacuo. The residue was dissolved in methanol (5
mL.) and then added to a column of silica gel 60; the column was eluted with
(CHCl3-MeOH, 9:1). The major fraction was evaporated to give 0.48 g.
(97.09%) of [56] as a white amorphous powder: m.p. 250 oC dec.; Rf 0.26;
FTIR (KBr disc), 3406.1 cm-1 (OH) ; UV (H2O) λmax at 245 nm; 1H-NMR
(EtOD): δ 8.05 (s, 1H), 5.85 (d, 1H), 4.15 (t, 1H), 4.05 (dd, 1H), 3.95-3.6
(complex m, 3H), 3.55-3.4 (s, 6H), 3.25 (s, 3H). 13C-NMR (CDCl3): δ 152.29,
149.05, 146.42, 138.91, 80.37, 69.45, 67.45, 64.04, 63.02, 26.72, 24.98.
This method was also used to obtain:
36
Chapter two
Experimental
10- β-D-Ribopyranosyl phenothiazine [57]
S
N
H
O
H
H
H
H
H
OH
OH
OH
The compound was obtained in 0.18 gm. (36.57%) as a pale yellow solid: Rf
0.04; mp 140oC dec.; FTIR (KBr disc), 3421 cm-1 (OH).
10- β-D-Ribopyranosyl phenoxazine [58]
O
N
H
O
H
H
H
H
H
OH
OH
OH
The compound was obtained in 0.18 g. (36.85%) of a white solid: Rf 0.03;
mp 170oC dec.; FTIR (KBr disc), 3450 cm-1 (OH); 1H-NMR (CDCl3): δ 8.2-6.9
(aromatic H), δ 6.66 (d, 1H), δ 5.02 (d, 1H), δ 4.82 (1H), δ 4.22 (dd, 1H), δ
3.6-3.15 (complex m, 5H).
1- β-D-Ribopyranosyl indol [59]
N
H
O
H
H
H
H
H
OH
OH
OH
37
Chapter two
Experimental
The compound was obtained in 0.27 g. (62%) of a pale yellow solid: Rf 0.04;
mp 158oC dec.; FTIR (KBr disc), 3398.3 cm-1 (OH); 1H-NMR (CDCl3): δ 7.85
(t, 2H), δ 7.6-6.9 (aromatic 4H), δ 6.45 (d,1 H), δ, δ 4.8 (t, 1H), δ 4.7 (m, 1H),
δ 4.55 (dd, 1H), δ 4 (dd, 2H), δ 3.35 (complex m, 3H); 13C-NMR (DMSO): δ
129.44, 127.5, 121.6, 121.16, 120.74, 120.23, 119.97, 118.6, 79.65, 71.02,
69.06, 67.26, 64.48.
9- β-D-Ribopyranosyl tetrahydro carbazol [60]
N
H
H
H
H
O
H
H
OH
OH
OH
The compound was obtained in 0.37 g. (69.8%) as a white solid: Rf 0.04; mp
146oC dec.; FTIR (KBr disc), 3425 cm-1 (OH).
2.2.2. Synthesis of Ribofuranonucleoside analogues
1, 2, 3, 5-Tetra-O-benzoyl-β-D-ribofuranose82 [61]
BOz
OBz
O
H
H
OBz
H
OBz
D-Ribose (5 g., 33.33 m.moL.) was dissolved in an anhydrous pyridine (50
mL.) and the solution held at 100oC for one minute. Benzoyl chloride (19 mL.)
was added at such a rate that the temperature of the rapidly stirred mixture
remained at (98-102oC), the reaction was monitored by TLC (dichloromethane :
ethanol, 8:2). When the mixture had cooled to 35oC, chloroform (50 mL.) was
38
Chapter two
Experimental
added and the solution washed successively with cold water, 3N sulfuric acid
and aqueous sodium bicarbonate. Moisture was removed with anhydrous
sodium sulfate, after the solution was filtered it was concentrated in vacuo (50o
bath) to a stiff, dark-brown sirup which was dissolved in absolute ethanol (25
mL.) and reconcentrated in vacuo. All attempts for crystallizing the syrup from
ethanol absolute were unsuccessful yielded 6.3g. (34%) of [61] as dark-brown
syrup: FTIR (film), 3076 cm-1of (C-H) aromatic, 1724.2 cm-1of (C=O).
1-O-Acetyl-2, 3, 5-tri-O-benzoyl-β-D-ribose83 [62]
BZO
OAc
O
H
H
OBZ
H
OBZ
β-D-Ribofuranose tetrabenzoate [61] (0.41 g., 0.07 m.moL.) was dissolved
in acetic anhydride (4 mL.) and the solution treated at 20oC with (1 mL.) of a
solution of fused zinc chloride (2 g.) in acetic anhydride (10 mL.). After (4 hr.)
the reaction mixture was monitored by TLC (dichloromethane : ethanol, 8:2)
and poured on ice. The gum was precipitated from absolute alcohol and
recrystallized from the same solvent to give o.1 g. (28%) of [62] as hexagonal
micaceous plates: mp 105-109 oC (literature mp 129-130oC); FTIR (KBr disc),
3075 cm-1of (C-H) aromatic, 2990 cm-1 of (C-H) aliphatic, 1720 cm-1of (C=O).
1, 2, 3, 5-tetra-O-acetyl-β-D-ribofuranose84 [63]
AcO
OAc
O
H
H
OAc
H
OAc
39
Chapter two
Experimental
D-Ribose (5 g., 33.33 m.moL.) was dissolved in acetic anhydride (20 mL.)
and acetic acid (15 mL.) and treated with sulfuric acid (0.6 mL.) with icecooling and left at room temperature for 1 hr. further sulfuric acid (1 mL.) was
then added with ice-cooling and the solution left at room temperature for
further (2 hr.), the reaction was monitored by TLC (dichloromethane : ethanol,
8:2) and the resulting dark-red solution was then treated with an excess of
sodium acetate (4.5 g.) and the mixture co-evaporated several times with
ethanol to give a stiff, brown syrup, to which was added chloroform (30 mL.).
the mixture was washed with water, the chloroform solution dried with
anhydrous sodium sulfate and after filtration it was evaporated to give a syrup
solid, which, on treatment with ice-cold ethanol, (6 mL.), gave 4.4 g. (42%) of
[63] as white crystals: mp 92-95 oC (literature mp 81-83oC); FTIR (KBr disc),
2947 cm-1 of (C-H) aliphatic, 1749.67 cm-1of (C=O).
Attempt for the synthesis of 7-(2/, 3/, 4/-Tri-0-acetyl-β-Dribofuranosyl) thiophylline [64]
O
N
N
AcO
O
H
H
OAc
H
OAc
N
N
CH3
O
CH3
A mixture of dry 1, 2, 3, 5-O-acetyl-β-D-ribofuranose84 [63] ( 0.25 g., 0.34
m.moL.), Bis(theophylline-7-yl) mercury(II) [46] (0.25 g., 1.25 m.moL.) and
celite (0.25 g.) was powdered and dissolved in dichloroethane (10 mL.).
1, 2-Dichloroethane was carefully purified before using by refluxed for (2 hr.)
over P2O5 and distilled and the procedure was repeated, finally it was stored
40
Chapter two
Experimental
over 5Ao molecular sieves. Stannic chloride (3 drops) was dissolved in
dichloroethane (5 mL.) and the mixture was added to the stirred solution. The
reaction was monitored by TLC (dichloromethane : ethanol, 8:2) . After (3
days) at 22oC no changes appeared on TLC sheets, the reaction temperature
was raised to 80oC for (20 hr.), also no changes had been detected. Acetonitrile
was used instead of dichloromethane and yet no development was discovered.
The reaction was stopped by filtration and the filtrate washed with hot
chloroform (3× 10 mL.). The filtrate was combined with the washings and
evaporated in vacuo, the residue was extracted with CHCl3 (20 mL.) and the
extract was washed with (30%) aqueous potassium iodide (2× 10 mL. portions)
and water (2× 10 mL. portions), then dried with anhydrous sodium sulfate and
filtered. The filtrate was evaporated to dryness in vacuo to give a pail yellow
syrup. IR(film) shows the same absorptions for the starting material.
The same method was used with different kind of nitrogen bases derivatives
[47-50]. The same above result were obtained.
41
CHAPTER
THREE
RESULTS &
DISCUSSION
Chapter three
Result & Discussion
Results and Discussion
In this work, a new type of nucleoside analogues have been synthesized
and characterized.
D-ribose has been chosen as a starting material due to its great
significance, not only as a component of ribonucleic acid RNA but also the
unique nature of D-ribose as compared with the other sugars.
3.1 Synthesis of Ribopyranonucleoside analogues
The strategy used for the synthesis of [56], [57], [58], [59] and [60]
nucleoside analogues using different kinds of heterocyclic compounds as a
nitrogen bases by the method of ( Keonigs-Knorr ), was started with D-ribose
in a series of reactions shown in ( scheme 3 ).
In order to obtain the target, 1, 2, 3, 4-tetra-O-benzoyl-β-D-ribopyranose
[41] was chosen as the starting step.
The pyranose structure of D-ribose (thermodynamically stable) had known
to be more reactive than the furanose structure, and moreover it had received
less attention than the furanose form which occurs in the natural nucleosides.
3.1.1. Synthesis of 2, 3, 4-tribenzoyl-β-D-ribopyranosyl bromide
[42]
In order to obtain [42], D-ribose was first converted to 1, 2, 3, 4-tetra-Obenzoyl-β-D-ribopyranose [41]. In the cyclic form of the ribopyranose, there
are three types of hydroxyl groups, namely the glycosidic hydroxyl group
attached to the anomeric carbon atom C1 and three secondary hydroxyl
groups attached to C2, C3 and C483a. Esters are generally used to block
hydroxyl groups to deactivate their oxygen atoms and, by doing so, we
prevent them from attacking nucleophile acceptors. The esters most
42
Chapter three
Result & Discussion
O
O
Ph CCl/Py
D-Ribose
BzO
BzO
OBz
BzO
BzO
OBz
Br
O
HBr/g.acetic acid
OBz
[42]
[41]
Het
condensation
xylene/ref.
N
2
Hg
Het
N
O
BzO
BzO
OBz
[51-55]
Protecting group
cleavage
NaOCH3/MeOH
Het
O
N
HO
OH
HO
[56-60]
O
N
Het
N
Hg
N
=
N
2
Hg
N
CH3
S
O
,
O
2
CH3
,
,
N
N
N
2
Hg
2
Hg
Hg
N
Hg
2
Scheme (3) : Synthesis of nucleoside analogues [51], [52], [53], [54], [55],
[56], [57], [58], [59] and [60].
43
2
Chapter three
Result & Discussion
commonly used for this purpose are the acetates and benzoates83b, so the
reason for conversion of D-ribose to [41] was to protect the hydroxyl groups
with benzoate group which is known to be more stable than the acetyl group
toward acid conditions, but are readily hydrolyzed by dilute alkaline, hence
they are very useful as blocking groups.
Benzoylation of D-ribose in pyridine with benzoyl chloride at low
temperature gave in 35% yield a crystalline β-D-ribopyranose tetrabenzoate
[41] and a 25.53% of amorphous mixture remaining in the mother liquor
which presumably contains the α-anomer75 [41a].
H
H
D-ribos
PhCOCl/PY
100 C, 24hr.
H
O
H
H
OBz
H
OBz
OBz
[41]
OBz
35%
H
H
+
H
O
H
H
H
OBz
OBz
OBz
[41a]
OBz
25.53%
The ribobenzoate [41] was characterized by its FT-IR spectrum and m.p.
which was identical with the literature. The FT-IR spectrum (Fig. 5) showed
stretching band at 3050cm-1 for aromatic (C-H), 2950cm-1 for aliphatic (C-H),
1728.1 cm-1 for (C=O) ester group and 1583.45 cm-1for (C=C) aromatic bands.
44
Figure (5): IR Spectrum of compound [41]
Chapter three
Result & Discussion
45
Chapter three
Result & Discussion
2, 3, 4-tri-O-benzoyl-β-D-ribopyranosyl bromide [42] was synthesized by
converting the benzoyl group of C1 (anomeric) in [41] to a good leaving
group, the bromide group. Both the crystalline tetrabenzoate [41] and its αisomer [41a] gave 71 to 85% of
2, 3, 4-tribenzoyl-β-D-ribopyranosyl
bromide as well-shaped prismatic crystals76 when treated with a solution of
hydrogen bromide in glacial acetic acid. At the same time there is formed in
relatively meager quantity (5% yield) a second compound crystallizing in
long needles84; which is found to be the α-isomer [42a] of the major product
of the reaction.
H
H
O
H
OBz
45% HBr in-
H
H
H
H
H
H
H
OBz
OBz
OBz
glical acitic acid
H
O
Br
H
OBz
OBz
[41]
H
H
+
H
O
H
H
Br
OBz
OBz
[42] 71-85%
H
OBz
[42a]
OBz
5%
The benzoylated D-ribopyranosyl halides appear to be considerably more
stable than their acetyl analogs and the tribenzoyl- β-D-ribopyranosyl bromide
[42], appears to be stable indefinitely when stored over calcium chloride and
potassium hydroxide at 5oC3. Thus the bromination of [41] with HBr in
CH2Cl2 resulted in a smooth conversion to the β-anomer [42].
The preparation of [42] is believed to follow the same mechanism
proposed by Fletcher and Ness86 which involves the formation of 1,2benzoxonium ion B as shown in (scheme 4).
46
Chapter three
Result & Discussion
Br
H
O
H
H
H
OBz
H
H
H
H
OBz
O
OBz
O
O
H
H
H
OBz
C
O
OBz
O
ph
ph
[A]
[B]
H
Br
O
H
H
H
H
H
OBz
OBz
OBz
[42]
Scheme (4): The mechanism of the bromoination reaction.
The FT-IR spectrum of [42] (fig. 6) showed stretching bands at 3055.03cm-1
aromatic (C-H), 2970cm-1 alifatic (C-H), 1722.31cm-1 carbonyl (C=O) (of the
benzoate groups), 1600.81cm-1 (C=C) and clearly showed the (C-Br)
stretching band at 651.89 cm-1.
47
Figure (6): IR spectrum of compound [42]
Chapter three
Result & Discussion
48
Chapter three
Result & Discussion
3.1.2. Synthesis of nitrogen bases and their mercury salts (II)
Phenothiazine [43]
The first syntheses of phenothiazine was reported by Bernthsen8. Interest
in phenothiazine was due to its pharmacological properties.
Phenothiazine drugs now play a very important part in chemotherapy. The
first comprehensive survey on reactivity of phenothiazine is that of Massie87
(1954), since then many significant results have been reported which are of
interest not only for worker in the phenothiazine field, but also for the entire
heterocyclic chemistry.
Phenothiazine was synthesized using the Ring-Clouser method by the
reaction of diphenylamine with sulfur at 250-260oC in the presence of a small
amount of Iodine as catalyst.
H
H
N
N
S,I2
+ H2S
0
260 C/6hours
S
[43]
The m.p. and IR spectrum were identical with that reported in the litriture.
The FT-IR spectrum of phenothiazine showed strong stretching band at
3340.48cm-1 for (N-H), at 1571.88cm-1 and 1596.95cm-1 assigned to the
aromatic system88 (see Fig. 7).
49
Figure (7): IR spectrum of compound [43]
Chapter three
Result & Discussion
50
Chapter three
Result & Discussion
Phenoxazine [44]
Phenoxazine was made first by Bernthsen89 in 1887. Phenoxazine was
prepared by the condensation of o-aminophenol in presence of H3PO4 and
ZnCl2 as Lewis acid catalyst at 270-275oC, this method was of choice because
it gives [44] in good yield77.
OH
O
H3PO4/ ZnCl2
NH2
270-2500C
N
H
[44]
The formed product [44] was characterized based on its infrared spectrum
bands. The FT-IR spectrum of phenoxazine [44] showed strong stretching
band at 3404cm-1 and 1500.5 cm-1 assigned to phenoxazine ring90 (see fig. 8).
1, 2, 3, 4-tetrahydrocarbazole [45]
1, 2, 3, 4-tetrahydrocarbazole [45] is a derivative of indole which may be
prepared by the Fisher Indolisation reaction80a.
The Fisher indolisation reaction occurs when the phenylhydrazone of a
suitable aldehyde or ketone undergoes cyclization with loss of ammonia, under
the influence of various reagents, such as zinc chloride, alcoholic hydrogen
chloride, or acetic acid80a.
1, 2, 3, 4-tetrahydrocarbazole [45] was synthesized by converting
cyclohexanone to its phenyl hydrazone, and the latter, without isolation, was
converted to 1, 2, 3, 4-tetra hydrocarbazole [45] by boiling with acetic acid for 5
minutes.
51
Figure (8): IR Spectrum of compound [44]
Chapter three
Result & Discussion
52
Chapter three
Result & Discussion
O
H2
4
3
C6H5NH.NH2 +
C6H5HNN
N
1
H2
H2
2
H2
H
[45]
It was obtained as a colorless crystals in 70.8% yield which was characterized
by comparing m.p with litriture m.p and by IR spectral data.
The FT-IR spectrum (fig. 9) showed a stretching band at 3400.27cm-1 for the
(N-H) band.
Bis(theophylline-7-yl) mercury (II) [46]
The theophylline mercury salt was prepared by dissolving the theophylline
in hot water and then adding sodium hydroxide and the resulting solution was
mixed with hot ethanolic mercury chloride solution which formed bis
(theophylline-7-yl) mercury81 [46]
Bis(phenothiazine) mercury (II) [47]
Bis(phenothiazine)
mercury
[47]
was
prepared
by
dissolving
the
phenothiazine in a boiling mixture of water, sodium hydroxide and ethanol,
and the resulting solution was mixed with hot ethanolic mercuric chloride
solution. After boiling for 10 min. the phenothiazine mercury salt [47] was
formed, its FT-IR spectrum showed the disappearance of (N-H) band at
3340.48 cm-1 (see fig. 10).
This method was used to prepare:
Bis(phenoxazine) mercury [48]
FT-IR spectrum showed the disappearance of (N-H) band at 3404.1cm-1 (see
fig. 11).
53
Figure (9): IR Spectrum of compound [45]
Chapter three
Result & Discussion
54
Figure (10): IR Spectrum of compound [47]
Chapter three
Result & Discussion
55
Figure (11): IR Spectrum of compound [48]
Chapter three
Result & Discussion
56
Chapter three
Result & Discussion
Bis(indol) mercury [49]
FT-IR spectrum showed the disappearance of (N-H) band at 3404.13cm-1
(see fig. 12).
Bis( 1, 2, 3, 4-tetrahydro-carbazole) mercury [50]
FT-IR spectrum showed the disappearance of (N-H) band at 3400.27cm-1
(see fig. 13).
3.1.3. Synthesis of benzoylated nucleoside analogues
7-(2/, 3/, 4/-tri-O-benzoyl-β-D-ribopyranosyl) theophylline [51]
The theophylline (1,3-dimethyl xanthine) base has been used because of
its availability and due to the fact that only one of its nitrogen atoms N-7 is
reactive91, 92.
Bis(theophylline-7-yl) mercury [46] was treated with 2, 3, 4-tribenzoyl-βD-ribopyranosyl bromide [42] under reflux for 3.5 hrs in xylene in presence of
Celite.
The 7-(2/, 3/, 4/-tri-O-benzoyl-β-D-ribopyranosyl) theophylline [51] was
obtained as a white amorphous precipitate in 63% yield and m.p. 90-92oC
which was characterized by IR, U.V spectral data.
The FT-IR spectrum (fig. 14) showed a stretching bands at 3090cm-1
aromatic (C-H), 2940cm-1 aliphatic (C-H), 1731.96cm-1 carbonyl (C=O) (of
the benzoate groups), 1662.52 cm-1 (C=O) (amide), 1590cm-1 (C=C), 1544.88
cm-1 (C=N), 1467.73 cm-1 (C-N) band.
The U.V spectrum (fig. 15) showed an absorption a λmax at 276.5 nm due
to π-π* transition of dienone system (C=C-C=O) of the theophylline ring, and
λmax at 246.5 nm due to π-π* transition which indicate the presence of the
(C=C) group of the benzoate.
The 1H-NMR spectrum as expected, showed (fig. 16 and 17) a signal at δ
8.25 ppm was assigned to H-6 proton (integrated for 1 H), between δ 8.3-7.2
was assigned to aromatic protons, at δ 6.78 ppm was assigned to H-1/ proton
57
Chapter three
Result & Discussion
(integrated for 1 H), at δ 6.34 was assigned to H-3/ proton (integrated for 1 H),
at δ 5.88 ppm was assigned to H-2/ proton (integrated for 1H), at δ 5.67 ppm
was assigned to H-4/ proton (integrated for 1H), between δ 4.5-4.2 ppm was
assigned to 2H-5/ protons (integrated for 2H), at δ 3.55 ppm was assigned to
CH3-3 proton (integrated for 3H) and at δ 3.38 ppm was assigned to CH3-1
proton (integrated for 3H).
The 13C-NMR spectrum showed (fig.18) a signal at δ 165.41 ppm assigned
to the carbonyl carbon of the benzoate group, a signal at δ 165 ppm assigned
to the carbonyl carbon of the benzoate group, a signal at δ 164.36 ppm
assigned to the carbonyl carbon of the benzoate group, a signal at δ 154.77
ppm assigned to C-2 (the carbonyl carbon) of the base, a signal at δ 151.35
ppm assigned to C-4 (the carbonyl carbon) of the base, a signal at δ 148.73
ppm assigned to C-6, a signal at δ 139.61 ppm assigned to C-9, a signal at δ
133.77 ppm assigned to C-8,a signals at δ beween 133.73 ppm and 128.08
ppm assigned to the aromatic carbons of the benzoyl croups, a signal at δ
80.89 ppm assigned to C/-1, a signal at δ 69.91 ppm assigned to C/-5, a signal
at δ 69.2 ppm assigned to C/-2, a signal at δ 66.77 ppm assigned to C/-3, a
signal at δ 64.31 ppm assigned to C/-4, a signal at δ 29.91 ppm assigned to C3, a signal at δ 28.07 ppm assigned to C-1.
O
CH3
N
H
H
O
H
Theophyline mercury salt
H
H
H
OBz
in xylen / refi.
H
H
H
OBz
[42]
[51]
58
CH3
H
OBz
OBz
N
O
H
H
OBz
N
Br
N
OBz
O
Figure (12): IR Spectrum of compound [49]
Chapter three
Result & Discussion
59
Figure (13): IR Spectrum of compound [50]
Chapter three
Result & Discussion
60
Figure (14): IR Spectrum of compound [51]
Chapter three
Result & Discussion
61
Figure (16): 1H-NMR spectrum of compound [51]
Chapter three
Result & Discussion
62
Figure (17): The expansion of 1H-NMR spectrum of compound [51]
Chapter three
Result & Discussion
63
Figure (18): 13C-NMR spectrum for compound [51]
Chapter three
Result & Discussion
64
Chapter three
Result & Discussion
10-(2/, 3/, 4/-tri-O-benzoyl-β-D-ribopyranosyl) phenothiazine [52]
Phenothiazine is the largest of the 5 main classes of antipsychotic drugs.
Antipsychotic agents are mainly used for prevention or treatment of
schizophrenia and serious psychotic diseases by improving mood and
behavior. They relieve delusions, hallucinations, agitation, thought disorder
and prevent relapse. Moreover, they produce emotional quieting, psychomotor
slowing and indifferece93, 94.
In addition to its formerly mentioned biological activity, we chose
phenothiazine because it possesses a three fused aromatic rings and a (C=S)
bond which enables use to investigate its effect on the new prepared
nucleoside analogues.
The phenothiazine salt [47] was, coupled with [42] under similar condition
described above for the synthesis of the nucleoside analogue [51]. Treatment
of [47]and the bromide [42] under reflux condition in xylene in presence of
celite afforded the expected phenothiazine nucleoside [52] as a syrup in
35.01% yield, which was characterized by FT-IR spectrum showed a
stretching bands at 3075 cm-1 aromatic (C-H), 2930 cm-1 aliphatic (C-H), 1730
cm-1 carbonyl (C=O), 1600 cm-1(C=C) (see fig. 19).
S
H
N
H
O
H
Br
Phenothiozine mercury salt
H
H
H
OBz
OBz
in xylene /refl.
O
H
H
H
H
H
H
OBz
OBz
OBz
[42]
[52]
65
OBz
Figure (19): IR Spectrum of compound [52]
Chapter three
Result & Discussion
66
Chapter three
Result & Discussion
10-(2/, 3/, 4/-tri-O-benzoyl-β-D-ribopyranosyl) phenoxazine [53]
Phenoxazine was also selected as a nitrogen base in the preparation of
ribonucleoside analogues because it possesses like phenothiazine a three
fused aromatic rings and it has a (C=O) bond which enables us to investigate
its biotosieal effect as part of the prepared nucleoside analogues. The
heterocyclic oxygen atom of the phenoxazine nucleus plaices certain
restriction on the aromaticity of this ring system, which appears to be some
what less aromatic than the phenothiazine system95.
Similarly, the reaction of [42] with phenoxazine mercury salt [48] gave 10-(2/,
3/, 4/-tri-O-benzoyl-β-D-ribopyranosyl) phenoxazine [53] as a dark brown
amorphous precipitate in 8.37% yield, m.p. 150oC dec., FT-IR spectrum
showed a stretching bands at 3075 cm-1 aromatic (C-H), 2927.74 cm-1
aliphatic (C-H), 1728 cm-1 carbonyl (C=O), 1600.81 cm-1 (C=C) and 1488.94
cm-1 (phenoxazine ring), 1450 cm-1 (C-N) (see fig. 20).
O
H
N
H
O
H
Br
Phenothiozine mercury salt
H
H
H
H
H
H
OBz
OBz
in xylene /refl.
O
H
H
H
OBz
OBz
OBz
[42]
[53]
67
OBz
Figure (20): IR Spectrum of compound [53]
Chapter three
Result & Discussion
68
Chapter three
Result & Discussion
1-(2/, 3/, 4/-tri-O-benzoyl-β-D-ribopyranosyl) indole [54]
The synthesis of a new type of indole nucleoside analogues shows
promising as biologically active agents.
Indole nucleoside analogues should be less susceptible to glycosidic bond
cleavage because, the 3-position of the indole nucleoside cannot be
protonated96. For example: the indole nucleosides 2, 5, 6-trichloro-1-(β-Dribofuranosyl) indole (TCRI [60a]) was synthesized but was found to be
devoid of antiviral activity97. In contrast, the three formyl indole nucleoside
[60b] was found to be both a potent and selective inhibitor of Human
Cytomegalovirus (HCMV) replication in antiviral assays.
CHO
Cl
Cl
Cl
Cl
Cl
N
Cl
HO
N
HO
O
OH
O
OH
OH
[60a] TCRI
OH
[60b]
The reaction of [42] with indole mercury salt [49] gave 1-(2/, 3/, 4/-tri-Obenzoyl-β-D-ribopyranosyl) indole [54] as brown syrup in 59.84% yield, FTIR spectrum showed a stretching bands at 3075 cm-1 aromatic (C-H), 2900cm1
aliphatic (C-H), 1730 cm-1 carbonyl (C=O), 1625 cm-1 (C=C) (see fig. 21).
H
N
H
O
H
H
H
Br
H
H
H
H
OBz
OBz
O
H
Indol mercury salt
in xylene /refl.
H
H
OBz
OBz
OBz
[42]
OBz
[54]
69
Figure (21): IR Spectrum of compound [54]
Chapter three
Result & Discussion
70
Chapter three
Result & Discussion
9-(2/, 3/, 4/-tri-O-benzoyl-β-D-ribopyranosyl) tetrahydrocarbazole
[55]
The reaction of [42] with 1, 2, 3, 4-tetrahydrocarbazole mercury salt [50]
gave 9-(2/, 3/, 4/-tri-O-benzoyl-β-D-ribopyranosyl) tetrahydrocarbazole [55] as
a brown syrup in 18.38% yield which was characterized by FT-IR and UV
spectral data, the FT-IR spectrum (fig. 22)showed a stretching bands at 3075
cm-1 aromatic (C-H), 2950 cm-1 aliphatic (C-H), 1730 cm-1 carbonyl (C=O),
1650 cm-1 (C=C), 1465.8cm-1 (C-N).
H
O
H
1,2,3,4Tetrahydrocarbazat-
H
H
H
OBz
mercury salt in xylene/ refl.
OBz
O
H
H
H
H
OBz
N
H
Br
H
H
OBz
OBz
[42]
OBz
[55]
The syntheses of β-ribonucleosides [56], [57], [58], [59] and [60] via the
Keonigs-Knorr method is believed to include a neighboring-group
participation by a 2/-benzoyloxy group which directs the nucleophile to the βform so the β-pyranonucleoside is the major reaction product98 and the
mechanism expected to follow the same mechanism proposed by Jeanlos and
Fletcher99 for the β-pyranosyl nucleosides showed in scheme (5). The β-Danomer [42] is rapidly dissociated, with C2-benzoyloxy-group participation,
to yield the 1, 2-α-D-cyclic carbonium ion (A). Thus, the fact that the rate of
formation of the β-D-nucleoside from the intermediate ions is much greater
than the rate of formation of the α-D-nucleoside [C] is understandable. The
71
Chapter three
Result & Discussion
formation of the α-D-anomer from intermediate carbonium ions would require
the formation of such ions as [B], and the rate controlling stage for the βàα
conversion was assumed to be the formation of this type of ion. Although the
main carbonium ion formed on the dissociation of the β-D-anomer [42]
undoubtedly possessed the 1, 2-α-D-cyclic structure[A], a fraction of the
dissociations may have led to the ion [B], which can form the α-nucleoside
[C], nevertheless, it was suggested that this rout of reaction may be less
favorable than one wich would involve the 1, 2-α-D-cyclic ion [A] as the main
source of ions capable of forming the α-nucleoside [C]. On this basis, the rate
controlling stage for the βàα conversion would be the rearrangement of the
readily formed 1, 2-α-D-cyclic ion to ions capable of forming the αnucleoside. An attractive argument in favor of this theory is the fact that the
free energy of activation for the formation of such an ion as [B] should be
considerably less from the ion [A] than from the β-nucleoside. Furthermore, it
seems reasonable to expect that, once formed, an ion such as [B] would have a
strong tendency to assume the relatively much more stable structure[A]; and,
consequently, in the absence of a tendency for the ion [A] to be rearranged to
ions capable of forming the α-nucleoside [B], the βàα conversion would be
extremely slow100.
72
Chapter three
Result & Discussion
Base
OBz
OBz
O
H
C
Br
O
C
H
H
O
Base
C
OBz
CH
OBz O
O C O
O
C
OBz
OBz OBz
[A]
[42]
O
C
H
[51-55]
O
Base
OBz
OBz OBz
OBz
Base
OBz OBz
[B]
[C]
Scheme (5): Mechanism of β and α nucleoside synthesis
73
Figure (22): IR Spectrum of compound [55]
Chapter three
Result & Discussion
74
Chapter three
Result & Discussion
3.1.4. Hydrolysis of the benzoate groups
A number of methods has been used for the hydrolysis of the benzoyloxy
group; for example, methanolic ammonia101 (liq NH3-MeOH) or sodium
methoxide102.
Treatment of the theophylline nucleoside [51] with sodium methoxide
solution under reflux gave (7-β-D-ribopyranosyl) theophylline [56] as white
crystals in 97.09% yield, with m.p. 250oC dec., and was characterized by IR
and UV spectral data.
The FT-IR spectrum (fig. 23) showed a stretching band at 3406cm-1
indicating the presence of hydroxyl groups.
The UV spectrum (fig. 24) showed absorption of λmax at 245 nm due to
the (π-π*) transition of dienone system (C=C-C=O) of the theophylline ring.
O
N
H
N
O
CH3
N
H
O
O
H
H
H
CH3
H
H
OBz
OBz
CH3
N
N
CH3ONa
Refl./InMeOH
OBz
[51]
H
N
O
H
H
H
HO
HO
N
N
O
CH3
H
OH
[56]
The 1H-NMR spectrum of [56] showed (fig. 25 and 26) a signal at δ 8.05
ppm which was assigned to H-6 proton (integrated for 1 H), a signal at δ 5.85
ppm was assigned to H-1/ proton (integrated for 1 H), a signal at δ 4.15 ppm
was assigned to H-3/ proton (integrated for 1 H), a signal at δ 4.05 ppm was
assigned to H-2/ proton (integrated for 1 H), a signal between δ 3.95-3.6 ppm
was assigned to H-4/ and 2H-5/ protons (integrated for 3 H), a signal between
75
Chapter three
Result & Discussion
δ 3.55-3.4 ppm was assigned to CH3-3 and 3H for –OH group protons
(integrated for 6 H) and a signal at δ 3.25 ppm was assigned to 3H-1 protons
(integrated for 3 H).
The 13C-NMR spectrum of [56] showed (fig. 27) a signal at δ 152.29 ppm
was assigned to C-2, a signal at δ 149.05 ppm was assigned to C-4, a signal at
δ 146.42 ppm was assigned to C-6, a signal at δ 138.91 ppm was assigned to
C-8 and C-9, a signal at δ 80.37 ppm was assigned to C/-1, a signal at δ 69.03
ppm was assigned to C/-5, a signal at δ 67.45 ppm was assigned to C/-2, a
signal at δ 64.04 ppm was assigned to C/-3, a signal at δ 63.02 ppm was
assigned to C/-4, a signal at δ 26.72 ppm was assigned to C-3 and a signal at δ
24.98 ppm was assigned to C-1.
76
Figure (23): IR Spectrum of compound [56]
Chapter three
Result & Discussion
77
Chapter three
Result & Discussion
Figure (15): UV Spectrum of compound [51]
Figure (24): UV Spectrum of compound [56]
78
Figure (25): 1H-NMR Spectrum of compound [56]
Chapter three
Result & Discussion
79
Figure (26): The expansion of 1H-NMR Spectrum of compound [56]
Chapter three
Result & Discussion
80
Figure (27): 13C-NMR Spectrum of compound [56]
Chapter three
Result & Discussion
81
Chapter three
Result & Discussion
Hydrolysis of the benzoate esters of [52], [53], [54] and [55] was
performed in the same manner which gave the expected products; [57], [58],
[59] and [60].
The FT-IR spectrum for [57], [58], [59] and [60] are presented in (fig. 28,
29, 31, 36) respectively.
The 1H-NMR spectrum of [58] (fig. 30) showed signals between δ 8.2 ppm
and δ 6.9 ppm were assigned to H-2, H-3, H-4, H-5, H-6, H-7, H-8, H-9
(aromatic protons) (integrated for 8 H), a signal at δ 6.65 ppm was assigned to
H-1/ proton (integrated for 1 H), a signal at δ 5.02 ppm was assigned to H-3/
proton (integrated for 1 H), a signal at δ 4.82 ppm was assigned to H-4/
proton (integrated for 1 H), a signal at δ 4.22 ppm was assigned to H-2/ proton
(integrated for 1 H), a signal between δ 3.6-3.15 ppm was assigned to 2H-5/
proton and 3 OH protons (integrated for 5 H).
The 1H-NMR spectrum of [59] showed (fig. 32) and (fig. 33) a signal at δ
7.85 ppm was assigned to H-7 and H-8 protons (integrated for 2 H), a signals
between δ 7.6 ppm and δ 6.9 ppm was assigned to H-2, H-3, H-4, H-5
(aromatic protons) (integrated for 4 H), a signal at δ 6.45 ppm was assigned to
H/-1 proton (integrated for 1 H), a signal at δ 4.8 ppm was assigned to H-3/
proton (integrated for 1 H), a signal at δ 4.7 ppm was assigned to H-4/ proton
(integrated for 1 H), a signal at δ 4.55 ppm was assigned to H-2/ proton
(integrated for 1 H), a signal at δ 4 ppm was assigned to 2H-5/ proton
(integrated for 2 H), a signal between δ 3.8-3 ppm was assigned to 3H for the
–OH group protons (integrated for 3 H).
The
13
C-NMR spectrum of (59) showed (fig. 34 and 35) a signal at δ
129.44 ppm was assigned to C-8, a signal at δ 127.5 ppm was assigned to C-9,
a signal at δ (121.6, 121.16, 120.74, 120.23, 119.97, 118.6) ppm was assigned
to C-2, C-3, C-4, C-5, C-6 and C-7, a signal at δ 79.65 ppm was assigned to
C/-1, a signal at δ 71.02 ppm was assigned to C/-5, a signal at δ 69.06 ppm
82
Chapter three
Result & Discussion
was assigned to C/-2, a signal at δ 67.26 ppm was assigned to C/-3, a signal at
δ 64.48 ppm was assigned to C/-4.
83
Figure (28): IR Spectrum of compound [57]
Chapter three
Result & Discussion
84
Figure (29): IR Spectrum of compound [58]
Chapter three
Result & Discussion
85
Figure (30): 1H-NMR Spectrum of compound [58]
Chapter three
Result & Discussion
86
Figure (31): IR Spectrum of compound [59]
Chapter three
Result & Discussion
87
Figure (32): 1H-NMR Spectrum of compound [59]
Chapter three
Result & Discussion
88
Figure (33): The expansion of 1H-NMR Spectrum of compound [59]
Chapter three
Result & Discussion
89
Figure (34): 13C-NMR Spectrum of compound [59]
Chapter three
Result & Discussion
90
Figure (35): The expansion of 13C-NMR Spectrum of compound [59]
Chapter three
Result & Discussion
91
Figure (36): IR Spectrum of compound [60]
Chapter three
Result & Discussion
92
Chapter three
Result & Discussion
3.2 Synthesis of Ribofuranonucleoside analogues
It is well known that while crystalline D-ribose exists in the pyranose form, it
appears to assume the furanose form with greater ease than the majority of sugars
and, indeed, occurs in nature largly, if not wholly, as furanose derivatives82. So In
this work attempts for the synthesis of different kind of ribofuranonucleoside
analogues have been admitted, using a modified Hilbert-Johnson procedure2e.
The synthetic strategy shown in scheme (6).
Modified Hilbert-Johnson procedure employed the peracylated carbohydrate
precursors instead of the highly reactive halosugar, along with a Lewis acid like
stanic chloride (SnCl4) and titanium chloride (TiCl4). Such a combination has
been used with mercuripurine derivatives2e.
The Lewis acid acts as a Friedel-Crafts catalyst like SnCl4 or TiCl4 which
activating the sugar by cleave the 1-O-acetyl bond to produse an electrophilic
sugar C1 cation103 [A] scheme (7), and therefore the thermodynamically formed
β-anomer is the only product2e.
3.2.1 Synthesis of 1-O-acetyl-2, 3, 5-tri-O-benzoyl-β-D-ribose [62]
Compound [62] is considered a common starting material used for the
synthesis of ribofuranonucleoside analogues.
In order to obtain [62], D-ribose was first converted to 1, 2, 3, 5-tetra-Obenzoyl-β-D-ribofuranose [61] to protect the hydroxyl groups at C-1, C-2, C-3
and C-5. 1, 2, 3, 5-tetra-O-benzoyl-β-D-ribofuranose [61] was prepared through
the benzoylation of D-ribose with benzoyl chloride, using pyridine as catalyst at
an elevated temperature to yield [62] in 34%. TLC of the reaction gave two spots
and according to Ness, Diehl and Fletcher82 this method formed a mixture of both
β-D-ribopyranose [41] and β-D-ribofuranose tetrabenzoate, this mixture consists
of 61.3% of [61] and 38.7% of [41]. On this bases the yield of furanose
tetrabenzoate is 21%-a yield never actually obtained because of the difficulty in
93
Chapter three
Result & Discussion
Ac
sulfuric acid
D-ribose
acetic anhydride/acetic acid
Ac
O
Ac
Ac
[63]
Het
condensation
N
2
SnCl4/ ClCH2CH2Cl
Hg
[46-50]
Het
N
Ac
O
Ac
Ac
[64-68]
protecting group
cleavage
Het
N
HO
O
OH
HO
[68-72]
O
N
Het
=
N
N
2
Hg
N
Hg
N
CH3
O
S
,
O
2
CH3
,
,
N
N
2
Hg
2
Hg
N
Hg
2
Scheme (6): Synthetic strategy.
94
,
N
Hg
2
Chapter three
Result & Discussion
AcO
O
OAc
OAc
AcO
O
SnCl4
O
OAc
OAc
O
SnCl4OAcCH3
[A]
[63]
Base
Base
AcO
O
OAc
OAc
Scheme 7: The action of SnCl4 in activating poly acetyl ribose.
95
Chapter three
Result & Discussion
H
BzO
phCOCl/py.
D-Ribose
OBz
O
H
H
98-102oC
H
OBz
OBz
[61]
O
H
H
H
+
OBz
H
H
OBz
OBz
OBz
[41]
Separating the mixture efficiently.
The ribobenzoate [61] was characterized by its IR spectrum. The FT-IR
spectrum (Fig. 37) showed stretching band at 3076cm-1 for aromatic (C-H),
2975cm-1 for aliphatic (C-H), 1724.2 cm-1 for (C=O) ester group and 1583.45 cm1
for (C=C) aromatic bands.
1-O-Acetyl-2, 3, 5-tri-O-benzoyl-β-D-ribose [62] was synthesized by
replacing the benzoyl attached to carbon 1 (anomeric) in [61] by acetyl group.
Such replacements at carbon 1 are accomplished by converting β-Dribofuranose tetrabenzoate [61] into 1-O-Acetyl-2, 3, 5-tri-O-benzoyl-β-Dribose [62] through the action of zink chloride in acetic anhydride to give [62]
as hexagonal micaceous plates.
The FT-IR spectrum of [62] (fig. 38) showed stretching bands at 3075cm-1
aromatic (C-H), 2990cm-1 alifatic (C-H), 1720cm-1 carbonyl (C=O) (of the
benzoate groups) and 1600.81cm-1 (C=C) aromatic bands. The TLC for the
reaction showed several spots and the melting point obtained was (105109oC) very far from the literature mp (129-130 oC) which shows that the
prepared 1-O-Acetyl-2, 3, 5-tri-O-benzoyl-β-D-ribose [62] was not pure and
according to Weygand and Wirth105 a mixture of 1-O-Acetyl-2, 3, 5-tri-Obenzoyl-β-D-ribose [62] and β-D-ribofuranose tetrabenzoate [61] was
obtained , so it was displaced by 1, 2, 3, 5-tetra-O-acetyl-β-D-ribofuranose84
[63] as a starting material.
96
Figure (37): IR Spectrum of compound [61]
Chapter three
Result & Discussion
97
Figure (38): IR Spectrum of compound [62]
Chapter three
Result & Discussion
98
Chapter three
Result & Discussion
BzO
BzO
OBz
O
H
H
OBz
H
OBz
Acetic anhydride
OAc
O
H
H
OBz
H
OBz
ZnCl2
[61]
[62]
3.2.2. Synthesis of 1, 2, 3, 5-tetra-O-acetyl-β-D-ribofuranose [63]
1, 2, 3, 5-tetra-O-acetyl-β-D-ribofuranose [63] could be used also as a
starting material for the synthesis of ribofuranonucleoside analogues,
compound [63] could be obtained in a pure form and in an easily crystallized
material, it was synthesized by treating D-ribose with sulfuric acid and acetic
acid, and using acetic anhydride as a solvent.
The FT-IR spectrum of [63] (fig. 39) showed stretching bands at 3075cm-1
aromatic (C-H), 2947cm-1 aliphatic (C-H), 1749 cm-1 carbonyl (C=O) (of the
acetate groups).
AcO
D-Ribose
sulfuric acid
OAc
O
H
H
OAc
H
OAc
acetic anhydride/acetic acid
[63]
3.2.3. Attempting for the synthesis of 7-(2/, 3/, 4/-Tri-0-acetyl-β-Dribofuranosyl) thiophylline [64]
In this experiment a dri mixture of 1, 2, 3, 5-tetra-O-acetyl-β-Dribofuranose [63] and Bis(theophylline-7-yl) mercury(II) [46] was treated
with SnCl4 in the present of celite and dichloroethan or acetonitrile as a
solvent in an elevated temperature, the TLC showed no proceeding of the
reaction and the FT-IR gave no absorptions between 1715-1650cm-1 for the
amide carbonyl group (fig 40). Two reasons may be advanced to explain this:
99
Chapter three
Result & Discussion
1- The catalyst is inactivated by traces of solvents in the sugar moiety like
ethanol or acetic acid104.
2- The present of traces of water is furthermore inactivating the Stannic
Chloride catalyst104.
The same method was used with different kind of nitrogen bases
derivatives [47-50] and The same above result were obtained.
AcO
O
H
OAc
OAc
H
H
OAc
theophillyne mercury salt
SnCl4 /CH2Cl2
[63]
100
(No reaction)
Figure (39): IR Spectrum of compound [63]
Chapter three
Result & Discussion
101
Figure (40): IR Spectrum for attempting of synthesis compound [64]
Chapter three
Result & Discussion
102
Chapter three
Result & Discussion
Conclusion
In this work it was established that the strategy which used the Keonigs-Knorr
method was useful for the preparation of the nucleoside analogues [56], [57],
[58], [59] and [60].
Future suggested work
a) Study of the biological activity for the prepared nucleoside analogues.
b) The inhibition effect of the prepared nucleoside analogues on ALP
marker in breast cancer patients.
c) Synthesis of new nucleoside analogues derived from the same nitrogen
bases: modification of the ribose moiety. The inhibition effect on ALP
may also be studded.
103
REFERENCES
References
1- Leven, P. A.; Jacobs, W. A. Ber. 1909, 42, 2474.
2- Kennedy, J. F. "Carbohydrate Chemistry", Birmingham; 1988; (a) p. 134 (b)
p. 135 (c) p. 177 (d) p. 154 (e) p. 149.
3- Hill, J. W.; Feigl, D. M.; Baum, S. J. "Chemistry and Life", forth ed., 1993,
(a) p. 614 (b) p. 610.
4- Voet, D.; Voet, J. G. "Biochemistry", 1990; (a) p. 409 (b) p. 740.
5- Robins, R. K.; Revankar, G. R. In Antiviral drug development. (Declerq, E,
and Walker, R. K., Eds.). Plenum, New York, 1988.
6- MacCoss, M.; Robins, M. J. In Chemistry of antitumour agents; Blackie and
son: UK (Wilman, D. E. V., ed.), 1990
7- Robins, R. K.; Kini, G. D. In Chemistry of antitumour agents, 1990.
8- Crooke, S. T.; Lebleu, B. Atisense research and application, CRC Press Inc.,
Baca Raton, FI. 1993.
9- Marry, R. K.; cranner, D. D.; Mayes, D. A.; Well, V. W. "Harper's
Biochemistry", 22nd ed, Appleton and large, Beirut, 1993; p. 340.
10-
Perigand, C.; Gosselin, G.; Imbach, J. L. Nucleosides and Nucleotides
1992, 11, 903.
11-
Wilman, D. E. "The chemistry of antitumour Agents", Chapman and
Hall, New Yourk; 1990; (a) p. 261 (b) p. 299.
12-
Harman, R. E.; Robins, R. K.; Townsend, L. B. "Chemistry and Biology
of Nucleosides and Nucleotides", Academic press, Inc: New Yourk,
1978; 98.
13-
Matsuda, A.; Takenuki, K.; Tanaka, M.; Sasaki, T.; Ueda, T. J. Med.
Chem. 1991, 34, 812.
14-
Okabe, M. M. Chem. Rev. 1992, 92,1745.
15-
Krayersky, A.; Watanabe, A. "Current Status and Perspectives
Bioniform", Mosco, 1993, p. 455.
16-
Balzarini, J.; McGuigan, C. Review, "Journal of Antimicrobial
Chemotherapy", 2002, 50, 5-9.
104
References
17-
Cooney, D.; Ahluwalia, G.; Mistsuya, H.; Fridl, A.; Hao, K. Z.; Dalal,
M.; Brooder, S.; Johns, D. Biochem, Pharmacol 1987, 36, 1763.
18-
Balzarini, J.; Pauwels, R.; Herdewijn, P.; Clercq, E. D.; Cooney, D.;
Kang, G.; Dalal, M.; Johns, D.; Broder, S. Biophys. Res. Comm. 1986,
140, 735.
19-
Herdwijn, P.; Balzarin, J. J. Med. Chem. 1987, 30, 1270.
20-
Chuand, C. J. Org. Chem. 1990, 55, 1418.
21-
Drorak, M. C.; Smee, M.; Mathews, T.; Verheyden, J. Antimicrob. Agent
and Chemotherapy 1983 23, 75.
22-
Dronk, M.; C. J. Med. Chem. 1983, 26, 75.
23-
Kumer, P.; Ohkura, K.; Balzarini, J.; DeClercq, E.; Seki, K.; Wiebe, L. I..
Nucleosides, nucleotides & Nucleic Acids 2004, Vol. 23, Nos. 1& 2, 729.
24-
Kohgo, S.; Yamada, K.; Kitano, K.; Iwai, Y.; Sakata, S.; Ashida, N,;
Hayakawa, H.; Nameki, D.; Kodama, E.; Matsuoka, M.; Hiroaki;
Mitsuya, H. O. Nucleosides, Nucleotides & Nucleic Acids 2004, Vol. 23,
No.4, 671-690.
25-
Gatzemeier, J.; Shepherd, Chevalier, F. A.; Weynants, P.; Cottier, B.;
Groen, H. J. M.; Rosso, R.; Mattson, K.; Cortes-Fune, H., Tonato, M.;
Burker, R. L.; Gottfried, M.; Voi, M. E.. J. Cancer 1996, 32, 234.
26-
Moore, M. Cancer 1996, 78, 633.
27-
Koshkin, A. A.; Fensholdt, J.; Pfundheller, H. M.; Lomholt,C. J. Org.
Chem. 2001, 66, No. 25, 8504-8512.
28-
Guianvarc'h, D.; Foutry, J.; Huu Dau, M. E. T.; Vincent Gu/erineau. J.
Org. Chem. 2002, 67, 3724-3732.
29-
Al-Tweigeri, F.; Nabholtz, J. M.; Macky, J. R. Cancer 1996, 78, 1359.
30-
Rang, H. R.; Dale, M. M.; Ritter, J. M. Pharmacology, 3rd ed., churchil
living stone, Inc., Ed in burgh, 1995; 696.
31-
Gupta, S. P. Chem. Rev. 1994, 94, 1507.
105
References
32-
Black, D. J.; Livimgston, R. B. Drugs 1990, 39, 489-501.
33-
Chabner, B. A. in Cancer and Chemotherapy, Crooke, S. T.; Prestayko,
A. W., Eds; Academic Press: New York, 1981, Vol. 3, PP. 3-24.
34-
Prath, W. B.; Ruddon, R. W. , Eds, The Anticancer Drugs, Oxford
University Press: New York, 1979.
35-
Grunewald, R.; Abbruzzese, J. L.; Tarassoff, P.; Plunkett, W. Cancer
Chemother. Pharmacol. 1991, 27, 258-262.
36-
Baker, C. H. ; Banzon, J. ; Bollinger, J. M. ; Stubbe, J. J. Med.
Chem.1991, 34, 1879-1884.
37-
Yamagami, K. ; Fujii, A. ; Arita, M. ; Okumoto, T. ; Sakata, S. ;
Matsuda, A. ; Ueda, T. Cancer Res. 1991, 51, 2319-2323.
38-
Haslett, C.; Chilvers, E. R.; Boon, N. A.; Colledge, N. R. Principles and
Practice of Medicine, Hunter, J. A. A. Ed. 19th edition 2002,p 110.
39-
Prusoff, W. H. ; Lin, T. S. ; Zucker, M. Antiviral Res. 1986, 6, 311-328.
40-
Shannon, W. M. in Antiviral Agents and Viral Diseases of Man; Galasso.
G. J. , Ed. ; Raven Press: New York, 1984; Chapter 3, PP. 55-121.
41-
Robis, R. K. ; Revenkar, G. R. in antiviral Drug Development. A
Multidisciplinary Approach; De Clercq, E.; Walker, R. T., Eds; Nato
Advanced Institutes Series. Series A: Life Sciences; Plenum Press: New
York, 1987; Vol. 143, PP. 11-36.
42-
De Clercq, E. in Advances in Drug Research; Testa, B., Ed.; Academic
Press LTD: London, 1988, Vol. 17, PP. 1-59.
43-
Pavan-Lanagston, D.; Buchanan, R. A. ; Alford, C. A. Adenine
Arabinoside: An Antiviral Agent , Eds; Raven-Press:New York, 1975.
44-
De Clercq, E.; Walker, R. T. Ed.; in Targets for the Design of Antiviral
Agents; Nato Advanced Institutes Series, Series A: Life Saences; Plenum
Press:New York, 1983, Vol. 73, PP. 203-230.
45-
Poiez, B. J. ; Ruscetti, F. W. ; Reitz, M. S. ; Kalyanamaran, V. S. ; Gallo,
R. C. Nature 1981, 294, 268-271.
106
References
46-
Barre-Sinoussi, F., Charmann, J. C. ; Rey, R.; Nugeyre, M. T. ;
Chamaret, S. ; Gruest, J. ; Dauguest, C. ; Axler-Blin, C.; Vezinet-Brun, F.
; Rouzioux, C. ; Rozenbaum, W. ; Montagnier, L. Science 1983, 220,
868-871.
47-
Gallo, R. C. ; Salahuddin, S. Z. ; Popvic, M. ;Shearev, G. M.; Kaplan, M.
; Haynes, B. F. ; Palker, T. J. ; Red Field, R. ; Olesky. J.; Safai, B. ;
White, G. ; Foster, P. ; Markham, P. D. Science 1984, 244, 500-504.
48-
Herdewijn, P.; De Clercq, E. in Design of Anti-AIDS Drugs; De
Clercq, E., Ed.; Elsevier; Amsterdam, 1990, Vol. 14, PP. 141-174.
49-
Sarin, P. S. Ann. Rev. Pharmacol. 1988, 28, 411-128.
50-
Suhadolnick, R. J. Nucleoside Antibiotics; Ed. ; Wiley-Interscience: New
York, 1970.
51-
Suhadolnick, R. J. in Progress in Nucleic Acid Research and Molecular
Biology, Cohn, W. E. , Ed. ; Academic Press: New York, 1979, Vol. 22,
PP. 193-291.
52-
Mistuya, H.; Yarohoam, R. J. Science 1990, 24, 1533.
53-
Hammer, C. L., Theses, SWISS FEDERAL INSTITUTE OF
TECHNOLOGY, ZURICH, 1997.
54-
Postema, M. H. D. Tetrahedron 1992, 40, 8545-8599.
55-
Von Kroigk, U.; Benner, S. A. J. Am. Chem. Soc. 1995, 117, 5361-5362.
56-
Michelson, A. M.; The Chemistry of
Nucleosides and Nucleotides;
Academic Press; London, 1963, chapter 2.
57-
Dekker, C. A.; Goodman, L.; The Carbohydrates; Academic Press; New
York, 1970, Vol. 2A, Page 1.
58-
Goodman, L.; Basic Principles in Nucleic Acid Chemistry; Academic
Press; New York, 1974, Vol. 1, Page 93.
59-
Walker, R. T.; Comprehensive Organic Chemistry; Pergamon Press;
Oxford, 1979, Vol. 5, Page 53-104.
60-
Davoll, J.; Lythgoe, B.; Todd, A. R. J. Chem. Soc. 1948, 967.
107
References
61-
Davoll, J. ;Lowy, B. A. J. Am. Chem. Soc. 1951, 73, 1650.
62-
Cass, C. E.; Lepage, W. D. J. Med. Chem. 1979, 22, 518.
63-
Wittenburg, E. Z. Chemic, Lpz, 1964, 4, 303.
64-
Nishimura, T.; Shimizu, B.; Iwai, I. Chem. Pharm. Bull, Tokyo, 1964,
12, 1471.
65-
Birkofer, L.; Ritter, A.; Kuhlthau, H.-P. Chem. Ber. 1964, 97, 934.
66-
Shimizu, B.; Saito, A. Agric. Biol. Chem. 1969, 33. 119.
67-
Sato, T. In Synthetic Procedures in Nucleic Acid Chemistry; Zorbach, W.
W., Tipson. R. S., Eds.; Wiley-Intersci:New York, 1968, P. 264.
68-
Diekmann, E.; Friedrich, K.; Fritz, H.-G. J. Prakt. Chem. 1993, 335, 415.
69-
Vorbruggen, H.; Rutt-PHLENZ, C. Syntheses of Nucleosides, 2000, P. 6.
70-
Lerch, U.; Burdon, M. G.; Moffatt, J. G. J. Org. Chem. 1971, 36, 1507.
71-
Brown, D. M.; Burdon, M. G.; Slatcher, R. P. J. Chem. Soc.; 1968, C,
1051.
72-
Ohrui, H.; Jones, G. H.; Moffatt, J. G.; Maddox, M. L.; Christensen, A.
T.; Byram, S. K. J. Am. Chem. Soc. 1975, 97, 4602.
73-
Cousineau, T. J. secrist III, J. A. J. Org. Chem. 1979, 44, 4351.
74-
Jeanloz,R.; Fletcher, H. G., JR.; Hudson, C. S. J. Am. Chem. Soc. 1948,
70, 4052-4054.
75-
Fletcher, H. G., JR.; Ness, R. K. J. Am. Chem. Soc. 1955, 77, 5337-5340.
76-
Bernthsen, A. Bur-Chem. Ges. 1883, 16, 2896.
77-
Bernthsen, A. Ber. 1887, 20, 942.
78-
Mann, F. G.; Bernard, C. S. PRACTICAL ORGANIC CHEMISTRY;
third edition, 1952.
79-
Freestone, A. G.; Hough, F. L.; Richaredson, A. C. Carbohydrate Res.
1973, 82, 387.
80-
Daroll, J.; Lowy, B. A. J. Am. Chem. Soc. 1951, 73, 1650-1655.
81-
Yung, N.; Fox, J. J. Methods in Carbohydrate Chemistry 1963, Volume
II, 108-112.
108
References
82-
Ness, R. K.; Diehl, H. W.; Fletcher, H. G. J. Am. Chem. Soc. 1954, 5,
763-767.
83-
Ness, R. K.; Fletcher, H. G. J. Am. Chem. Soc. 1954, 20, 1663-1667.
84-
Guthrie, R. D.; Smith, S. C. Chem. Ind. (London) 1968, 27, 547-548.
85-
Hassan S. El Khadem. Carbohydrate Chemistry Monosaccharides and
their Oligomers, 1988 sixth Avenue a.p 138, b. p 139.
86-
Ness, R. K.; Fletcher, H. G.; JR.; Hudson, C.S. J. Am. Chem. Soc. 1951,
73, 959-963.
87-
Jeanloz, R. W.; Fletcher, H. G.; JR. Advances in carbohydrate Chem.
1951, 6, 135.
88-
Ness, R. K.; Fletcher, H. G. J. Am. Chem. Soc. 1956, 78, 4710.
89-
Bernthsen, A. Bur-Chem. Ges. 1883, 16, 2896.
90-
Randal, F.; Dangl, F. Infrared Determination of Organic Chemistry,
D. Van Naustrand Co., New York, 1949, 95.
91-
Bernthsen, A. Ber. 1887, 942, 20.
92-
Vanderhaeghe, H. J. Org. Chem. 1960, 25, 747.
93-
Montgomery, J. A.; Thomas, H. J. J. Org. Chem. 1966, 31, 1411.
94-
Bühler,E.; Pfliderer, W. Angew. Chem. Inter. Ed. 1964, 90, 638.
95-
Fang, W.; Jie, T. Acta Pharmacol. Sin. 2003 Oct, 24(10), 1001-1005.
96-
Alwan, A. A. S.; Abou, Y. Z. IRAQI DRUG GUIDE, First edition, 1990,
71.
97-
Al-Mashkur, I. M. theses, college of Science University of Baghdad,
2003.
98-
WilliAMS, J. D.; Drach, J. C.; Townsend, L. B. NUCLEOSIDES,
NUCLEOTIDES & Nucleic acids 2004, 23, No. 5, 805-812.
99-
Chen, J.J.; Wei, Y.; Drach, J.C.; Townsend. L.B. J. Med. Chem. 2000,
43, 2449-2456.
100- Lemieux, R. U. Advances in carbohydrate chemistry 1954, 9, 1.
101- Fox, J. J.; Wempen, I. Advances in Carbohydrate Chem. 1959, 14, 283.
109
References
102- Jeanloz, R. W.; Fletcher, H. G. Jr. Advances in Carbohydrate Chem.
1951, 6, 135.
103- Niedballa, U.; Vorbrüggen, H. J. Org. Chem. 1974, 39, 3664-3667.
104- Niedballa, U.; Vorbrüggen, H. J. Org. Chem. 1974, 39, 3554-3660.
105- Weygand, F.; Wirth, F. Chem. Ber. 1952, 85, 1000.
110
‫ﻭﺯﺍﺭﺓ ﺍﻟﺘﻌﻠﻴﻢ ﺍﻟﻌﺎﻟﻲ ﻭﺍﻟﺒﺤﺚ ﺍﻟﻌﻠﻤﻲ‬
‫ﺟﺎﻣﻌﺔ ﺑﻐﺪﺍﺩ‬
‫ﻛﻠﻴﺔ ﺍﻟﻌﻠﻮﻡ‬
‫ﻗﺴﻢ ﺍﻟﻜﻴﻤﻴﺎء‬
‫ﺗﺤﻀﻴﺮ ﻣﻤﺎﺛﻼت اﻟﻨﻴﻮﻛﻠﻴﻮﺳﻴﺪ ﺗﺤﺘﻮي ﻋﻠﻰ اﻧﻮاع ﺟﺪﻳﺪة‬
‫ﻣﻦ اﻟﻘﻮاﻋﺪ اﻟﻨﻴﺘﺮوﺟﻴﻨﻴﺔ‬
‫ﺭﺳﺎﻟﺔ ﻣﻘﺪﻣﺔ ﺇﻟﻰ ﻛﻠﻴﺔ ﺍﻟﻌﻠﻮﻡ – ﺟﺎﻣﻌﺔ ﺑﻐﺪﺍﺩ‬
‫ﻛﺠﺰء ﻣﻦ ﻣﺘﻄﻠﺒﺎﺕ ﻧﻴﻞ ﺩﺭﺟﺔ ﺍﻟﻤﺎﺟﺴﺘﻴﺮ‬
‫ﻓﻲ ﻋﻠﻮﻡ ﻛﻴﻤﻴﺎء‪/‬ﺍﻟﻜﻴﻤﻴﺎء ﺍﻟﻌﻀﻮﻳﺔ‬
‫ﻣﻘﺪﻣﺔ ﻣﻦ ﻗﺒﻞ‬
‫ﺃﺳﻤﺎء ﻣﺎﺯﻥ ﻋﺒﺪ ﺍﻟﺤﻤﻴﺪ ﻛﺎﻅﻢ ﺍﻟﺴﺎﻣﺮﺍﺋﻲ‬
‫ﺑﻜﺎﻟﻮﺭﻳﻮﺱ ﻋﻠﻮﻡ ﻛﻴﻤﻴﺎء‬
‫)ﺟﺎﻣﻌﺔ ﺑﻐﺪﺍﺩ ‪(۱۹۹۹‬‬
‫ﺑﺈﺷﺮﺍﻑ‬
‫ﺍﻷﺳﺘﺎﺫ ﺍﻟﺪﻛﺘﻮﺭ ﻳﻮﺳﻒ ﻋﻠﻲ ﺍﻟﻔﺘﺎﺣﻲ ﻭ ﺍﻷﺳﺘﺎﺫﺓ ﺳﻌﺎﺩ ﺍﻻﻋﺮﺟﻲ‬
‫ﺣﺰﻳﺮﺍﻥ ‪ ۲۰۰٥‬ﻡ‬
‫ﺟﻤﺎﺩﻱ ﺍﻷﻭﻝ ‪۱٤۲٦‬ﻫـ‬
‫اﻹﻫﺪاء‬
‫إﻟﻰ ﻣﺜﻠﻲ اﻷﻋﻠﻰ ‪ ...‬واﻟﺪي اﻟﻐﺎﻟﻲ‬
‫إﻟﻰ ﻗﺮة ﻋﻴﻨﻲ‪ ...‬واﻟﺪﺗﻲ اﻟﺤﻨﻮﻧﺔ‬
‫إﻟﻰ ﻣﻦ ﻛﺎن ﺧﻴﺮ ﺳﻨﺪ ﻟﻲ ‪ ...‬زوﺟﻲ اﻟﺤﺒﻴﺐ‬
‫إﻟﻰ ﻣﻦ وﻗﻔﻮا داﺋﻤﺎ ﺑﺠﺎﻧﺒﻲ ‪ ...‬ﻋﺎﺋﻠﺔ زوﺟﻲ‬
‫إﻟﻰ أﺣﺒﺎﺋﻲ ‪ ...‬اﺧﻮاﺗﻲ‬
‫وﻣﻦ اﷲ اﻟﺘﻮﻓﻴﻖ‬
‫أﺳﻤﺎء ﻣﺎزن‬
‫ﺍﻟﺧﻼﺻﺔ‬
‫ﻟﻘ���ﺩ ﺗﺿ���ﻣﻥ ﺍﻟﻌﻣ���ﻝ ﻓ���ﻲ ﻫ���ﺫﻩ ﺍﻻﻁﺭﻭﺣ���ﻪ ﺛﻼﺛ���ﺔ ﺃﺟ���ﺯﺍء‪ ،‬ﻳﻌﻧ���ﻰ ﺍﻟﺟ���ﺯء ﺍﻻﻭﻝ "ﺍﻟﻣﻘﺩﻣ���ﺔ" ﺑﻛﻳﻣﻳ���ﺎء‬
‫ﺍﻟﻧﻳﻭﻛﻠﻳﻭﺳ��ﻳﺩﺍﺕ ﻭﺍﻟ��ﺫﻱ ﻳﺗﺿ��ﻣﻥ ﺑﻧﺎﺋﻬ��ﺎ ﺍﻟﻔﺭﺍﻏ��ﻲ‪ ،‬ﺍﻧﻭﺍﻋﻬ��ﺎ‪ ،‬ﻣﺳ��ﺢ ﻭﺍﺳ��ﻊ ﻟﻔﻌﺎﻟﻳﺗﻬ��ﺎ ﺍﻟﺑﻳﻭﻟﻭﺟﻳ��ﺔ ﻛﻣﺿ��ﺎﺩﺍﺕ‬
‫ﻟﻠﺳﺭﻁﺎﻥ ﻭﺍﻟﻔﻳﺭﻭﺳﺎﺕ ﻭﺍﻟﺑﻛﺗﻳﺭﻳﺎ ﺇﺿﺎﻓﺔ ﺇﻟﻰ ﻁﺭﻕ ﺗﺣﺿﻳﺭﻫﺎ‪.‬‬
‫ﻳﺗﺿ���ﻣﻥ ﺍﻟﺟ���ﺯء ﺍﻟﺛ���ﺎﻧﻲ ﺗﺣﺿ���ﻳﺭﺍﻧﻭﺍﻉ ﺟﺩﻳ���ﺩﺓ ﻣ���ﻥ ﻣﻣ���ﺎﺛﻼﺕ ﺍﻟﻧﻳﻭﻛﻠﻳﻭﺳ���ﻳﺩﺍﺕ ﺗﺣﺗ���ﻭﻱ ﻋﻠ���ﻰ ﻗﻭﺍﻋ���ﺩ‬
‫ﻧﻳﺗﺭﻭﺟﻳﻧﻳﺔ ﺟﺩﻳﺩﺓ ﻟﺳﻛﺭ ﺍﻝ ‪-D‬ﺭﺍﻳﺑﻭﺯ‪ .‬ﻳﺗﺿﻣﻥ ﺍﻟﺟﺯء ﺍﻟﻌﻣﻠﻲ ﺗﺣﺿﻳﺭ ﻣﺭﻛﺏ ‪ -٤ ،۳ ،۲ ،۱‬ﺗﺗﺭﺍ‪– O -‬‬
‫ﺑﻧﺯﻭﺍﺕ – ‪ – D -β‬ﺭﺍﻳﺑﻭﺑﻳﺭﺍﻧﻭﺯ ]‪ [٤۱‬ﻛﻣ�ﺎﺩﺓ ﺍﻭﻟﻳ�ﺔ ﺣﻳ�ﺙ ﺍﻣﻛ�ﻥ ﺍﻟﺣﺻ�ﻭﻝ ﻋﻠ�ﻰ ]‪ [٤۱‬ﻣ�ﻥ ﺗﻔﺎﻋ�ﻝ ‪-D‬‬
‫ﺭﺍﻳﺑﻭﺯ ﺍﻟﻼﻣﺎﺋﻲ ﻣﻊ ﻛﻠﻭﺭﻳﺩ ﺍﻟﺑﻧﺯﻭﻳﻝ ﺑﻭﺟﻭﺩ ﺍﻟﺑﺭﺩﻳﻥ‪ .‬ﻋﻧ�ﺩ ﻣﻌﺎﻣﻠ�ﺔ ]‪ [٤۱‬ﻣ�ﻊ )‪ (HBr‬ﺗ�ﻡ ﺍﻟﺣﺻ�ﻭﻝ ﻋﻠ�ﻰ‬
‫ﺑﺭﻭﻣﻳ���ﺩ ‪ ،٤ ،۳ ،۲ ،۱‬ﺗ���ﺭﺍﻱ‪-O-‬ﺑﻧ���ﺯﻭﺍﺕ‪-D-β-‬ﺭﺍﻳﺑﻭﺑﻳﺭﺍﻧ���ﻭﺯ ]‪ [٤۲‬ﻣ���ﻊ ﺍﻧ���ﻭﺍﻉ ﻣﺧﺗﻠﻔ���ﺔ ﻣ���ﻥ ﺍﻟﻘﻭﺍﻋ���ﺩ‬
‫ﺍﻟﻧﻳﺗﺭﻭﺟﻳﻧﻳ�ﺔ )ﺛﻳ��ﻭﻓﻠﻳﻥ‪ ،‬ﻓﻳﻧﻭﺛﺎﻳ��ﺎﺯﻳﻥ‪ ،‬ﻓﻳﻧﻭﻛﺳ��ﺎﺯﻳﻥ‪ ،‬ﺍﻧ��ﺩﻭﻝ ﻭﻛﺎﺭﺑ��ﺎﺯﻭﻝ( ﺑﻌ��ﺩ ﺗﺣﻭﻳﻠﻬ��ﺎ ﺍﻟ��ﻰ ﺍﻣ��ﻼﺡ ﺍﻟﺯﺋﺑ��ﻕ‬
‫ﺍﻟﺛﻧ����ﺎﺋﻲ ﺍﻟﻣﻘﺎﺑﻠ����ﺔ ]‪ [٤۹] ،[٤۸] ،[٤۷] ،[٤٦‬ﻭ ]‪ [٥۰‬ﻋﻠ����ﻰ ﺍﻟﺗﺭﺗﻳ����ﺏ‪ ،‬ﻭﺑﺎﻋﺗﻣ����ﺎﺩ ﻁﺭﻳﻘ����ﺔ ‪(Keonigs-‬‬
‫)‪ Knorr‬ﺗﻡ ﺍﻟﺣﺻﻭﻝ ﻋﻠﻰ ﻣﺷﺗﻘﺎﺕ ﻣﺗﻣﺎﺛﻼﺕ ﺍﻟﻧﻳﻭﻛﻠﻳﻭﺳﻳﺩﺍﺕ ]‪ [٥٤] ،[٥۳] ،[٥۲] ،[٥۱‬ﻭ]‪.[٥٥‬‬
‫ﺍﺩﻯ ﺍﻟﺗﺣﻠ���ﻝ ﺍﻟﻣ���ﺎﺋﻲ ﺍﻟﻘﺎﻋ���ﺩﻱ ﻟﻣﺟﻣﻭﻋ���ﺔ ﺍﻟﺑﻧ���ﺯﻭﺍﺕ ﻝ]‪ [٥٤] ،[٥۳] ،[٥۲] ،[٥۱‬ﻭ ]‪ [٥٥‬ﺑﺎﺳ���ﺗﺧﺩﺍﻡ‬
‫ﻣﻳﺛﻭﻛﺳﻳﺩ ﺍﻟﺻ�ﻭﺩﻳﻭﻡ ﺍﻟ�ﻰ ﺍﻟﺣﺻ�ﻭﻝ ﻋﻠ�ﻰ ﻣﻣ�ﺎﺛﻼﺕ ﺍﻟﻧﻳﻭﻛﻠﻳﻭﺳ�ﻳﺩﺍﺕ ﺍﻟﺣ�ﺭﺓ ]‪ [٥۹] ،[٥۸] ،[٥۷] ،[٥٦‬ﻭ‬
‫]‪.[٦۰‬‬
‫ﺍﻣﺎ ﺍﻟﺟﺯء ﺍﻟﺛﺎﻟﺙ ﻣﻥ ﺍﻟﻌﻣﻝ ﻓﻘﺩ ﺗﺿﻣﻥ ﻣﻧﺎﻗﺷﺔ ﻧﺗﺎﺋﺞ ﺍﻟﻘﻳﺎﺳﺎﺕ ﺍﻟﻁﻳﻔﻳﺔ ﻟﻠﻣﺭﻛﺑﺎﺕ ﺍﻋﻼﻩ ﺑﺎﺳﺗﺧﺩﺍﻡ ‪،FT-IR‬‬
‫‪1H-NMR ،UV‬ﻭ ‪.13C-NMR‬ﻭﻗﺩ ﺩﻟﺕ ﺍﻟﻧﺗﺎﺋﺞ ﺍﻟﻧﻬﺎﺋﻳﻪ ﺍﻟﻣﺳﺗﺣﺻﻠﻪ ﻋﻠﻰ ﺻﺣﺔ ﺍﻟﺗﺭﺍﻛﻳﺏ ﺍﻟﻣﻘﺗﺭﺣﻬﺯ‬
‫ﺻﺪق اﷲ اﻟﻌﻈﻴﻢ‬
‫)ﺳﻮﺭﺓ ﺍﻻﻧﻌﺎﻡ ‪ ،‬ﺍﻵﻳﺔ ‪(٥۹‬‬