143
Molybdopterin-Modeling: The Synthesis of Pterin Dithiolene Ligands I n a u g u r a l d i s s e r t a t i o n zur Erlangung des akademischen Grades eines Doktors der Naturwissenschaften (Dr. rer. nat.) der Mathematisch-Naturwissenschaftlichen Fakultät der Universität Greifswald vorgelegt von Ivan Trentin Greifswald 15. April 2019

epub.ub.uni-greifswald.de · Table of contents 1. Pteridines and pterins

  • Upload
    others

  • View
    4

  • Download
    0

Embed Size (px)

Citation preview

Page 1: epub.ub.uni-greifswald.de · Table of contents 1. Pteridines and pterins

Molybdopterin-Modeling: The Synthesis of Pterin Dithiolene Ligands

I n a u g u r a l d i s s e r t a t i o n

zur

Erlangung des akademischen Grades eines

Doktors der Naturwissenschaften (Dr. rer. nat.)

der

Mathematisch-Naturwissenschaftlichen Fakultät

der

Universität Greifswald vorgelegt von Ivan Trentin

Greifswald 15. April 2019

Page 2: epub.ub.uni-greifswald.de · Table of contents 1. Pteridines and pterins

Dekan: Prof. Dr. Werner Weitschies 1. Gutachter : Prof. Dr. Carola Schulzke 2. Gutachter: Prof. Dr. Albrecht Berkessel Tag der Promotion: 15. April 2019

Page 3: epub.ub.uni-greifswald.de · Table of contents 1. Pteridines and pterins

Table of contents 1. Pteridines and pterins ................................................................................................................................... 1

1.1 Historical background .............................................................................................................................. 1

1.2 Nomenclature .......................................................................................................................................... 5

1.3 Pteridines chemistry ................................................................................................................................ 6

1.3.1 Aromaticity, π-excessive and π-deficient .......................................................................................... 6

1.3.2 Stability of pteridines and pterins ..................................................................................................... 8

1.3.3 Covalent hydration reaction ............................................................................................................. 9

1.3.4 Reactivity of pteridine ..................................................................................................................... 11

2. Biological Background ................................................................................................................................ 15

2.1 Molybdenum dependent enzymes ......................................................................................................... 15

2.2 Molybdenum cofactor natural synthesis and maturation ..................................................................... 18

2.2.1 Cyclization of GTP ........................................................................................................................... 18

2.2.2 Sulfuration of cPMP ........................................................................................................................ 19

2.2.3 Molybdenum uptake ....................................................................................................................... 20

2.2.4 Insertion of the metal ..................................................................................................................... 21

2.2.5 Cofactor insertion into apoenzyme ................................................................................................. 22

2.3 Molybdenum cofactor and Human diseases-MoCD and ISOD .............................................................. 24

2.3.1 Synthetic cofactor, drug for a possible cure ................................................................................... 25

3. Results and discussion ................................................................................................................................ 26

3.1 Modelling molybdopterin (MPT)............................................................................................................ 26

3.2 Protection of Pterins .............................................................................................................................. 31

3.3 First attempt for the synthesis of pterin-dithiolene ligand, preparation of 6-acyl ................................ 35

pterins by condensation .............................................................................................................................. 35

3.4 Synthesis of pterin-dithiolene ligands through the “Minisci reaction” .................................................. 57

3.5 Acylation protocol variations, targeting MPT ....................................................................................... 81

3.5.1 Pyran ring variation ........................................................................................................................ 81

3.5.2 Phosphate variation, northern and southern functionalization ..................................................... 94

4 .Conclusion ................................................................................................................................................... 95

Experimental section ...................................................................................................................................... 98

IR and UV-VIS spectra ................................................................................................................................... 115

Molecular structures and X-ray data ........................................................................................................... 124

References ..................................................................................................................................................... 131

Eigenständigkeitserklärung .......................................................................................................................... 135

Lebenslauf - Ivan Trentin .............................................................................................................................. 136

Acknowledgement ........................................................................................................................................ 139

Page 4: epub.ub.uni-greifswald.de · Table of contents 1. Pteridines and pterins

«The period of fifteen years of investigation in two laboratories on the structure of the butterfly

pigments indicates already some principal difficulties encountered with pteridines which are

attributed to their incomplete combustibility in elemental analysis, their high melting point and

decomposition point, their poor solubilities in water and most organic solvent and hence problem

purification.

Nevertheless the natural pteridines reveal so many exceptional physical and extraordinary chemical

properties that we cannot only learn basic principles of heterocyclic chemistry, in general, but also

get detailed knowledge and under-standing why special structures are prerequisite for specific

biochemical and enzymatic reaction.

The fascination to discover the unexpected and anomaly is one of the reasons why I have devoted

already forty years of research efforts to a large extent to pteridine chemistry».

“Chemistry and Biology of Pteridines and Folates

Natural Pteridines – A Chemical Hobby”

Prof. Dr. Wolfgang Eugen Pfleiderer

Page 5: epub.ub.uni-greifswald.de · Table of contents 1. Pteridines and pterins

1

1. Pteridines and pterins

1.1 Historical background

The interesting story of the pteridines compounds family has been described in different books and by different

scientists all around the word. The best narrative remains the one written by the main characters of this tale who could

directly have influenced the development of this attractive field of research. The reading of books like “Chemistry and

Biology of Pteridine and Folates” or “Fused Pyrimidines” offers not only amazing reports of experimental data but mainly

the ideas and passion of the authors and the researchers [1]. Therefore, if we want to fully describe the story of

pteridines, we can’t but emphasize the devotion of the researchers and the spirit of a topic that W. Pfleiderer defined

as a “chemical hobby“ [2].

The development of pteridine research can be ascribed to the organic chemistry of the second half of the 19th century.

The first publications came from two different fields — synthetical chemistry and isolation of natural products. It is

interesting to observe how different scientists were able to isolate some products but due to the particular chemical

features of pteridines, the complete elucidations of the structures were obtained only many years later at the beginning

of the 20th century. Exemplary are the publications of F. Wöhler [3] and H. Hlasiwetz [4] in 1857, which describe the

formation of a yellow product by heating uric acid in water; eighty-five years later in 1942 F. G. Hopkins repeated the

experiment and suggested the formation of a compound analogous to the wings’ pigments of the sulfur-yellow butterfly

“Pieridae” [5]. The final elucidation of the reaction was achieved only in 1959 by W. Pfleiderer [6] with the isolation and

characterization of twelve different substances of which six are main products, represented in Figure 1.

FIGURE 1 – FORMATION OF PTERIDINES FROM URIC ACID DESCRIBED BY W. PFLEIDERER : 2,4,7-TRIHYDROXY PTERIDINE (I); 2,4,7-TRIHYDROXY-6-METHYL PTERIDINE (II); 2,4,6-TRIHYDROXY PTERIDINE (III); 2,4,7-TRIHYDROXY-6- PTERIDINECARBOXYLIC ACID (IV); 2,4,6,8-TETRAHYDROXY

PYRIMIDO-[4.5-G]-PTERIDIN (V); 2,4,5,7-TETRAHYDROXY-PYRIMIDO[5.4-G]- PTERIDIN (VI)

Page 6: epub.ub.uni-greifswald.de · Table of contents 1. Pteridines and pterins

2

The summary of the first directed synthesis of pteridines does not disclose the difficulties faced by the pioneers of this

field (Scheme 1); in fact, despite the development of the organic chemistry between the end of the 19th and beginning

of the 20th century, the analysis of particular compounds like pteridines represented a tripping stone for the

researchers.

The first rational protocol to obtain a pteridine was reported by O. Kühling [7] in 1894. He synthesized through an

oxidation and decarboxylation of alloxazine a 2,4(1H,3H)-pteridinedione named alloxazin. In 1907 S. Gabriel and A. Sonn

[8] in Berlin obtained the same product of O. Kühling starting from 2,3-pyrazinedicarboxyamide and in 1937 R. Kuhn

with A. Cook [9] prepared the same pteridine, this time condensing a 2,4-dihydroxy-5,6-diaminopyrimidine with a

glyoxal molecule and changing its name from alloxazin to lumazine (Scheme 1).

SCHEME 1 – FIRST SYNTHESIS OF PTERIDINES FROM THE BOOK “FUSED PYRIMIDINES”

In 1901 S. Gabriel with J. Colman prepared the first pteridine from a pyrimidine ring, condensing a 4,5-

pyrimidinediamine with diphenylglyoxal and calling the final product “das Azin” (6,7-diphenyl-pteridine) [10]. After 5

years in the same laboratory the same reaction was curiously described and it was used for the synthesis of various

“Azin-purin” derivatives [10, 11] (Scheme 1).

Despite the analytical issues faced by the synthesis of pteridines, at the beginning of the 20th century the basic protocols

of this chemistry were already delineated; many research groups started to work on this topic and the number of

respective publications increased. Still the main authors of this field refer the origins of pteridine chemistry to the

Page 7: epub.ub.uni-greifswald.de · Table of contents 1. Pteridines and pterins

3

natural products; it is in fact the isolation of the natural occurring pterins which had the largest impact on the scientific

community.

Surely the first and most representative author in this field is F. G. Hopkins, who between 1889 and 1895 described the

isolation of two rich fractions of butterfly pigments – the first from the English brimstone butterfly and the second from

the white cabbage butterfly (Figure 2) [12]. However, as other contemporary chemists, he did not manage to give any

structural elucidations, mainly due to the unusual physical and chemical properties of the substances, like poor solubility

and high melting point.

In 1924 C. Schöpf, a young butterfly collector and junior professor in Freiburg, persuaded his professor H. Wieland to

work on the publication of Hopkins and tried to identify the structure of the interesting pigments. The two scientist

worked many years on the characterization of the molecules isolated by Hopkins, they named the class of pigments

“pterins” referring to their origin (from Greek pteron = wing) and the two substances according to their colouration –

xanthopterin (yellow) and leucopterin (white, colourless). Two years later they managed to isolate a third

isoxanthopterin pigment but unfortunately the composition of all the pigments remained unknown until 1940 when R.

Purrmann, one of Wieland’s PhD student, was able to elucidate the structure of the three pigments represented in

Figure 2 [13-16].

FIGURE 2 – PIGMENTS ISOLATED BY C. SCHÖPF AND H. WIELAND

Representative of many years of research is the opening address to the third Pteridine Symposium in Stuttgart in 1962,

when C. Schöpf described the practical and chemical difficulties encountered in almost fifteen years of research:

«However, I want to call attention to a most paradoxical situations. The chemistry of those pyrimidopyrazines, which to-

day we call pteridines, had been very successfully investigated in Berlin from 1884 onwards; in Munich, it was not realized

that the butterfly pigments belonged to this family of compounds until 1940, nearly half a century later. Why did natural

products cause so many difficulties? Collection was fitful, because of the season, and the material precious. The lack of

melting points removed an important criterion for purity. Not until 1940 was xanthopterin freed from an impurity that

gave a red colour with hydrogen peroxide. The poor solubility in all common solvents diminished ease of purification and

no molecular weights were obtainable. The resistance to complete combustion, leading to nitrogenous coke, produced

errors in the analytical results. Particularly misleading was the supposed analogy with purines based on the fact that

pterins and purines gave similar substances upon degradation» [17].

Schöpf ’s lecture expressed in detail the challenges faced by the research group and emphasizes the interpretative

mistake to consider pterins purine-like molecules.

Page 8: epub.ub.uni-greifswald.de · Table of contents 1. Pteridines and pterins

4

Figure 3 shows the proposed structure of xanthopterin during the fifteen years of research until 1940 when finally R.

Purrmann managed to discover its correct composition and conformation [17].

FIGURE 3 – PROPOSED MOLECULAR STRUCTURES OF XANTHOPTERIN FROM THE BOOK “CHEMISTRY AND BIOLOGY OF PTERIDINES”

Page 9: epub.ub.uni-greifswald.de · Table of contents 1. Pteridines and pterins

5

1.2 Nomenclature

For most chemical compounds, their nomenclature is influenced by the historical development of their chemistry. As

previously described, the pteridine family grew up in two different fields of research — the synthetical chemistry and

the isolation of natural compounds. Due to this pteridines literature is overcrowded with trivial names and various

numbering system.

The discovery of many natural pteridines or biomolecules containing the pteridine scaffold, has introduced into the

entire scientific literature a variety of different terminology, among which the term “pterins” represents the first

historical example. It was in 1941, after the structure elucidations achieved by R. Purrmann, that H. Wieland firstly used

the term “pteridine” to describe a pyrazine-pyrimidine ring system. In 1963 W. Pfleiderer suggested to use the name

“pterins” only for the pteridines 2-amino-4(3H)-pteridinone derivatives [11], this rule is nowadays almost universally

accepted (Figure 4).

Two numbering systems have been proposed by the scientific community – the first was used by F. Sachs and G.

Mayerheim at the beginning of the 20th century to define the “azine-purine” products and it beholds the idea of a

purine-like structure (1, Figure 4); the second was described by R. Kuhn and his A. Cook in 1937 and is nowadays the

most commonly accepted nomenclature (2, Figure 4) [11, 18].

FIGURE 4 – NOMENCLATURES

Page 10: epub.ub.uni-greifswald.de · Table of contents 1. Pteridines and pterins

6

1.3 Pteridines chemistry

In the book “Fused Pyrimidines”, D. J. Brown describes pteridines as a: «semiaromatic system with no great stability

towards ring fission, prone to nucleophilic but not electrophilic reactions and subject to covalent addition reaction». In

the following respective aspects will be detailed with a focus on the particular subclasses “pterins” [1].

1.3.1 Aromaticity, π-excessive and π-deficient

In order to better understand the π-deficiency of pteridines, it is useful to recall the concept of “aromaticity” and the

meaning of “aromatic character”.

The most of the organic chemistry books contain a chapter dedicated to benzene and its low reactivity compared to a

normal alkene system; a typical example is the bromination reaction which leads to a monosubstituted benzene and

not to an addition product (Scheme 2).

SCHEME 2 – BROMINATION ALKENE AND AROMATIC SYSTEM

The benzene ring has six carbon atoms, each bearing three sp2 and one unhybridized p orbitals. Two of these sp2 orbitals

are used for σ bonding with the adjacent carbons and the third one binds with an hydrogen atom. The remaining p

orbital, which is perpendicular to the planar ring, participates to a π-electron delocalization that makes the ring more

stable and provides it with particular features making it an “aromatic system” [18].

The aromaticity exemplified for the case of benzene is then generalized for all compounds showing a similar

delocalization and defined by the Hückel rule: «an aromatic compound contains a cyclic system of atoms whit a cyclic

conjugated, π bond system of (4n+2) π-electrons (Hückel number)» [19].

The effect of the aromaticity on reactivity and physical properties of a molecule can be summarized in few points [20].

1. Aromatics tend to undergo ring substitution reactions rather than addition reactions which are

characteristics of alkenes.

2. Cyclic system are more stable to oxidation and reduction than typical alkenes.

3. The existence of a “delocalization energy”.

4. Bond length in aromatic rings are intermediate, between normal σ bond and normal π bonded atoms.

5. Characteristic absorbance in the U.V. or visible region of the spectrum.

6. The exhibition of a ring current when an aromatic compound is placed in a magnetic field.

The so called “aromatic character” described above is an ideal situation and it is possible to find many aromatic

compounds deviating from this. An example are the heterocycles containing nitrogen compounds characterized by a

loss in aromaticity directly proportional to the N/C ratio [18].Three representative cases are pyrrole, pyridine and

pteridine heterocycles shown in the Figure 5. All these molecules are aromatic and the nitrogen atoms can be described

with exactly the same terms as benzene.

Page 11: epub.ub.uni-greifswald.de · Table of contents 1. Pteridines and pterins

7

In the case of the pyridine and pteridine, nitrogen carries the lone pare on one of the sp2 orbitals and only one electron

is localized in the p orbital perpendicular to the plane. The pyrrole ring is a different system which by binding hydrogen

atoms carries its lone pare, two electrons, on the unhybridized p orbital responsible for the π-electron delocalization.

The higher electronegativity of nitrogen, compared to carbon, effects directly the electron density distribution in the

ring, resulting in a distortion of the π-orbitals and a consequent loss of aromaticity [20]. In the case of the pyrrole this

effect is compensated by the higher number of electrons participating to the delocalization, which produces a higher

electron density on the carbon close to the nitrogen, also compared to the benzene model. That’s why these systems

are termed π-excessive. For pyridines or pteridines the vicinal carbon atoms are partially positive charged and the π-

delocalization is drastically reduced, therefore those heterocycles are termed π-deficient [20].

Based on this D.J. Brown describes this particular π-deficiency as a “semi-aromatic character”, meaning that all chemical

and physical properties of these heterocycles are strongly influenced. Pteridines represent among the heterocycles the

most effected class of compounds.

π – deficient π – excessive

FIGURE 5 – AROMATICITY IN HETEROCYCLES

Page 12: epub.ub.uni-greifswald.de · Table of contents 1. Pteridines and pterins

8

1.3.2 Stability of pteridines and pterins

The pteridine ring has 10 π-electrons contributing to the aromaticity like for instance naphthalene but, the presence of

nitrogen atoms results in a loss in stabilization energy, which makes the system susceptible to cleavage [20, 21]. The

carbon atoms of the pyrimidine moiety carriyng a partial positive charge easily undergo hydrolytic cleavage, acid or base

catalysed, resulting in the formation of substituted pyrazines (Figure 6) [20]. This instability can be softened by the

introduction of an electron donating group. For example the xanthopterin natural product carries an amino group and

is stable to boiling in 7 M hydrochloric acid and only slightly effected by boiling with barium hydroxide for 20 hrs [22]

(Figure 6).

FIGURE 6 – HYDROLYTIC CLEAVAGE PRODUCT AND XANTHOPTERIN

The photosensitivity of pteridines and pterins [23-25] is another important aspect that must be taken into account

during the synthesis and characterization of these compounds; reactions and work-up with pteridines has to be carried

out as prophylaxis in dark-room. The photodegradation of pteridines has been investigated and in most cases they

proceed through a Norrish-type II mechanism. One of the first examples in literature is the irradiation of biopterin under

aerobic conditions (Scheme 3) [26-28].

SCHEME 3 – PHOTOCHEMISTRY OF BIOPTERIN IN AQUEOUS SOLUTION FROM [26-28]

Page 13: epub.ub.uni-greifswald.de · Table of contents 1. Pteridines and pterins

9

1.3.3 Covalent hydration reaction

The addition of water to a double bound is a typical reaction in organic chemistry but few examples of them have a

covalent character. In most of the cases the stability of the final product and the nature of the double bond play the

main role. Many studies on a large range of aldehydes revealed almost fifty per cent of their respective hydrated form

in aqueous solution, while the reaction with alkenes has a high activation energy and to be overcome it is necessary the

application of a catalyst like hydrogen or hydroxy ions [29].

A different scenario is represented by the C=N bound which has been investigated for a long time and results in most

of the cases in a broken bond between carbon and nitrogen, like for the hydration of benzylidene-aniline to a Dimroth

base shown in Scheme 4 [29].

SCHEME 4 – ADDITION OF WATER TO C=N BOND

Pteridines are one of the few cases in which the formation of the hydration product does not lead to the expected

scission. This peculiarity is even more interesting if we consider that the additional reaction is associated to a further

loss in aromaticity, which as previously described, has already been pauperized by the distortion of the electron density.

A possible explanation was postulated by professor A. Albert from the University of Canberra. He attributes the easy

covalent addition to pteridines to two main factors — the first is the high electron affinity of the nitrogen that produces

an exposed polarized double bond and the second is an extra stabilization energy arisen from the formation of

resonance structures [29].

It was A. Albert who in 1951 observed for the first time the hydration of a pteridine. The professor and his co-worker

were investigating the pKa of different organic bases like pteridines and purines, when a strange phenomenon was

observed (Scheme 5); the titration curve of 6-hydroxypteridine with alkali revealed a weak acidity with a pKa of 9.7 as

value, but the back-titration with acids exhibited the formation of a new curve with a stronger acidity around a pKa of

6.7 [29].

SCHEME 5 – COVALENT HYDRATION

The interesting behaviour of this pteridine found an explanation in 1955 with the proposal of a covalent hydration

mechanism on one of the free carbon atoms; the exact structure of the neutral hydrated product 6,7-dihydroxy-7,8-

dihydropteridine (Scheme 5) was proven one year later by D.J. Brown and S.F. Mason [30] through their UV

spectroscopic studies on the species formed by titrations.

Page 14: epub.ub.uni-greifswald.de · Table of contents 1. Pteridines and pterins

10

Afterwards, the covalent hydration became an important parameter for the characterization of pteridines, since this

kind of reaction influences conformation, redox potential and pKa value in aqueous solution, which are fundamental

features in the biological system. The observation of this phenomenon had stimulated a large number of studies in

spectrometric and potentiometric research with pteridines. Therefore, it became necessary to establish a method to

reveal the presence of hydration. With this exact purpose A. Albert in cooperation with F. Reich in 1961 used the methyl

derivatives of 6-hydroxypteridine to confirm the attack of the water at position 7 (Figure 7) [31]. In their method the

presence of the bulky methyl-group on the carbon atom blocks the addition of water and other nucleophilic attacks.

This allows us to determine which of the derivative does not give the hydrated form and to localise the position involved

in the reaction mechanism (Figure 7).

FIGURE 7 – METHOD WITH METHYL SUBSTITUENT

Pterins are a particular subclass of pteridines and as previously described, the presence of an electron donating group

stabilizes the entire ring system. Nevertheless, examples of covalent addition are reported in the literature, like the

natural product xanthopterin which partially reacts with water and methanol (Scheme 6) [32].

SCHEME 6 – COVALENT ADDITION WITH METHANOL OF XANTHOPTERIN

The role of this mechanism in biology becomes extremely relevant for pterins, which are a class of compound occurring

in a huge variety of natural products and consequently having an echo in medicine, physiology, biotechnology and many

other disciplines. It is therefore always suggested the study of a possible addiction mechanism on pterins and, as

recommended by A. Albert «any substances which obstinately retains a molecule of water (on the evidence of

elementary analysis) should be examined for covalent hydration» [29].

Page 15: epub.ub.uni-greifswald.de · Table of contents 1. Pteridines and pterins

11

1.3.4 Reactivity of pteridine

The main topic of this dissertation is the chemistry of pterins and particularly the synthesis of the pterin-dithiolene

systems. It is not therefore my intention to describe each reaction about pteridines, but to focus on the most relevant

reactivity of this compounds’ family. Nevertheless, it is important to stress the utility of pteridine chemistry in

understanding the behaviour of pterins and its derivatives in chemical and biological fields [11].

Many reactions have been tested on pteridine ring systems resulting in a clear preference towards nucleophilic attack.

It is indeed the specific high depauperate π-electron layer that suppresses the typical affinity of aromatic system for

electrophiles; this reactivity is amplified by the introduction of an electron withdrawing group and repressed by electron

donating substituent.

Pteridines participate in four main classes of reactions — nucleophilic addition, nucleophilic metatheses, reductive

reactions and oxidative reactions. The homolytic c-acylation will be fully discussed later in a following chapter, since it

is the core of the synthetical strategy reported in this dissertation.

Nucleophilic addition

The covalent hydration and its implications on the biology for the natural occurring pteridines was already discussed. It

is, hence, likely that there will be the same mechanism with other nucleophiles, stronger then water, like alcohols,

amines and in general Michael reagents [33].

The link between the nucleophilic addition reaction on pteridine and the Michael reaction is interesting. They are both

deactivated by the introduction of a methyl group, for electronical and steric reasons and most of the Michael reagents

attack the pteridine ring (Scheme 7).

There is a general tendency to prefer positions 4,7 and in few cases position 6, but the introduction of substituents

make any substrate a particular case. The best example are pterin derivatives with their have two positions blocked and

an amino and hydroxy group that make the substrate more stable against nucleophilic additions [11].

SCHEME 7 – DIMEDONE ADDUCT FORM ADDITION OF A MICHAEL REAGENTS

Page 16: epub.ub.uni-greifswald.de · Table of contents 1. Pteridines and pterins

12

Nucleophilic metatheses

The presence of a leaving group on the pteridine ring makes the nucleophilic substitution easier. It is in fact present in

the literature a large number of nucleophilic metathesis; common leaving group are halogen, sulfo and alkylsulfonyl

(Scheme 8). Chloro is the most often used living group and has been employed in many syntheses as part of starting

material or intermediate product [34].

SCHEME 8 – NUCLEOPHILIC SUBSTITUTION ON PTERIDINES

Other substituents can be replaced when working with harsher conditions, a typical example is the reaction from

pteridinone to chloropteridines (Scheme 9), which requires high temperatures but at the same time permits obtaining

an extremely versatile intermediate product [35].

SCHEME 9 – NUCLEOPHILIC SUBSTITUTION

The nature of other substituents influences the reactivity of the substrate. An electron donating group like an amino

function makes the replacement of a good leaving group like chloro more difficult (Scheme 10).

Page 17: epub.ub.uni-greifswald.de · Table of contents 1. Pteridines and pterins

13

SCHEME 10 – NUCLEOPHILIC SUBSTITUTION

Oxidative reactions

While pyrimidines can be further oxidized to alloxan, the pteridines are already in a high oxidation state. The oxidative

nuclear reactions therefore usually involve the di- or tetra derivatives obtained by chemical reduction; a strong oxidant

reagent like potassium permanganate but also normal oxygen in basic condition can be successfully adopted. The

chemistry involved in those reactions is not really interesting, but it becomes fundamental for the study of biomolecules

like tetrahydrobiopterin (Scheme 11).

SCHEME 11 – OXIDATION OF PTERIDINES

The oxidation of pteridines with peroxide or peroxycarboxylic acid results in the formation of N-oxide derivatives, like

for the other heterocycles this product can be used as versatile intermediate. The regioselectivity of this reaction is

influenced by steric factors, like the introduction of a bulky group adjacent or in peri position, also the choice between

two different peroxycarboxylic acids has an effect. The 2,4(1H,3H)-pteridinedione and its 6,7 dimethyl derivatives are

oxidized at position 5 with trifluoroacetic acid and at position 8 using peroxide and formic acid. The targeted

regioselectivity is not always possible and in many case the product is a mixture (Scheme 12).

SCHEME 12 – FORMATION OF N-OXIDE

The N-oxide form has been largely used for the synthesis of 6-substituted pterins. The best example is the synthesis of

6-chloropterin (Scheme 13), in this case the formation of the N(8)-oxide actives the carbon 6 and permits introducing a

chloro substituent only in one position.

Page 18: epub.ub.uni-greifswald.de · Table of contents 1. Pteridines and pterins

14

SCHEME 13 – REGIOSELECTIVE INTRODUCTION OF CHLORO ON THE N-OXIDE DERIVATIVE

Reductive reactions

The reduction of the pteridine ring is in general an easy reaction. Depending on the stability of substrate and product is

possible to use sodium borohydride, sodium dithionite, catalytic hydrogenation, or metal amalgams (Scheme 14). The

reaction will preferably take place at the carbon atom without substituent [36].

SCHEME 14 – REDUCTION OF PTERIDINES

Also pterins have been successfully reduced using Platinum and hydrogen gas [37]. The synthesis of pterin molecules in

their reduced form is of particular interest for the biological application; the pterin-motive is present in different

important natural systems, with particular implications in the medicine and enzymology [38]. Three important examples

are the folic acid, the tetrahydrobiopterin and ultimately the molybdenum cofactor.

Page 19: epub.ub.uni-greifswald.de · Table of contents 1. Pteridines and pterins

15

2. Biological Background

2.1 Molybdenum dependent enzymes

Among the metals of biological relevance, molybdenum represents an interesting and valuable example for the

bioinorganic field. From the simplest bacterial to the human organism, this 4d transition metal has been selected by

evolution as cofactor constituent of many enzyme families. Despite the low percentage of molybdenum in the earth’s

crust, this metal is bioavailable as molybdate, a high water soluble form that makes molybdenum the most abundant

transition metal presence in sea water (MoO42-, molybdate 10 μg/L) [39]. The access to different oxidation states and

its redox-activity also under physiological condition make molybdenum a perfect and flexible candidate for a biological

system [40].

Depending on the nature of the active site, molybdenum dependent enzymes are divided in two groups – the

nitrogenase and the oxidoreductases. The first group is characterized by an iron-sulfur cluster (Figure 8), an unique

molybdenum complex that was centre of an intensive crystallographic discussion from 1992 to 2011 [41-43]; in

particularly the central atom was at the beginning unidentified than postulated as nitrogen and finally assigned as

hypervalent carbide.

FIGURE 8 – ATOMIC STRUCTURE OF FEMOCO OF NITROGENASE

The strong interest of the scientific community toward this enzymes is motivated by its peculiar and powerful ability to

catalyse the conversion of atmospheric nitrogen to ammonia, a reaction known as nitrogen fixation. The analogue

industrial conversion, called Haber-Bosch process requires extremely high temperature and pressure, while the

enzymatic way can be conducted at room temperature and atmospheric pressure [44].

The oxidoreductase group are characterized by the Molybdenum cofactor (Figure 9), which is probably the most

relevant discovery in the recent history of pterin chemistry and biochemistry. Indeed, the finding of pterin natural

products or derivatives had always stimulated great interest on this class of compounds, as for biopterin and folic acid

a huge number of publications, studies and stimulating ideas have been reported. The structural elucidation of Moco

was achieved by K.V. Rajagopalan, J.L. Johnson in 1992 [45], when the two scientists postulated the structure of the

molybdenum cofactor to be a pterin derivative.

From then on, the synthesis of Moco model compounds became an important field of research and after more than

twenty years of studies some good models have been synthetized [21], nevertheless many aspects of the cofactor need

to be clarified.

The structure described by K.V. Rajagopalan, J.L. Johnson was a 6-substituted fully reduced pterin with a phosphorylated

dihydroxybutyl side chain and carrying a cis-dithiolene functionality attached to the Molybdenum (Figure 9). This

composition was in 1995 largely confirmed with the first reported crystal structure of a molybdoenzyme, the aldehyde

ferrodoxin oxidoreductase (AOR) from Pyrococcus furiosus [46].

Page 20: epub.ub.uni-greifswald.de · Table of contents 1. Pteridines and pterins

16

The proposal of K.L. Rajagopalan was corresponding with the exception of the additional pyran ring, in half-chair

conformation, formed by the hydroxy group of the sidechain attacking the position 7 of the pterin ring (Figure 9). Further

crystal structures have later accredited the tricyclic form as representing the actual natural cofactor structure.

All these enzyme families catalyse oxygen and two electron transfer reactions from or to a substrate playing a key role

in the metabolic process of carbon, nitrogen and sulfur manly with detoxification purposes. A malfunction in the

biosynthesis of Moco or in the enzyme’s maturation causes two severe diseases the molybdenum cofactor deficiency

(MoCD) and the isolated sulfite oxidase deficiency (ISOD), described in the following.

Escherichia coli represent the best characterized system and it was chosen for the biological experimentation of the

Moco model compounds. It is therefore of utility to have a general overview of the enzymes characterized in this system.

The Molybdenum cofactor has been determined in three different configuration in Escherichia coli [40] and therefore

historically divided in the families — xanthine oxidase (XO), DMSO reductase (DMSOR) and the sulfite oxidase (SO) as

reported in Figure 10.

The active sites of these three families are characterized by the presence of the MPT ligand system and differ each other

by the coordination sphere of the metal centre. The supplement of a cytosine monophosphate in case of xanthine

oxidase and of guanine monophosphate in DMSO reductase constitutes an additional variation (Figure 10).

The xanthine oxidase family bears an oxidized MPT- MoVIOS(OH) nucleus with one equivalent of metal and MPT, they

are participating in a two electron transfer hydroxylation with water as source of oxygen. The sulfite oxidase family is

characterized by an MPT- MoVIO2 nucleus with one equivalent of metal to MPT and a further cysteine coordinated to

the meatal centre. Oxo transfer reactions are catalysed by these enzymes, with atom translocation occurring potentially

in both direction, to or from the substrate.

The DMSO reductase family is the most numerous group of proteins in Escherichia coli, it is characterized by a two

equivalents of MPT for one metal centre. The MPT2-MoVIO(X) core catalyses mainly oxygen atom transfer and

dehydrogenation reactions. In absence of the substrate they are active as terminal reductases, sulfur or proton transfer

catalyst, and non-redox catalyst. The oxygen in the coordination sphere is substituted by a sulfur in formate

dehydrogenase H, while the extra ligand X can be a cysteine, serine, selenocysteine and aspartate; in many cases they

are also involved in the respiratory system [47].

FIGURE 9 – MOCO STRUCTURE HYPOTHESIS AND MOLYBDOPTERIN (MPT)

Page 21: epub.ub.uni-greifswald.de · Table of contents 1. Pteridines and pterins

17

FIGURE 10 – DIFFERENT STRUCTURE OF MOCO EXPRESSED IN ESCHERICHIA COLI, SCHEME FROM REF. [40]

Page 22: epub.ub.uni-greifswald.de · Table of contents 1. Pteridines and pterins

18

2.2 Molybdenum cofactor natural synthesis and maturation

The biosynthesis of Moco may appear not directly of interest for the synthesis of Moco model compounds, but the

biological mechanism, intermediates and the proteins involved open the research view and permit the expression of

concepts and ideas for the understanding of the working principals in the molybdenum dependent enzymes. Therefore

the Moco synthesis in Escherichia coli is here shortly summarized with the intention to emphasize the possibilities and

the value of the synthetic protocol reported in this scientific research.

The natural synthesis of Moco is basically divided in three parts — cyclization of GTP) [48] (Scheme 15) to form the

cyclopyranopterin monophosphate (cPMP), sulfuration of cPMP to give the mature pyranopterin and finally the

coordination of molybdate to the cis-dithiolene anticipated by molybdenum uptake. The complete maturation of this

enzyme, namely the insertion of MPT into the apoenzyme will be described in the following text as an important

mechanism for the biological investigation on the Moco model compounds.

2.2.1 Cyclization of GTP

The first step involves a radical abstraction of a proton at the position 3’ of GTP carried out by MoaA, an enzyme which

belongs to the radical SAM family (S-adenosylmethionine) [49, 50], subsequent radical migration leads to the

intermediate 3,8-cH2GTP that was isolated and fully characterized. MoaC-catalyses the hydrolytic ring opening and

formation of the pyran cyclophosphate ring resulting in the intermediate cPMP [51] as reported in Scheme 15 (other

possible mechanism are reported in literature) [52].

SCHEME 15 – CYCLIZATION OF GTP, SCHEME FROM [40]

Page 23: epub.ub.uni-greifswald.de · Table of contents 1. Pteridines and pterins

19

2.2.2 Sulfuration of cPMP

The second step is catalysed by the MPT synthase, this enzyme is a heterotetramer constituting two small subunits-

MoaD and two larger MoaE. As shown in the Scheme 16 below two sulfur equivalents are required for the complete

sulfuration of the substrate. These sulfur atoms are carried by the smaller MoaD subunit while MoaE is binding the

cPMP intermediate.

SCHEME 16 – SULFURATION OF CPMP IMAGE TAKEN FROM REF. [40]

Page 24: epub.ub.uni-greifswald.de · Table of contents 1. Pteridines and pterins

20

2.2.3 Molybdenum uptake

While most of the first-row transition metals are already available as cation in the cell, molybdenum with its anionic

form access the cellular membrane via a highly specific transporter called ModABC [53]. This system belongs to the ATP-

binding cassete superfamily of membrane transporters, and it consists of two integral membrane proteins (ModB, B-

subunit), two ATP-binding peripheral membrane protein (ModC) and a periplasmic binding protein (ModA) (Figure 11)

[53-55].

Briefly summarised, the external periplasmic A-protein scavenges the oxoanion molybdate as tetrahedral complex,

which binds to a pair of translocating, transmembrane B-subunits (ModB), these prepare the cavity for the transport

and deliver to the cytoplasmic pair C-subunits with ATPase activity. The molybdate is finally released into the cytoplasm

[53]. The interaction of ModABC in molybdate-bound form with another protein called ModE [56] produces an

enhancement in the transcription of molybdenum enzymes like DMSO reductase, nitrate reductase [53].

FIGURE 11 – CELLULAR UPTAKE OF MOLYBDENUM AND TUNGSTEN, IMAGE TAKEN FROM REF. [16]

Page 25: epub.ub.uni-greifswald.de · Table of contents 1. Pteridines and pterins

21

2.2.4 Insertion of the metal

Once the molybdate is transported into the cytoplasm, as described above, the final step for the complete construction

of the molybdenum cofactor is the combination with the biosynthesized molybdopterin (MPT). In Escherichia coli this

task is manged by two enzymes MogA and MoeA, both participate in the process with different function. It has been

shown in vitro reaction that MoeA is able to mediate the anchoring of molybdate to MPT; MogA in absence of ATP

inhibits the reaction and accelerates if in presence of the same [40].

The phenomenon has been explained based on a crystal structure of a MPT-AMP product. MogA catalyses the

adenylation of the MPT substrate preliminary to the coupling with the molybdenum oxoanion (Scheme 17). The role of

MogA with the formation of the adenylated MPT is of relevant at physiological concentration of the metal, but it has

been proven that in concentrated solution the molybdenum insertion is accomplished by the MoeA alone [47, 57].

SCHEME 17 – METAL INSERTION SCHEME FROM [40]

Page 26: epub.ub.uni-greifswald.de · Table of contents 1. Pteridines and pterins

22

2.2.5 Cofactor insertion into apoenzyme

The insertion of the cofactor into the apoenzyme represents the final process of maturation for the molybdoenzymes,

namely the acquisition of the catalytic activity specific to the protein’s family. Therefore it is of extreme interest for the

researchers, which want to understand the mechanism of function in molybdenum enzymes carrying Moco. It is useful

to have a simplified introduction of this process looking at the acting proteins, but keeping in mind that many aspects

are postulated and many questions are still open (Figure 12) [40].

The synthetic address developed in this dissertation targets the synthesis of bis-dithiolene molybdenum complexes, i.e.

compounds that would act as model for a bis-MGD cofactor. Hence, it is appropriate to describe the maturation of a

DMSO reductase family ‘s enzyme, in particular the most explored TMAO reductase (TorA) with its chaperon TorD.

FIGURE 12 – IMAGE TAKEN FROM REF. [40]

The biosynthesis of the molybdenum cofactor is a highly conserved process in all organisms employing molybdenum

enzymes. In Escherichia coli after Moco-basic formation, depending on the family of enzymes, further modifications are

needed before the complete maturation takes place (Figure 12). To xanthine oxidase is added a CMP (cytosine

monophosphate, from CTP) via pyrophosphate bond and also a further sulfuration of the Mo=O bond. In the DMSO

reductase family the cofactor receives a GMP always via pyrophosphate bond, but it couples itself with another MPT,

resulting in a bis-MGD form. Different case is the sulfite oxidase family where the cofactor is directly inserted into the

apoenzyme and it does not require any other steps [40].

As discussed above after the coordination of molybdate to MPT, such Moco basic form is involved in structural

modifications by two proteins MobA and MobB, while the first plays definitely an important role, MobB has probably

some functions as adapter protein working in concert with MobA. However the maturation process has been

reproduced in vitro also in absence of MobB.

MobA is a highly specific GTP binding protein and as first step combines Moco with a nucleotide resulting in a MGD,

which is later coupled with another molecule MPT to produce bis-MGD. This passage (step 2) represented in Figure 12

which culminates in the insertion of bis-MGD into ApoTorA, can occur direct on MobA (which has enough space) or on

Page 27: epub.ub.uni-greifswald.de · Table of contents 1. Pteridines and pterins

23

the surface of the enzyme (Apo-Tor A, TorA). In both cases this happens in collaboration with the chaperon TorD [40,

57]. The mechanism adopted is still an enigma and requires more in depth studies and experimentation, possibly with

the employment of model compounds.

TorD is probably the protein that fulfils the majority of the tasks in the entire maturation process. It belongs to a family

of molecular chaperons [58] present in bacterial and in archaea cells. Its ability to interact has been proven with most

of the players of this natural process. First and foremost it binds with the molybdenum enzyme TorA, which in absence

of it decompose (70-80%) under thermal stress; thus it is reasonable to assume that the first job of TorD is to protect

the enzyme and to carry it to the Tat-machinery for the periplasmic release.

Furthermore TorD is involved in the maturation of bis-MGD. First he interacts with MobA and Mo-MPT for the addition

of a GMP from GTP and later most probably he works as platform for the formation of bis-MGD. It has also been proven

that the chaperone influences the maturation level of ApoTorA in vitro with the source of the molybdenum cofactor,

which in presence of TorD increases from twenty at basal level to eighty percent. Thus, the chaperone somehow

facilitates the insertion of bis-MGD in ApoTorA in a short time in order to avoid the proteolytic attack on the holoprotein

(Figure 12).

The last step of the maturation process involves preTorA and TorD, which remains bound to the protein until the Tat-

machinery is prompted to transport the molybdenum enzyme to the periplasm. This passage is till now not clear, but

most probably the release of TorD plays also in this case a role for the activation of the Tat-machinery.

Page 28: epub.ub.uni-greifswald.de · Table of contents 1. Pteridines and pterins

24

2.3 Molybdenum cofactor and Human diseases-MoCD and ISOD

The molybdenum dependent enzymes, count around one-hundred of components, present in all kingdoms of life. As

previously described except for the nitrogenase carrying the FeMoco active site, the majority harbours the molybdenum

cofactor, a unique biological motif central for the carbon, sulfur, and nitrogen cycles [59].

So far four molybdenum dependent enzymes are formed in the human — sulfite oxidase (SO), xanthine oxidoreductase

(XOR), aldehyde oxidase (AO) and only recently discovered was the mitochondrial amidoxime-reducing component

(mARC) [60, 61]. These proteins are mainly involved in redox reactions using water as acceptor or donor of oxygen; in

particular SO and XOR have been intensely studied for their important role in the catabolic reactions in cysteine and

purine metabolism whereas AO and mARC require an “in depth-analysis” about their exact physiological function in the

human organism.

Two important pathologies are related to Moco — the molybdenum cofactor deficiency (MoCD) and the isolated sulfite

oxidase deficiency (ISOD). Both are autosomal recessive disorders and lead to a severe impairment or directly to the

death of the patients. The molybdenum cofactor deficiency was reported for the first time in 1978 when a baby of a

Danish family died after it had shown the typical symptomatology of MoCD, namely therapy-resistant seizures,

neurological abnormalities, lens dislocation, dysmorphic shape of the head, feeding difficulties, occurring during the

neonatal period (0-28 days of life) [59]; most probably the other siblings, which had previously died, were also affected

but no analysis was reported. Since this first case, about one hundred patients have been recorded, this is a quite rare

pathology that counts around 1 case in 100.000 to 200.000 live births; nevertheless many studies have been started and

also some treatments addressed [59].

The biosynthesis of Moco is a highly conserved biological process, meaning that the basic pathway is the same in all

kingdoms of life with the employment of related enzymes specific for the living organism. At the beginning of this

chapter a detailed description of the Moco biosynthesis is given. It is therefore sufficient to show the main passages

constituting the formation of the cofactor and consequently maturation in order to understand at which steps MoCD

and ISOD are induced (Scheme 18).

SCHEME 18 – STEPS OF THE BIOLOGICAL SYNTHESIS OF MOCO AND MATURATION OF THE ENZYMES

Four main passages delineate the natural synthesis and maturation of the enzymes — cPMP synthesis, MPT synthesis,

coordinatoion to the molybdenum ( synthesis of Moco) and final insertion into the apoenzymes [59].

MoCD is caused by biallilic phatogenic variants in the enzymes MOCS1, MOCS2 or GEPH [62] involved in the formation

of Moco, and is clasified in types A, B or C depending on which step of the process is impaired; the ISOD is localized in

the last step of coordination with the apoezyme. In all types of pathology the accumulation of sulfite, taurine, thiosulfate

Page 29: epub.ub.uni-greifswald.de · Table of contents 1. Pteridines and pterins

25

and all the related toxins is the most dangerous and deleterious effect. It causes permanent damage to the patients or

directly their death.

2.3.1 Synthetic cofactor, drug for a possible cure

The MoCD type A desease has been already succefully treated by the employment of a biothechnology prepared cPMP.

This method was developed by Professor G. Schwarz at the University of Cologne [63] and it helps many patients every

year.

Unfortunately, there is not yet a treatment for the MoCD type B, C and the ISOD; in particular the instability of Moco

and of the enzyme does not permit their employment as drugs.

The final goal of the work reported in this dissertation is the preparation of a synthetic cofactor, which can be used as

drug for the MoCD type B,C and ISOD. The synthetical strategy developed in this research makes possible the

preparation of model compounds of MPT and in the future also of Moco. Indeed, the study of these models allows a

deeper comprehension of the natural cafactor and its enzymes families.

The general features of our hypothetical cofactor can be summarized in four points:

✓ The cofactor has to be of about the same size as the biological cofactor or smaller and able to bind inside the

pocket.

✓ The cofactor has to be able to catalyse the oxygen transfer reactions.

✓ Because MPT bind to molybdenum by its dithiolene function this feature will be part of every model.

✓ The synthetised cofactor has to be more stable than the natural cofactor.

In these context the chemistry reported in the following are basic studies for the development of possible treatment

for MoCD and ISOD.

Page 30: epub.ub.uni-greifswald.de · Table of contents 1. Pteridines and pterins

26

3. Results and discussion

3.1 Modelling molybdopterin (MPT)

The modelling of natural products is of utmost importance for the medical research and in general for fundamental

biological investigations. Succeeding the elucidation of the Moco structure by K.V. Rajagopalan, J.L. Johnson in 1992

[64] the literature output on MPT, its chemistry and biology increased. Many studies were centred on MPT models, but

many aspects of the fundamental behaviour of the Moco enzymes have to be clarified still. In the last quarter of the

20th century different synthetical approaches were developed and some good model compounds were synthesized.

The numerous and rich portfolio of results make a comprehensive schematic résumé extremely difficult, but some

important works are worthy to be mentioned.

The first is the entire synthesis of the masked form of MPT reported by B. Bradshaw, C. D. Garner and J. A. Joule et al.

in 2001 [65]. This publication is the closure of an intense research started in the 90’s with the aim to develop a protocol

for the synthesis of MPT [66-68]. Initially, the quinoxaline-halogen derivative II was investigated as an easier platform

for the coupling reaction and later they prepared pterin substrate III comprising all the necessary protections (Figure

13).

The laborious protocol permits the preparation of product VI, which represents the closest synthetic system to the MPT

natural product. Many issues were encountered during the development of the coupling reaction, in particular low

yield, no reactivity and homocoupling of the dithiolene partner I. Finally, it was optimized a Stille cross-coupling with

copper thiophene-2-carboxylate as a stochiometric mediator with sixty percent of yield [69].

Complexation reactions of this ligand have been heralded, but never published, probably due to the long and laborious

procedure that makes the following step extremely costly and difficult. Speculations are not helping at all, rather, it is

useful to contemplate this huge work as the first big effort in the synthesis of the natural product MPT and extract all

the information necessary for future research.

FIGURE 13 – REAGENTS OF THE COUPLING REACTION OF B. BRADSHAW

Page 31: epub.ub.uni-greifswald.de · Table of contents 1. Pteridines and pterins

27

In this publication are reported, in fact, many important mechanisms besides the cross-coupling reaction. First of all,

the formation of the pyran ring was carried out by an elegant reduction-protection, protocol introduced by F.W. Fowler

in 1972 as a protected-stabilized form of reduced pyridines using sodium borohydride in the presence of methyl

chloroformate [70]. Interestingly, in the case of the quinoxaline derivative (Scheme 19) the reaction with benzyl

chloroformate in absence of solvent or added-base and at room temperature proceeds toward the formation of the

pyran ring without reductant; moreover only one diastereoisomer was found [68].

SCHEME 19 – REDUCTION-PROTECTION PROTOCOL WITH SIMULTANEOUS RING CLOSING STEP

In the case of MPT synthesis, the ring protection-reduction leads to the formation of two diastereoisomers, among

which the unwanted one comes as minor product and can be recycled by separation through chromatography column.

Another important aspect is the second reduction of the pyrazine moiety with cyanoborohydride, producing only one

diastereoisomer as for quinoxaline, without cleaving the dithiolene masked form, promising a sufficient stability also

for the model compounds synthesized in this dissertation.

The final step is the introduction of the protected phosphate unit carried out with N,N-diisopropyl-bis-

[2(methylsulfonyl)ethyl]phosphoramidite in the presence of triazole [71]; also this reaction could be used to implement

the compounds reported in the following.

Page 32: epub.ub.uni-greifswald.de · Table of contents 1. Pteridines and pterins

28

The best model-compound for the molybdenum cofactor was synthesized by Burgmayer Group in 2007, supported even

by crystal structures [72]. The synthetic approach described in Scheme 20 employs the direct reaction between a

molybdenum polysulfide with an unsaturated bond, inserted through Pd C-C coupling reaction at the 6 position of a

pterin-substrate [21]. This protocol is of extreme utility and permitted the production of different molybdenum

complexes; the only limitation is the necessary presence of an electron-withdrawing group at the alkyne functionality.

The exceptionality of this model is even more amplified by the observation of an interesting solvent-dependent

pyranopterin cyclization; it has been, in fact, proven an interconversion between tricyclic and bicyclic forms, thought

the insertion of the hydroxy group close to the 7 position of the pterin moiety. This phenomenon, reminiscent of the

covalent addition as classical behaviour of pteridines has been hypothesized to being part of the catalytic mechanism

of Moco [73]. Presumably the only downside of this model compound is the water insolubility (not favoured by the huge

organic moiety), which is a critical point for the planning of biological studies.

The last example is the most relevant for this dissertation and it permits the preparation of pterin-dithiolene ligands.

The most common protocol for the synthesis of dithiolene in masked form starts from a carbonyl functionality [74]. On

this basis, J.A. Joule et. al. have optimised a protocol for the synthesis of asymmetrical dithiolene ligands from a carbonyl

attached on the 6 position of pterin. Moreover they proved the feasibility of this method for the preparation of bis-

dithiolene complexes [75] (Figure 14). Besides the great achievement, the synthesis of the pterin substrate remains

quite complicated starting from 6-formylpterin, prepared with a fifty percent of yield by the hydrobromic acid

degradation of commercial folic acid; furthermore the construction of the acetic groups requires different synthetical

passages [76].

SCHEME 20 – SYNTHETICAL SCHEME TAKEN FROM REF.[72]

Page 33: epub.ub.uni-greifswald.de · Table of contents 1. Pteridines and pterins

29

FIGURE 14 – COMPLEXES BY J.A. JOULE ET AL WITH PTERIN-DITHIOLENE LIGANDS

Like for the model compound of Burgmayer’s group, the poor water solubility remains a disadvantage. In fact, the

isolation of the molybdenum complex in pure form had required, the exchange of the counter cation with consequent

decrease of the water solubility.

The main observation arising from these three approaches are the particular hurdles in Moco modelling research; the

topic requires high familiarity with the organic synthesis of natural products, one of the most challenging fields in organic

synthesis and last but not least high competence in handling the complexation reactions.

This dissertation is part of a wider project centred exactly on this problematic, with the scope to put aside the

complicated task of the entire MPT synthesis and focus its attention on the subunits composing Moco; namely three

important moieties are targeted – the pterin rings, the pyrazine-pyran bicyclic system and the phosphate lateral chain

(Figure 15). Each of these subunits has a particular influence on the cofactor activity and the study of relative complexes

with molybdenum represents a possible key to a better understanding of the Moco principals of functions.

Theoretical calculations have suggested a partial influence of the pyrazine ring on the active site; the extension with

also the pyran unit which has been already indicated as participating in the activity with its scission-cyclization

mechanism, constitutes a desirable model compound [77]. The phosphate functionality does not have an electronic

influence on the main structure, however it is surely able to create hydrogen bonding and keep the cofactor tightly

inside of the apoenzyme’s pocket.

The pterin moiety has been also exonerated from any electronic role in Moco [77], but it’s great ability to create

intermolecular hydrogen bonding plays most probably a role; recent vibrational studies have suggested a mechanism

of fine-tuning Moco for electron and atom-transfer reactivity in catalysis [78]. Finally, the dithiolene moiety is of extreme

importance, since it is directly connected to the metal centre and well known as a non-innocent ligand system [79].

In this research work, the pterin unit has been investigated and the results are discussed in the following text, moreover

the cooperation with a co-worker has permitted the synthesis of new interesting structure combinations of pterin with

the phosphate subunit.

Further biological characterization is planned, namely the study of a possible interaction between proteins involved in

the maturation of the molybdenum dependent enzymes and the apoenzymes (see chapter 2 “Biological Background”).

Page 34: epub.ub.uni-greifswald.de · Table of contents 1. Pteridines and pterins

30

FIGURE 15 – BREAK DOWN APPROACH FOR MPT

Page 35: epub.ub.uni-greifswald.de · Table of contents 1. Pteridines and pterins

31

3.2 Protection of Pterins

The synthesis of pterin-dithiolene ligands has already been successfully achieved by the two approaches described

above and in both cases the researchers had to face the solubility problems of pterins. Mother issue has been the

selection of suitable protection for the amine and amide functionalities in order to increase the solubility in organic

solvents. [80].

The protecting group was chosen taking into account the stability during the synthetical pathway and also the potential

cleavage at the end of the process, which permit the modelling of the natural free form. The literature is rich of possible

pterin derivatives with blocked functionality, and in most of the cases an increment of solubility has been described,

but a detailed respective evaluation of the large assortment is not reported. Thus, the most often used protections with

relative good stability were chosen. Two different pterins were prepared according to literature carrying two different

protecting groups concomitant – methanimidamide N'-(6-chloro-3,4-dihydro-4-oxo-2-pteridinyl)-N,N-dimethyl-(5) and

2-pteridinamine, 4-(pentyloxy)-(10) (Figure 16).

Substrate 5 is the starting material of the coupling protocols developed by C. D. Garner and J.A. Joule (Figure 13) for the

synthesis of the masked form of MPT previously describe [65]. This product was protected at the amino group employing

a particular product, called Bredereck’s reagent, usually used in α-methylation, α-amination of various carbonyl systems,

ring-opening polymerization and indole synthesis [81]. Substrate 10 was prepared following a procedure by D. Mohr, Z.

Kazimierczuk and W. Pfleiderer for the synthesis of 6-thioxanthopterin and 7-thioisoxanthopterin [82].

Both protecting groups are cleaved under strong basic conditions, which is typical step of a complexation reaction with

molybdenum, thus, removable at the end of the entire synthetic pathway. Product 10 can be also hydrolysed by acid,

albeit with lower efficiency [82].

As illustrated in Scheme 21, the synthesis of 5 starts with the formation of the pyrazine ring 1 and builds up the

pyrimidine by condensation with a guanidine salt. After hydrolysis of the amino group product 3 is obtained. The

interesting and critical transformation is the insertion of the chlorine atom (see chapter 1 “Pteridine and Pterins”). The

use of the N(8)-oxide form induces a stereospecific insertion at position 6 of the pyrazine ring and yields the chemical

structure 4. While the chemistry of N-oxide heterocycles would suggest an α-rearrangement, in this case the position 6

is preferred by a faster β-rearrangement accompanied by the release of the oxide group [83]. In the last step, the amino

group reacts with Bredereck’s reagent with a consequent increase of the solubility.

FIGURE 16 – 5 BREDERECK’S REAGENT AS PROTECTION, 10 PENTHYLOXY

PROTECTION

Page 36: epub.ub.uni-greifswald.de · Table of contents 1. Pteridines and pterins

32

SCHEME 22 – CARBONYL PROTECTION, PENTYLOXY

The preparation of substrate 10 is an Isay-Gabriel condensation [80], that is the reaction between a pre-protected

pyrimidine 7 with a glyoxal molecule. Using the previous N(8)-oxide protocol a chlorine atom is then stereospecifically

attached at position 6 (Scheme 22).

SCHEME 21 – AMINO PROTECTION, BREDERECK’S REAGENT

Page 37: epub.ub.uni-greifswald.de · Table of contents 1. Pteridines and pterins

33

Comparison of the two products reveals a higher solubility of 10 in a larger variety of solvent, in particular in those with

low polarity, while product 5 surprisingly exibits a better solubility in more polar solvents. This led us to first attempting

to use the pentyloxy as protecting group.

Moreover, the high solubility of the substrate in a broad range of solvents allowed the application of different crystal-

growing techniques and eventually x-ray characterization which hasn’t been avalable before (Figures 17,18,19).

FIGURE 19 – MOLECULAR STRUCTURE OF PRODUCT 10

BASED ON NOT ENTIRELY RELIABLE STRUCTURAL DATA

FIGURE 17 – MOLECULAR STRUCTURE OF PRODUCT 6

FIGURE 18 – MOLECULAR STRUCTURE OF PRODUCT 9

Page 38: epub.ub.uni-greifswald.de · Table of contents 1. Pteridines and pterins

34

In this dissertation the major part of the synthetic work is focused on the synthesis of 6-acyl pterins as starting point for

the construction of the dithiolene ring. Nevertheless, some attempts to prepare also a coupling partner were

undertaken, e.g. the reaction reported below in Scheme 23 for the preparation of the iodine derivative was tested with

the undesired cleavage of the pentyloxy protection as result.

SCHEME 23 – ATTEMPT TO EXCHANGE THE CHLORINE BY IODINE USING HYDROIODIC ACID

Page 39: epub.ub.uni-greifswald.de · Table of contents 1. Pteridines and pterins

35

3.3 First attempt for the synthesis of pterin-dithiolene ligand, preparation of 6-acyl

pterins by condensation

The first attempt for the synthesis of a pterin-dithiolene ligand started with a stereoselective Timmis condensation

reaction [80, 84] as reported by W. Pfleiderer and G. Heizmann [85]. This condensation allows the preparation of 6-

acetyl-4-cyclohexyloxy-7-methylpterin (Scheme 24).

SCHEME 24 – SYNTHESIS OF A 6-ACYL PTERIN REPORTED BY PFLEIDERER

Encouraged by the positive crystallographic results and increased solubility due to employing the pentyloxy protecting

group, it was decided to modify the reaction and use the nitroso pyrimidine intermediate 6 and proceed with the

condensation with acetylacetone (2,4-pentandione, Scheme 25).

SCHEME 25 – SYNTHESIS OF 4 PENTYLOXY-6-ACYL-7 METHYL PTERIN BY A MODIFIED PROTOCOL OF W.PFLEIDERER

The targeted condensation product 11 was obtained with a yield of eighty percent. Growing crystals by the slow

evaporation method gave, as desired, a good and reliable crystal structure reported in Figure 20 below.

Page 40: epub.ub.uni-greifswald.de · Table of contents 1. Pteridines and pterins

36

FIGURE 20 – MOLECULAR STRUCTURE OF PRODUCT 11

The successive transformations, following the procedure for the masked form of dithiolenes of C. D. Garner, comprise

bromination in α-position of the carbonyl and substitution with isopropyl xanthate. Among a large number of

brominating agents it was decided to use PHT (2‐Pyrrolidone hydrotribromide) as mild reagent that permits maintening

the protecting group; moreover PHT is described in the literature as highly selective for ketones [86, 87].

In particular, the combination of PHT with an excess of 2-pyrrolidone prevents any acidic or basic hydrolysis due to the

formation of a hemihydrobromide salt with the leaving hydrobromic acid as described in Scheme 26.

SCHEME 26 – BROMINATION WITH PHT

As a precautionary measure the bromination of the synthesized 6-acyl pterin 11 was carried out in dried tetrahydrofuran

since the brominating agent is sensitive to water. Moreover, this avoids the possible involvement of an acidic or basic

environment and also the possible substitution of the bromine atom. After bromination the crude product was treated

with potassium isopropyl dithiocarbonate as a source of the protected dithiolene sulfur and oxygen atoms. This reaction

proceeds at room temperature in acetone with the precipitation of potassium bromide as clear white solid. The product

obtained and purified was supposed to be the expected product with a xanthate moiety in α-position of the carbonyl

attached to position 6 (Scheme 27).

SCHEME 27 – BROMINATION WITH PHT AND SUBSTITUTION WITH ISOPROPYL XANTHATE

Page 41: epub.ub.uni-greifswald.de · Table of contents 1. Pteridines and pterins

37

The elemental analysis and 1H and 13C NMR supported the success of the synthesis of the targeted structure. However

results obtained later revealed a misinterpretation of the analytical data and an unexpected synthetical side reaction

(Table 1).

TABLE 1 – ELEMENTAL ANALYSIS OF THE EXPECTED PRODUCT

Even though the clarification of this synthetic struggle is not the topic of this dissertation, the problems faced are of

value for the next person working with these structures and of analytical interest for the understanding of future studies.

In particular for the interpretation of NMR spectra, described below, previous publications reporting pterins carrying

the pentyloxy protecting group [88-92] were taken into considerations, together with the analysis of the new products

prepared in this dissertation. A mechanistic assumption about the result and a crystal structure is provided in the final

part of this section, with the confidence that the substrate obtained is not suitable for the main aim of this dissertation,

or at least not with the applied protocols. Therefore, after really intense and long work dedicated to this product, it was

decided to shelve it and focus the efforts on the acylation protocol described in the following.

Expected product N C H S

Calculated 16.53 51.04 5.95 15.14

Measured 16.31 50.53 5.64 16.31

Page 42: epub.ub.uni-greifswald.de · Table of contents 1. Pteridines and pterins

38

The original assignment of the 1H NMR signals of the pentyloxy protected addresses the typical overlap of the CH2 peaks

on the alkyl chain B – C and the broad signal of the amino function H, which is in many cases not visible. The other

signals were easily assigned as shown in Spectrum 1, except for the singlets E and F which belong either to the CH3 at

the 7 position of the pterin or to the CH3 in α-position of the carbonyl (Spectrum 1).

7.5 7.0 6.5 6.0 5.5 5.0 4.5 4.0 3.5 3.0 2.5 2.0 1.5 1.0 0.5

Chemical Shift (ppm)

0

0.05

0.10

0.15

0.20

0.25

0.30

0.35

0.40

0.45

0.50

0.55

0.60

0.65

0.70

No

rma

lize

d In

ten

sity

3.024.062.182.993.002.011.94

0.9

30

.96

0.9

8

1.3

81

.391.4

1

1.4

51

.461.4

71

.48

1.4

81

.49

1.5

01

.51

1.5

51

.56

1.9

11

.93

1.9

6

2.7

6

2.9

2

4.5

34

.55

4.5

8

6.0

0

7.2

8

Spectrum 1 – 1H NMR of product 11 1H NMR (300MHz ,CHLOROFORM-d) δ = 6.00 (br. s., 1 H), 4.55 (t, J = 6.8 Hz, 2 H), 2.92 (s, 3 H), 2.76 (s, 3 H),

2.00 - 1.85 (m, 2 H), 1.60 - 1.35 (m, 4 H), 1.04 - 0.88 (m, 3 H) ppm

The 13C NMR spectra were assigned based on the precedent literature and the additional application of a DEPT 135 13C

NMR experiment, which clearly shows the presence of the CH2 negative signals, the CH/CH3 positive signals and the

disappearance of the quaternary carbons. Like for the proton NMR, the exact assignment of the C7-CH3 and CH3-C=O

signals at 27.47 and 25.15 ppm is not possible using this method (Spectrum 2).

D

A

C B

D

E or F

G

H

A

B – C

H F

E G

Page 43: epub.ub.uni-greifswald.de · Table of contents 1. Pteridines and pterins

39

95 90 85 80 75 70 65 60 55 50 45 40 35 30 25 20 15

Chemical Shift (ppm)

0

0.05

0.10

0.15

0.20

0.25

0.30

0.35

0.40

0.45

0.50

0.55

0.60

0.65

0.70

0.75

0.80

0.85

0.90

0.95

1.00

No

rma

lize

d In

ten

sity

13

.96

22

.33

25

.13

27

.46

28

.06

68

.50

95 90 85 80 75 70 65 60 55 50 45 40 35 30 25 20 15

Chemical Shift (ppm)

-1.0

-0.9

-0.8

-0.7

-0.6

-0.5

-0.4

-0.3

-0.2

-0.1

0

0.1

0.2

0.3

0.4

0.5

0.6

0.7

No

rma

lize

d In

ten

sity

13

.97

22

.32

25

.1427

.47

28

.07

68

.49

Spectrum 2 – 13C NMR DEPT 135, product 11

13C NMR (75 MHz): δ = 14.0 (CH3), 22.3 (CH2), 25.1 (CH3), 27.5 (CH3), 28.1 (CH2, CH2 ), 68.5 (CH2) ppm

The original interpretation of the 13C NMR spectrum assigned an overlap of the γ and δ carbon atoms at 22.32 ppm. Nevertheless, the structure C’ which will be described later, reveals the presence of two almost overlapped signals around 28 ppm. This belongs most probably to the β and γ CH2 of the extranuclear alkyl chain (Spectrum 2).

ε

δ β

α γ

C7-CH3 / CH3-C=O

β

δ

α

γ

ε

CH3-C=O

C7-CH3

Page 44: epub.ub.uni-greifswald.de · Table of contents 1. Pteridines and pterins

40

200 195 190 185 180 175 170 165 160 155 150 145 140 135 130 125 120 115 110 105 100

Chemical Shift (ppm)

0

0.05

0.10

0.15

0.20

0.25

0.30

0.35

0.40

0.45

0.50

0.55

0.60

0.65

0.70

0.75

0.80

0.85

0.90

0.95

1.00

No

rma

lize

d In

ten

sity

12

0.6

1

14

2.6

6

15

7.0

6

16

1.3

7

16

3.0

0

16

7.8

7

19

9.8

9

Spectrum 3 – 13C NMR of product 11, quaternary carbons 13C NMR (75MHz ,CHLOROFORM-d) δ= 199.9, 167.9, 163.0, 161.4, 157.1, 142.7, 120.6 ppm

The bromination and substitution with sodium isopropyl xanthate is accompanied by the appearance of three important

signals – the peaks L and I belonging to the O-isopropyl and a singlet at 4.9 ppm (E’ or F’) with integration 2, which is

ascribable to a CH2 that has been brominated first and then received the xanthate unit. Indeed, one of the CH3 has

disappeared (Spectrum 4).

8.5 8.0 7.5 7.0 6.5 6.0 5.5 5.0 4.5 4.0 3.5 3.0 2.5 2.0 1.5 1.0

Chemical Shift (ppm)

0

0.05

0.10

0.15

0.20

0.25

0.30

0.35

0.40

0.45

0.50

0.55

0.60

0.65

0.70

0.75

0.80

0.85

0.90

0.95

1.00

No

rma

lize

d In

ten

sity

3.016.114.041.992.612.002.011.042.00

DMSO

Water

0.8

80

.90

0.9

3

1.2

91

.31

1.4

11

.42

1.4

6

1.7

91

.821.8

41

.86

1.8

9

2.6

4

4.4

94

.51

4.5

3

4.9

0

5.5

45.5

65

.58

5.6

05

.62

5.6

45

.677

.68

7.8

0

7.9

3

8.0

6

Spectrum 4 – 1H NMR product of bromination and substitution with xanthate (expected product) 1H NMR (DMSO-d6) δ = 7.80 (br. s., 1 H), 7.68 (br. s., 1 H), 5.60 (td, J = 6.3, 12.3 Hz, 1 H), 4.90 (s, 2 H), 4.51

(t, J = 6.6 Hz,2 H), 2.64 (s, 3 H), 1.84 (quin, J = 6.9 Hz, 2 H), 1.52 - 1.35 (m, 4 H), 1.30 (d, J = 6.4 Hz, 7 H), 0.94 - 0.85 (m, 3 H) ppm

L

I

E ’or F’

C=O

C-2 C-8A

C-4

C-6

C-7

C-4a

A

C B

D

E or F

G

H

I

E’ or F’

L

C=O

Page 45: epub.ub.uni-greifswald.de · Table of contents 1. Pteridines and pterins

41

The relative good matching of the analytical with the expected data encouraged us to proceed to the second and final

step for the synthesis of a pterin-dithiolene ligand. This step was therefore carried out using concentrated sulfuric acid,

according to the method described by C. D. Garner et al. [76]. Unfortunately, applying the reported protocol did not

give any result; In particular, the only effect observed was the hydrolysis of the protecting group within two or three

days in pure sulfuric acid.

Subsequently sodium diethyldithiocarbamate was used in the nucleophilic substitution of bromine.The typical signals

of the carbamate chain N and O arrised coherently with the previous product. After purification and characterization

the same protocol was applied with the hope of a different outcome, but unfortunately also in this case no reaction was

observed.

10.5 10.0 9.5 9.0 8.5 8.0 7.5 7.0 6.5 6.0 5.5 5.0 4.5 4.0 3.5 3.0 2.5 2.0 1.5 1.0 0.5

Chemical Shift (ppm)

0

0.05

0.10

0.15

0.20

0.25

0.30

0.35

0.40

0.45

0.50

0.55

0.60

0.65

0.70

0.75

0.80

0.85

0.90

0.95

1.00

No

rma

lize

d In

ten

sity

3.143.283.194.012.042.562.202.392.032.172.04

DMSO

Water

0.8

80

.90

0.9

31

.12

1.1

41

.24

1.2

61

.38

1.4

21

.44

1.4

61.8

21.8

41

.86

2.6

4

3.7

53

.77

3.9

03

.93

4.4

84

.51

4.5

3

4.9

9

7.6

57

.70

7.7

8

Spectrum 5 – 1H NMR product of bromination and substitution with carbamate (experimental section, B’- carbamate

derivative) 1H NMR (300MHz ,DMSO-d6) δ = 7.65 (s, 1 H), 7.70 (s, 1 H), 4.99 (s, 2 H), 4.50 (t, J = 6.6 Hz, 2 H), 3.91 (q, J = 6.5 Hz, 2 H), 3.76 (q, J =

6.9 Hz, 2 H), 2.64 (s, 3 H), 1.84 (quin, J = 6.9 Hz, 2 H), 1.53 - 1.33 (m, 4 H), 1.24 (t, J = 7.0 Hz, 3 H), 1.20 - 1.09 (m, 3 H), 0.97 - 0.85 (m,

3 H) ppm

The protonation of the amino group, which leads to a change in the electronic distribution of the pterin ring has been

postulated to be the cause of the unreactivity of the substrate. The previous examples in the literature were in fact

either protected or proposed to have a different basicity. Therefore it was also tried to reduce the acidic strength and

concentration using a diluted solution of sulfuric acid, hydrochloric acid, perchloric acid and acetic acid. Later, it was

also tried to reduce the contact surface between the reagent by the employment of HCl in gas form, with the

understanding that the ring closing is an acid catalyzed reaction.

The hydrochloric acid in gas form was obtained by the controlled reaction between sodium chloride and concentrated

sulfuric acid. The apparatus was constructed by joining two schlenks, one containing sodium chloride and the other

equippedt with a filter carrying the substrate and an outlet directed to a basic water solution (sodium hydroxide). During

the dropwise introduction of sulfuric acid, the developed HCl(g) was transported through the filter with the help of a soft

nitrogen gas flow. The experiment resulted in a color change of the powder product, but successive 1H NMR analysis

shows the intact isopropyl xanthate chain and only a shifting of one of the aminic protons to high-field, probably due to

the lower shielding caused by protonation (Figure 21, Spectrum 6).

H

i

A

C B

D

G

N

O

N

O

E’ or F’

E or F

E’ or F’

Page 46: epub.ub.uni-greifswald.de · Table of contents 1. Pteridines and pterins

42

FIGURE 21 – PROTONATION OF THE AMINO FUNCTION WITH HYDROCHLORIC ACID GAS

10.0 9.5 9.0 8.5 8.0 7.5 7.0 6.5 6.0 5.5 5.0 4.5 4.0 3.5 3.0 2.5 2.0 1.5 1.0 0.5

Chemical Shift (ppm)

0

0.05

0.10

0.15

0.20

0.25

0.30

0.35

0.40

0.45

0.50

0.55

0.60

0.65

0.70

0.75

0.80

0.85

0.90

0.95

1.00

No

rma

lize

d In

ten

sity

3.053.183.524.142.023.122.092.091.982.040.880.93

0.9

30

.95

0.9

81

.20

1.3

01

.32

1.3

41

.47

1.4

91

.50

1.5

21.9

31.9

61

.98

2.0

0

2.8

2

3.6

93

.71

3.7

43

.85

3.8

73

.90

4.7

04

.72

4.7

54

.90

7.7

2

9.8

2

Spectrum 6 – 1H NMR of protonation of the amino group by reaction with hydrochloric acid in gas form(Experimental section, B’-

Carbamate derivative protonated) 1H NMR (300MHz ,CHLOROFORM-d) δ = 9.82 (br. s., 1 H), 7.72 (br. s., 1 H), 4.90 (s, 2 H), 4.72 (s, 2 H), 3.86 (q, J = 6.9 Hz, 2 H), 3.73 (q,

J = 6.7 Hz, 2 H), 2.82 (s, 3 H), 1.96 (quin, J = 6.8 Hz, 2 H), 1.57 - 1.37 (m, 4 H), 1.32 (t, J = 7.0 Hz, 3 H), 1.18 (t, J = 7.2 Hz, 3 H), 1.02 - 0.89 (m, 3 H) ppm

In order to avoid protonation, the amino group was protected by a mixture of acetic acid and acetic anhydride

(AcOH/AcO2), a typical method for pterins and previously described in different pubblications [93]. After three hours of

refluxing the reaction mixture, two products were isolated. The first, a white solid was characterized by 1H NMR and

recognised to be the protected form of the starting material by the formation of the CH3 signal M and the shifting of

one of the aminic protons H’ (Spectrum 7).

The second product, a deep red solid, shows in the 1H NMR the protection of the amino group, the disappearance of

the xanthogenate unit and, most importantly a singlet signal P, at 7.78 ppm, that was first wrongly assigned to a proton

of the dithiolene ring (Spectrum 8).

The hydrolysis of the protecting group in the red powder product was carried out in methanol at room temperature

overnight (experimental section- red solid). As aspected the amino group signal H reappeared around 5.7 ppm and the

methyl group of the protection disappeared (Spectrum 9). Aside the formation of a small broad signal at 2.04 ppm was

observed and initially assigned to some impurities due to variation of the integration values. Later, the position,

stabilized integration of 1 and the characteristic broadness of this new signal was ascribed to a hydroxy or thioxy

functionality and the appearance over time was attributed to the general instability or reactivity of the targeted product

(Spectrum 9).

H

H

A

C

B

D

G

N

O

E or F

E’ or F’

8.3 8.2 8.1 8.0 7.9 7.8 7.7 7.6 7.5 7.4 7.3 7.2 7.1 7.0 6.9

Chemical Shift (ppm)

2.00

7.6

5

7.7

0

7.7

7

Page 47: epub.ub.uni-greifswald.de · Table of contents 1. Pteridines and pterins

43

11.0 10.5 10.0 9.5 9.0 8.5 8.0 7.5 7.0 6.5 6.0 5.5 5.0 4.5 4.0 3.5 3.0 2.5 2.0 1.5 1.0 0.5

Chemical Shift (ppm)

0

0.05

0.10

0.15

0.20

0.25

0.30

0.35

0.40

0.45

0.50

0.55

0.60

0.65

0.70

0.75

0.80

0.85

0.90

0.95

1.00N

orm

aliz

ed

Inte

nsi

ty

3.006.104.342.013.012.821.961.941.000.95

DMSO

Water

0.8

80

.91

0.9

3

1.3

01

.32

1.4

31

.45

1.4

6

1.8

61

.88

1.9

11

.93

2.3

5

2.7

1

4.6

14

.63

4.6

5

4.9

8

5.5

45.5

65.5

85.6

05

.62

5.6

45

.66

10

.92

Spectrum 7 – 1H NMR of protection of the amino group, first product isolated (experimental section, B’ - white solid) 1H NMR (300MHz ,DMSO-d6) δ = 10.92 (s, 1 H), 5.60 (spt, J = 6.2 Hz, 1 H), 4.98 (s, 2 H), 4.63 (t, J = 6.6 Hz, 2 H), 2.71 (s, 3 H), 2.35 (s, 3

H), 1.88 (quin, J = 6.9 Hz, 2 H), 1.51 - 1.36 (m, 4 H), 1.32 (s, 3 H), 1.30 (s, 3 H), 0.94 - 0.88 (m, 3 H) ppm

8.0 7.5 7.0 6.5 6.0 5.5 5.0 4.5 4.0 3.5 3.0 2.5 2.0 1.5 1.0 0.5

Chemical Shift (ppm)

0

0.05

0.10

0.15

0.20

0.25

0.30

0.35

0.40

0.45

0.50

0.55

0.60

0.65

0.70

0.75

0.80

0.85

0.90

0.95

1.00

No

rma

lize

d In

ten

sity

3.104.382.153.003.012.091.001.01

0.9

30

.95

0.9

7

1.3

81.4

01

.46

1.4

71

.48

1.5

0

1.8

61.8

81

.91

2.6

62

.68

4.4

84

.50

4.5

2

7.2

67.7

8

8.1

6

Spectrum 8 – 1H NMR of protection of the amino group, second product isolated (experimental section, C’ - red solid) 1H NMR (300MHz ,CHLOROFORM-d) δ = 8.16 (s, 1 H), 7.78 (s, 1 H), 4.50 (t, J = 6.8 Hz, 2 H), 2.68 (s, 3 H), 2.66 (s, 3 H),

1.95 - 1.83 (m, 2 H), 1.53 - 1.36 (m, 4 H), 1.03 - 0.87 (m, 3 H) ppm

L

H’

I

i

M

A

C

B

D

G

E or F

I

M

H’

A

C

B

D

G

P

E or F

E’ or F’

H’

M

H’

M

P

Page 48: epub.ub.uni-greifswald.de · Table of contents 1. Pteridines and pterins

44

7.5 7.0 6.5 6.0 5.5 5.0 4.5 4.0 3.5 3.0 2.5 2.0 1.5 1.0 0.5

Chemical Shift (ppm)

0

0.05

0.10

0.15

0.20

0.25

0.30

0.35

0.40

0.45

0.50

0.55

0.60

0.65

0.70

0.75

0.80

0.85

0.90

0.95

1.00

No

rma

lize

d In

ten

sity

3.034.062.021.062.952.001.810.96

0.9

20

.95

0.9

7

1.3

81.4

21

.44

1.4

51

.46

1.4

71

.81

1.8

31.8

61

.88

2.0

4

2.6

2

4.4

14

.43

4.4

6

5.7

1

7.7

0

Spectrum 9 – 1H NMR of hydrolysis of protection, second product isolated (experimental section - C’)

1H NMR (300MHz ,CHLOROFORM-d) δ = 7.70 (s, 1 H), 5.71 (br. s., 2 H), 4.43 (t, J = 6.8 Hz, 2 H), 2.62 (s, 3 H),

1.86 (quin, J = 7.1 Hz, 2 H), 1.52 - 1.38 (m, 4 H), 0.99 - 0.90 (m, 3 H) ppm

The elemental analysis was partially fitting except for the sulfur content, which was lower than expected. Also, the

deprotected derivative obtained by hydrolysis with hydrochloric acid, exhibited the same decrease in sulfur content

(Scheme 28, Table 2, experimental section, C’- C’ without pentyloxy chain).

TABLE 2 – ELEMENTAL ANALYSIS OF THE EXPECTED PRODUCT (THEORETICAL) AND OF THE ISOLATED PRODUCT C’ AND THE DEPROTECTED FORM

Mass spectrometry showed a peak at 348.3 m/z and not the expected 364(H+) m/z. Nevertheless, considering possible

fragmentation and the basic instability of the compound, the achievment of the targeted structure could not be

excluded and the complexation reaction with K3Na[(MoO2)(CN)4] was tried as shown in Scheme 29.

Red powder h. N C H S

Theoretical 19.27 49.57 4.71 17.64

Experimental 19.54 51.57 4.68 9.19

Expected C’ dep. N C H S

Theoretical 23.88 40.95 2.41 21.86

Experimental 21.90 41.27 3.22 10.06

A

C

B

D

G

H

– CH – OH

– SH

E or F

– CH

SCHEME 28 – ABOVE DEPROTECTION WITH HYDROCHLORIC ACID(YIELD 42%)

Page 49: epub.ub.uni-greifswald.de · Table of contents 1. Pteridines and pterins

45

SCHEME 29 – COMPLEXATION REACTION WITH THE ISOLATED PRODUCT

TABLE 3 – POSSIBLE ELEMENTAL ANALYSIS OF THE COMBINATION WITH THE COUNTER CATIONS

The protocol for the complexation with molybdenum contemplates a final purification step of the dianionic product.

This can be achieved by exchange of the counter cations with tetraphenylphosphonium chloride, by precipitation with

crown ether or by extraction with acetonitrile in case of enough solubility in organic solvent. Employment of all these

methods did not give any results and the presence of inorganic salts, mainly cyanide, was always detected by IR

spectroscopy. The best result was obtained by washing the inorganic salts with a small amount of methanol

(experimental section, C’- complexation product).

The NMR spectra did not give interpretable results and the elemental analysis, coherently with the previous step,

showed a lower content of sulfur. The IR spectrum shows two bands in the range of the double bond of the dithiolene

C=C at 1506 cm-1 and the characteristic Mo=O at 835 cm-1 typical of the bis-dithiolene complexes [75] (Spectrum 10).

Complexation N C H S

theoretical with 2 Na+

20.34 31.40 2.05 18.63

theoretical with 2 K+

19.43 29.99 1.96 17.80

experimental 18.91 32.11 2.73 4.78

SPECTRUM 10 – IR OF THE COMPLEXATION PRODUCT

Page 50: epub.ub.uni-greifswald.de · Table of contents 1. Pteridines and pterins

46

An explanation of the collected data, incongruent with the synthetical aim, arises from two crystal structures obtain

from the complexation mixture and from the red powder product after reaction with the acetic acid and acetic

anhydride mixture (Figure 23, 23).

FIGURE 23 – COLOURLESS CRYSTALS FROM COMPLEXATION MIXTURE

Both structures may result from the decomposition of the desired product. However the presence of a totally intact

methyl group on position 6 of the pterin, points toward a failure already during the bromination reaction. Most

probably, the reaction has accurred on the methyl group at position 7 of the pterin, and this apparently with a high

selectivity since no other side products were isolated or observed. Therefore this 6-acyl pterin can aparently not be used

as a substrate in this specific protocol and the high selectivity for the 7 methyl position over the α-position must be

taken into account for future work. Further attempts to brominate the α-carbonyl position with bromine (and acetic

acid as catalyst) leads to the cleavage of the protecting group and it was eventually not contemplated as a possible

solution any further.

A comprehensive explanation of the side reaction can be derived from a study reported in 2004 by A. Kalinin and V. A.

Mamedov for the synthesis of thiazole-quinoxaline from chloroderivatives with the use of carbon disulfide [94]. The

group observed the formation of thiazole C by the reaction of the quinaxoline-chloride substrate in basic condition in

presence of carbon disulfide (Scheme 30)

This reaction is similar to a Cook–Heilbron synthesis [95] and has been postulated to go through a xanthogenate

derivative in the reported work. Therefore, in order to confirm their hypothesis, they synthesized product B and

recovered the same product C with an acid catalysed ring closing reaction (Scheme 30)

Notably the applied reaction conditions were very similar to our protection protocol (AcOH/Ac2O at reflux). Even

through TFA (trifluoroacetic acid) is far stronger than acetic acid, the prolonged refluxing time may eventually lead to

the same product. It is, thus, possible to propose a similar pathway (Scheme 31).

FIGURE 22 – CRYSTAL STRUCTURE FROM THE RED POWDER

Page 51: epub.ub.uni-greifswald.de · Table of contents 1. Pteridines and pterins

47

SCHEME 30 – SYNTHESIS OF THIAZOLE-QUINOXALINE [94]

As depicted in Scheme 31, the first step presumably is the bromination of the methyl group at 7 position (instead of the

α-position) and formation of the product A’ followed by substitution with isopropyl xanthate that leads to product B’.

Finally the reflux in acetic acid reults in the formation of the five membered cyclic thiazole between position 7 and 8.

SCHEME 31 – PROPOSED MECHANISM AS DERIVED FROM THE SYNTHESIS OF THIAZOLE QUINOXALINE

Page 52: epub.ub.uni-greifswald.de · Table of contents 1. Pteridines and pterins

48

The proposed final product C’ is in perfect accordance with the elemental analysis of the C’, including the sulfur content (Table 4).

TABLE 4 – ELEMENTAL ANALYSIS OF PRODUCT C’

Also MS-APCI spectrometry shows the exact mass peak and a fitting fragmentation, i.e. corrisponding to a loss of the

pentyloxy chain , as observed for the majority of the 6-acyl pterins synthesized in this dissertation (Figure 24).

FIGURE 24 – MASS SPECTRA OF THE PRODUCT C’ POSITIVE AND NEGATIVE MODUS

Product-C’ N C H S

Theoretical 20.16 51.86 4.93 9.23

Experimental 19.54 51.57 4.68 9.19

348.3 [M + H]+

278.2 [M1 + H]+

[M1] = [M] =

346.2 [M - H]-

Page 53: epub.ub.uni-greifswald.de · Table of contents 1. Pteridines and pterins

49

Also the 1H and 13C NMR are in good accordance with the proposed structure of C’ (Spectra 11, 12, 13)

7.5 7.0 6.5 6.0 5.5 5.0 4.5 4.0 3.5 3.0 2.5 2.0 1.5 1.0 0.5

Chemical Shift (ppm)

0

0.05

0.10

0.15

0.20

0.25

0.30

0.35

0.40

0.45

0.50

0.55

0.60

0.65

0.70

0.75

0.80

0.85

0.90

0.95

1.00

No

rma

lize

d In

ten

sity

3.034.062.021.062.952.001.810.96

0.9

20

.95

0.9

7

1.3

81.4

21

.44

1.4

51

.46

1.4

71

.81

1.8

31.8

61

.88

2.0

4

2.6

2

4.4

14

.43

4.4

6

5.7

1

7.7

0

Spectrum 11 – 1H NMR product C’

1H NMR (300MHz ,CHLOROFORM-d) δ = 7.70 (s, 1 H), 5.71 (br. s., 2 H), 4.43 (t, J = 6.8 Hz, 2 H), 2.62 (s, 3 H), 1.86 (quin, J = 7.1 Hz, 2 H), 1.52 - 1.38 (m, 4 H), 0.99 - 0.90 (m, 3 H) ppm

200 195 190 185 180 175 170 165 160 155 150 145 140 135 130 125 120 115 110

Chemical Shift (ppm)

0

0.05

0.10

0.15

0.20

0.25

0.30

0.35

0.40

0.45

0.50

0.55

0.60

0.65

0.70

0.75

0.80

0.85

No

rma

lize

d In

ten

sity

10

8.2

9

12

4.9

5

14

1.4

8

14

9.4

1

16

2.0

0

16

6.1

71

67

.05

19

8.0

1

Spectrum 12 – 13C NMR of C’ quaternary carbons only

13C NMR (75MHz ,CHLOROFORM-d) δ = 198.0, 167.0, 166.2, 162.0, 149.4, 141.5, 125.0, 108.3 ppm

D

A

C B

D

E

F

G

A

B

G

E F

H H

C

C=O

C-2

C-8a C- 4

C-6

C-7

C-4a

S-C=O

C=O

S-C=O

Page 54: epub.ub.uni-greifswald.de · Table of contents 1. Pteridines and pterins

50

105 100 95 90 85 80 75 70 65 60 55 50 45 40 35 30 25 20 15 10

Chemical Shift (ppm)

0

0.05

0.10

0.15

0.20

0.25

0.30

0.35

0.40

0.45

0.50

0.55

0.60

0.65

0.70

0.75

0.80

0.85

0.90

0.95

1.00

No

rma

lize

d In

ten

sity

14

.00

22

.35

25

.65

28

.08

28

.26

67

.94

76

.57

76

.997

7.4

1

10

3.3

5

105 100 95 90 85 80 75 70 65 60 55 50 45 40 35 30 25 20 15 10

Chemical Shift (ppm)

-1.0

-0.9

-0.8

-0.7

-0.6

-0.5

-0.4

-0.3

-0.2

-0.1

0

0.1

0.2

0.3

0.4

0.5

0.6

0.7

0.8

No

rma

lize

d In

ten

sity

13

.99

22

.33

25

.65

28

.06

28

.25

67

.92

10

3.3

4

Spectrum 13 – 13C NMR, DEPT 135, product C’

13C NMR (75 MHz): δ = 14.0 (CH3), 22.3 (CH2), 25.7 (CH3), 28.1 (CH2), 28.2 (CH2), 67.9 (CH2), 103.3 (CH)ppm

The 13C NMR spectrum, as previously mentioned, shows an overlap of signals β and γ carbon atoms of two CH2 groups

of the pentyloxy chain. Taking into account the proposted structure of C’ the signal at 25.65 ppm is assigned to CH3-C=O

(Spectrum 13).

Product B’ is a constitutional isomer of the expected product. Therefore the elemental analysis and the mass peak are

the same. However, some interesting differences can be observed between the carbamate and the xanthogenate

derivatives. The spectrum with the quaternary carbons shows the appearance of a peak at 212.5 ppm of the thio

carbonyl S–C=S and the shifting of C–7 to high field from 163 ppm to 157 ppm, according to the reaction occurring close

to this position (Spectrum 15).

β

γ

α δ

ε

β

δ

α

γ

ε

C-H

CH3-C=O

CH3-C=O

Page 55: epub.ub.uni-greifswald.de · Table of contents 1. Pteridines and pterins

51

8.5 8.0 7.5 7.0 6.5 6.0 5.5 5.0 4.5 4.0 3.5 3.0 2.5 2.0 1.5 1.0

Chemical Shift (ppm)

0

0.05

0.10

0.15

0.20

0.25

0.30

0.35

0.40

0.45

0.50

0.55

0.60

0.65

0.70

0.75

0.80

0.85

0.90

0.95

1.00

No

rma

lize

d In

ten

sity

3.016.114.041.992.612.002.011.042.00

DMSO

Water

0.8

80

.90

0.9

3

1.2

91

.31

1.4

11

.42

1.4

6

1.7

91

.821.8

41

.86

1.8

9

2.6

4

4.4

94

.51

4.5

3

4.9

0

5.5

45.5

65

.58

5.6

05

.62

5.6

45

.677

.68

7.8

0

7.9

3

8.0

6

Spectrum 14 – 1H NMR of product B’ 1H NMR (DMSO-d6) δ = 7.80 (br. s., 1 H), 7.68 (br. s., 1 H), 5.60 (td, J = 6.3, 12.3 Hz, 1 H), 4.90 (s, 2 H), 4.51

(t, J = 6.6 Hz,2 H), 2.64 (s, 3 H), 1.84 (quin, J = 6.9 Hz, 2 H), 1.52 - 1.35 (m, 4 H), 1.30 (d, J = 6.4 Hz, 7 H), 0.94 - 0.85 (m, 3 H) ppm

210 205 200 195 190 185 180 175 170 165 160 155 150 145 140 135 130 125 120 115

Chemical Shift (ppm)

0

0.05

0.10

0.15

0.20

0.25

0.30

0.35

0.40

0.45

No

rma

lize

d In

ten

sity

12

0.3

9

13

9.8

6

15

6.8

615

7.1

6

16

3.3

8

16

6.7

6

19

8.9

3

21

2.5

0

Spectrum 15 – 13C NMR of B’, quaternay carbons only

13C NMR (DMSO-d6) δ = 212.5, 198.9, 166.8, 163.4, 157.2, 156.9, 139.9, 120.4 ppm

L

I

E

L

I

E

F’

F’ A

B

C

D

G

H

A C

B

D

G

H

C – 4a

C=O C – 2

C – 8A

C – 4 C – 6

C – 7

C=S

C=S

C=O

Page 56: epub.ub.uni-greifswald.de · Table of contents 1. Pteridines and pterins

52

110 105 100 95 90 85 80 75 70 65 60 55 50 45 40 35 30 25 20 15

Chemical Shift (ppm)

0

0.05

0.10

0.15

0.20

0.25

0.30

0.35

0.40

0.45

0.50

0.55

0.60

0.65

0.70

0.75

0.80

0.85

0.90

0.95

1.00

No

rma

lize

d In

ten

sity

13

.88

20

.88

21

.80

26

.79

27

.61

67

.56

78

.29

110 105 100 95 90 85 80 75 70 65 60 55 50 45 40 35 30 25 20 15

Chemical Shift (ppm)

-0.9

-0.8

-0.7

-0.6

-0.5

-0.4

-0.3

-0.2

-0.1

0

0.1

0.2

0.3

0.4

0.5

0.6

0.7

0.8

0.9

1.0

No

rma

lize

d In

ten

sity

13

.88

20

.88

21

.79

26

.79

27

.60

40

.22

67

.56

78

.29

Spectrum 16 – 13C NMR DEPT 135 of B’

13C NMR (DMSO-d6) δ = 13.9 (CH3), 20.9 (CH3, CH3), 21.8 (CH2), 26.8 (CH3), 27.6 (CH2, CH2), 40.2 (CH2), 67.6 (CH2), 78.3 (CH) ppm

The 13C NMR and DEPT 135 experiments support the assignment of the CH3 -C=O moiety similar to the structure of C’

at 26.70 ppm. Also, the S-CH2 carbon atoms can be ascribed to the signal at 40.22 ppm (Spectrum 16).

CH3 -C=O

ε

Xa-(CH3)2

Xa-CH

Xa-(CH3)2

Xa-CH

S-CH2

δ β σ

γ

S-CH2

ε

δ

β γ

α

CH3 -C=O

Page 57: epub.ub.uni-greifswald.de · Table of contents 1. Pteridines and pterins

53

Following this hypothesis also the carbamate derivative can be identified unambiguously by its carbon and proton NMR (Spectra 17, 18, 19).

10.5 10.0 9.5 9.0 8.5 8.0 7.5 7.0 6.5 6.0 5.5 5.0 4.5 4.0 3.5 3.0 2.5 2.0 1.5 1.0 0.5

Chemical Shift (ppm)

0

0.05

0.10

0.15

0.20

0.25

0.30

0.35

0.40

0.45

0.50

0.55

0.60

0.65

0.70

0.75

0.80

0.85

0.90

0.95

1.00

No

rma

lize

d In

ten

sity

3.143.283.194.012.042.562.202.392.032.172.04

DMSO

Water

0.8

80

.90

0.9

31

.12

1.1

41

.24

1.2

61

.38

1.4

21

.44

1.4

61.8

21.8

41

.86

2.6

4

3.7

53

.77

3.9

03

.93

4.4

84

.51

4.5

3

4.9

9

7.6

57

.70

7.7

8

Spectrum 17 – 1H NMR of carbamate derivative 1H NMR (300MHz ,DMSO-d6) δ = 7.65 (s, 1 H), 7.70 (s, 1 H), 4.99 (s, 2 H), 4.50 (t, J = 6.6 Hz, 2 H), 3.91 (q, J = 6.5 Hz, 2 H), 3.76 (q, J = 6.9 Hz, 2 H), 2.64 (s, 3 H), 1.84 (quin, J = 6.9 Hz, 2 H), 1.53 - 1.33 (m, 4 H), 1.24 (t, J = 7.0 Hz, 3 H), 1.20 - 1.09 (m, 3 H), 0.97 - 0.85 (m,

3 H) ppm

200 195 190 185 180 175 170 165 160 155 150 145 140 135 130 125 120 115

Chemical Shift (ppm)

0

0.05

0.10

0.15

0.20

0.25

0.30

0.35

0.40

0.45

No

rma

lize

d In

ten

sity

11

9.8

6

14

1.1

5

15

6.8

81

58

.01

16

3.1

6

16

6.7

6

19

3.6

3

19

8.9

7

Spectrum 18 – 13C NMR of carbamate derivative, quaternary carbons

13C NMR (75MHz ,DMSO-d6) δ = 199.0, 193.6, 166.8, 163.2, 158.0, 156.9, 141.1, 119.9 ppm

N

H

i

A

C

B

D

G

N

O

F’

E

F’

O

E

C-4a C=O

C-2 C-8A

C-4 C-6 C-7 N-C=S

N-C=S

C=O

Page 58: epub.ub.uni-greifswald.de · Table of contents 1. Pteridines and pterins

54

The quaternary carbon spectrum reveals a similar situation with the C-7 shifted to 158 ppm. The N-C=S of the carbamate

unit, appears at higher field compared to the other derivative, probably because of the inductive effect of the two alkyl

substituents and the lower electronegativity of nitrogen compared to oxygen (Spectrum 18).

The carbamate derivative shows a similar S-CH2 signal as the xanthate and the signals which belong to Car-(CH2)2 and

Car-(CH3)2 (Spectrum 19).

110 105 100 95 90 85 80 75 70 65 60 55 50 45 40 35 30 25 20 15 10

Chemical Shift (ppm)

0

0.05

0.10

0.15

0.20

0.25

0.30

0.35

0.40

0.45

0.50

0.55

0.60

0.65

0.70

0.75

0.80

0.85

0.90

0.95

1.00

No

rma

lize

d In

ten

sity

11

.31

12

.3813

.87

21

.79

26

.902

7.6

1

41

.79

46

.65

49

.04

67

.52

110 105 100 95 90 85 80 75 70 65 60 55 50 45 40 35 30 25 20 15 10

Chemical Shift (ppm)

-1.0

-0.9

-0.8

-0.7

-0.6

-0.5

-0.4

-0.3

-0.2

-0.1

0

0.1

0.2

0.3

0.4

0.5

0.6

No

rma

lize

d In

ten

sity

11

.30

12

.38

13

.88

21

.80

26

.91

27

.60

41

.79

46

.64

49

.03

67

.51

Spectrum 19 – 13C NMR DEPT 135 of carbamate derivative 13C NMR (75MHz ,DMSO-d6) δ= 11.3 (CH3), 12.4 (CH3), 13.9 (CH3), 21.8 (CH2), 26.9 (CH3), 27.6 (CH2, CH2), 41.8 (CH2),

46.6 (CH2), 49.0 (CH2), 67.5 (CH2) ppm

Car-(CH3)2

Car-(CH2)

CH3 -C=O

ε – CH3

γ

σ S-CH2

δ

β

Car-(CH3)2

Car-(CH2)2

S-CH2

CH3 -C=O

ε

δ

γ

β

α

Page 59: epub.ub.uni-greifswald.de · Table of contents 1. Pteridines and pterins

55

The signal at 2.04 ppm, in the 1H NMR of C’, appearing over time may arise from S-oxidation, which occurs in solution

and results in the formation of the sulfur bearing an -OH functionality. This is, in fact, a possible mechanism leading to

the chemical structure shown in Scheme 32, accompanied by the re-aromatization of the pyrazine ring. Keeping the

product in solution with methanol or chloroform for a prolonged time does not change the MS peak and no other signals

arise, indicating a chemical species not stable enough to be observed or the presence of an equilibrium only in solution.

Possibly by a light driven mechanism during data collection with the diffractometer or due to long exposure to light a

transformation to species E’ occured. This would be in accordance with pterins being natural photosensitizer [25].

SCHEME 32 – POSSIBLE STEPS OF S-OXIDATION AND ACTUAL RESULT OF THE COMPLEXATION ATTEMPT

Intrestingly, a similar reaction is described as first step in the metabolism of the thiazolidinedione (TZD) ring, catalyzed

by cytochrome P450 [96, 97]. The employment of this structure as model compounds represents an interesting research

opportunity.

Refering to structures C and C’, respectively, the presence of a phenyl substituent, a typical electron-pushing group,

instead of a proton plays most probably a role in the stabilization of the thioazole ring. Indeed, the distinct aromaticity

of the quinoxaline compare to the pteridine rings must be taken into account.

The attempted complexation reaction in – basic condition under the false presumption that the targeted ligand

precursor had been made, leads to the formation of product F’ by hydrolisis of the nitrogen at postion 8 and formation

of a new five membered ring by the attack of sulfur on carbons (Scheme 32). Similar structure arises from the oxidation

of the natural Moco [98] and they are quite common in other research work on pterins [99]. Further speculations

regarding the mechanism of this side reaction would require more in depht-studies, which are not inherent to the main

goal of this dissertation. Therefore it was decided to proceed to the synthesis of other 6-acyl pterins by the acylation

protocol described in the following. Beside the formation of colorless crystal of F’ a brownish powder was isolated G’

(experimental section,C’- complexation product, code IT 114, Scheme 32). Interestingly, its incubation with the protein

apoTor A indicates in the respective UV spectrum the formation of specific binding compared to the unspecific binding

Page 60: epub.ub.uni-greifswald.de · Table of contents 1. Pteridines and pterins

56

on the external protein suface developed with BSA (bovine serum albumin). The content of molybdenum measured by

ICP-OES was close to zero, meaning that the species effectively bound to the protein was most probably neither a

complex nor molybdate.

This observation does not prove any specific association between pterin and protein. Nevertheless, the particularity of

this result should be emphasized, i.e it was not observed with many other model compounds as of yet. In this context

it is important to consider the purification process after the incubation with protein, which is a size exclusion

chromatography as shown in Figure 26. With this method the molecules are separated by size and if the product binds

to the protein they are collected in the same fraction; therefore it is possible to assume the formation of a kind of

interaction between the protein and the not unambiguously identified pterin product, which most likely bears free

amino and amide fuctionalities as shown in the crystal structure of F’ (Scheme 32). During the experiment the product

has shown signs of instability, which suggests a certain sensitivity towards oxygen and light. This should be taken into

account for future studies.

300 400 500 600

0

400

800

1200

1600

Ab

sorp

tio

n, a

.u.

Wavelength, nm

IT 114 with apoTorA

IT 114 with BSA

FIGURE 25 – UV STUDIES AFTER INCUBATION WITH BSA AND APOTOR A

FIGURE 26 - GEL FILTRATION, SIZE-EXCLUSION

Page 61: epub.ub.uni-greifswald.de · Table of contents 1. Pteridines and pterins

57

3.4 Synthesis of pterin-dithiolene ligands through the “Minisci reaction”

The particular π-deficient character of heteroaromatic bases was already described above In the chapter “Pteridines

and Pterins”. Pteridines are a prototype of this electronic condition and possess, hence, a characteristic chemical

behaviour compared to other aromatic cycles. The high reactivity toward nucleophilic reagents is the best example of

this characteristic reactivity and the covalent hydration a direct consequence [100].

The presence of electron donating substituents on the pteridine ring like amino and hydroxy group does not change this

behaviour; therefore, subclasses like pterins and lumazines maintain this reactivity and the nucleophilic attack remains

a powerful method for their derivatization. The protonation of heteroaromatic bases enhances this reactivity, but

usually most of the nucleophiles act as deprotonating agent instead of proceeding to substitution. This inconvenience

does not occur if the nucleophile is a radical species and exactly in this perimeter plays the so called “Minisci reaction”,

a chemical protocol that has been developed mainly for C-C bond formation [101].

This reaction has various interesting aspects — high reactivity rates, good selectivities and specular synthetical

applications as the Friedel-Craft reaction [102]. The reaction mechanism in Scheme 33 can be divided in three parts —

the formation of the radicals (for example with a Fenton reaction [103]), the addition to the heteroaromatic ring and

the re-aromatization. While the first two transformations are quite easily understood, the final step has not been clearly

deciphered [104].

SCHEME 33 – HOMOLYTIC ACYLATION MECHANISM

Page 62: epub.ub.uni-greifswald.de · Table of contents 1. Pteridines and pterins

58

The nucleophilic character of the acyl radicals is due to the stability of its cations [105] and it influences the reaction

rate together with the so called polar effect [106], namely the high reactivity increment due to the presence of an

electron withdrawing group or in the case reported above the protonation of the substrate [104]. The synthetic interest

for this reaction is also related to its selectivity; in fact, the presence of other activated positions does not represent an

issue, since the introduction of the first acyl activates further the substrate but also decreases the basic character of the

same changing the condition for the protonation.

The reaction has the same synthetic capability as the Friedel-Crafts aromatic alkylation but with opposite reactivity and

selectivity [102, 106]. All electrophilic species used in the aromatic electrophilic substitution are suitable, as

corresponding radicals, for the homolytic acylation, since more stable carbonium ions give more nucleophilic radicals

[101, 102].

Pterins have been used as substrate for this protocol in a few examples in the literature, in particular by W. Pfleiderer

in the chemical synthesis of deoxysepiapterin [35] (Scheme 34) and the derivatization of Lumazine [18]; also in the book

“Fused Pyrimidines” the reaction is described as not completely explored on pteridines with a stress for wider future

employment [11].

SCHEME 34 – CLOSELY FOLLOWING THE PROCEDURE INTRODUCED BY W. PFLEIDERER FOR THE SYNTHESIS OF 6-ACYL PTERIN

Page 63: epub.ub.uni-greifswald.de · Table of contents 1. Pteridines and pterins

59

Among a large number of methods for the generation of acyl radicals W. Pfleiderer used a Fenton reagent with TBHP

(tert-Butyl hydroperoxide solution 70%), iron sulfate, 17 equivalents of aldehyde (propionaldehyde) and as solvent a 3:1

acetic/water mixture [107]. Product 6 is the same as used before for the synthesis of the 6 chloro derivative 10 with the

pentyloxy chain as protection.

The acylation reaction is particularly facile and, as shown in Scheme 34 requires only 2 minutes of reaction time. The

quenching and isolation of the product is carried out by adding water and consequent precipitation of the pterin

substrate. In most of the cases no further purification is necessary except for washing with water and n-hexane or

recrystallization from a methanol and water mixture. The synthetic work of W. Pfleiderer describes also other pterin

substrates and aldehydes with consequent variation of the reaction conditions [35]. Nevertheless the work-up and

general procedure remain simple and do not require particular modifications [35]. The main aim of this dissertation is

only indirectly connected to this protocol, therefore the reaction parameters have not been further optimized for the

synthesis of the other 6-acyl pterins reported in this work.

Reliable crystallographic data was obtained of the intermediates 14, 15 and 16 (Figure 27, 28, 29); the orientation of

the carbonyl is also in this molecular structures toward the substituent at position 7, exactly as for the previously

described product 11.

FIGURE 27– MOLECULAR STRUCTURE OF INTERMEDIATE 14

FIGURE 28 – MOLECULAR STRUCTURE OF INTERMEDIATE 15

Page 64: epub.ub.uni-greifswald.de · Table of contents 1. Pteridines and pterins

60

The reaction using acetyl aldehyde as reactant also gave with good yield the expected product 17, which shows the

same orientation of the carbonyl like in the previous molecular structures above. This derivative was synthesized in

order to appreciate the possible differences in stability of dithiolene-pterin ligands or complexes, related to the variation

in steric hindrance of the dithiolene substituents (Scheme 35). This aspect requires further engagement in this topic.

FIGURE 29 – MOLECULAR STRUCTURE OF INTERMEDIATE 16

SCHEME 35 – ACYLATION REACTION WITH ACETYL ALDEHYDE

FIGURE 30 – MOLECULAR STRUCTURE PRODUCT 17

Page 65: epub.ub.uni-greifswald.de · Table of contents 1. Pteridines and pterins

61

The signals in the NMR spectra of product 17 are assigned by comparison with those of 16, which are already reported

in the literature (Spectra 20, 21, 22).

7.5 7.0 6.5 6.0 5.5 5.0 4.5 4.0 3.5 3.0 2.5 2.0 1.5 1.0 0.5

Chemical Shift (ppm)

0

0.05

0.10

0.15

0.20

0.25

0.30

0.35

0.40

0.45

0.50

0.55

0.60

0.65

0.70

0.75

0.80

0.85

0.90

0.95

1.00

No

rma

lize

d In

ten

sity

3.063.054.131.252.152.002.942.002.011.80

0.9

1

1.0

4

1.3

91.4

11

.44

1.4

8

1.6

31

.70

1.7

31

.75

1.8

51

.87

1.9

01

.92

2.6

9

3.1

83.2

13

.23

4.4

74

.49

4.5

1

5.6

8

7.2

2

Spectrum 20 – 1H NMR of product 17

1H NMR (300MHz ,CHLOROFORM-d) δ = 5.68 (br. s., 1 H), 4.49 (t, J = 6.8 Hz, 2 H), 3.27 - 3.11 (m, 2 H), 2.69 (s, 3 H), 1.86 (d, J = 7.2

Hz, 2 H), 1.81 - 1.65 (m, 2 H), 1.52 - 1.31 (m, 4 H), 1.04 (t, J = 7.4 Hz, 3 H), 0.98 - 0.81 (m, 3 H) ppm

200 195 190 185 180 175 170 165 160 155 150 145 140 135 130 125 120 115

Chemical Shift (ppm)

0

0.05

0.10

0.15

0.20

0.25

0.30

0.35

0.40

0.45

0.50

0.55

0.60

0.65

0.70

0.75

0.80

0.85

0.90

0.95

1.00

No

rma

lize

d In

ten

sity

11

7.7

3

14

0.1

1

15

6.5

116

3.2

6

16

5.5

5

16

8.1

9

19

8.7

8

Spectrum 21 – 13C NMR of product 17, quaternary carbons 13C NMR (75MHz ,CHLOROFORM-d) δ = 198.8, 168.2, 165.5, 163.3, 156.5, 140.1, 117.7 ppm

E

C

H

I

L

C – D

G

L

F

E

I

A

C

B

C

C

C

D

C

F

C

G

A

C

B

C

H

C=O C-2 C-8A

C-4 C-6 C-7

C-4a

C=O

Page 66: epub.ub.uni-greifswald.de · Table of contents 1. Pteridines and pterins

62

110 105 100 95 90 85 80 75 70 65 60 55 50 45 40 35 30 25 20 15 10

Chemical Shift (ppm)

0

0.05

0.10

0.15

0.20

0.25

0.30

0.35

0.40

0.45

0.50

0.55

0.60

0.65

0.70

0.75

0.80

0.85

0.90

0.95

1.00

No

rma

lize

d In

ten

sity

13

.77

13

.98

21

.31

22

.33

26

.43

28

.08

28

.11

32

.32

68

.46

110 105 100 95 90 85 80 75 70 65 60 55 50 45 40 35 30 25 20 15 10

Chemical Shift (ppm)

-1.0

-0.9

-0.8

-0.7

-0.6

-0.5

-0.4

-0.3

-0.2

-0.1

0

0.1

0.2

0.3

0.4

0.5

0.6

0.7

0.8

0.9

No

rma

lize

d In

ten

sity

13

.77

13

.99

21

.31

22

.35

26

.43

28

.08

32

.32

68

.47

Spectrum 22 – 13C NMR DEPT 135, product 17 13C NMR (75 MHz): δ = 13.8 (CH3), 14.0 (CH3), 21.3 (CH2), 22.3 (CH2), 26.4 (CH3), 28.1 (CH2), 28.1 (CH2), 32.3 (CH2), 68.5 (CH2) ppm

The bromination and substitution reactions were conducted with PHT and 2 pyrrolidone in order to prevent the cleavage

of the protection (Scheme 36). The achievement of the wanted product was confirmed by the collection of x-ray data

of both derivatives 18 and 19 (Figures 31, 32).

β

δ

α

γ

ζ

ϑ

ι

ε

ι

ζ

β

γ α

ϑ

δ

η

ε

η

ζ

Page 67: epub.ub.uni-greifswald.de · Table of contents 1. Pteridines and pterins

63

SCHEME 36 – BROMINATION AND SUBSTITUTION WITH XANTHATE AND CARBAMATE

FIGURE 31 – MOLECULAR STRUCTURE OF PRODUCT 18

Page 68: epub.ub.uni-greifswald.de · Table of contents 1. Pteridines and pterins

64

FIGURE 32 – MOLECULAR STRUCTURE OF PRODUCT 19

The molecular structure, 13C NMR DEPT-135, mass and IR spectra constitute novel information about the product 16.

The signal I (CH2) of the thiol chain is overlapped with the peak of CH2 H of the α-position of the carbonyl at around 3.26

ppm; the amino group is always squashed around 6 ppm and difficult to integrate (Spectrum 23).

7.0 6.5 6.0 5.5 5.0 4.5 4.0 3.5 3.0 2.5 2.0 1.5 1.0

Chemical Shift (ppm)

0

0.05

0.10

0.15

0.20

0.25

0.30

0.35

0.40

0.45

0.50

0.55

0.60

0.65

0.70

0.75

0.80

0.85

0.90

0.95

1.00

No

rma

lize

d In

ten

sity

3.013.013.014.052.272.044.002.001.83

0.9

30

.96

0.9

81

.07

1.0

91

.11

1.2

01

.22

1.2

5

1.4

11

.44

1.4

61

.47

1.4

81

.49

1.5

01

.50

1.5

21

.73

1.7

61

.78

1.8

01

.88

1.9

21

.95

1.9

7

3.2

03

.23

3.2

53

.27

4.5

24

.54

4.5

6

5.9

2

7.2

8

Spectrum 23 – 1H NMR of product 16 1H NMR (300MHz ,CHLOROFORM-d) δ = 5.92 (br. s., 2 H), 4.54 (t, J = 6.8 Hz, 2 H), 3.35 - 3.13 (m, 4 H), 1.92 (quin, J = 7.0 Hz, 2 H),

1.79 (sxt, J = 7.3 Hz, 2 H), 1.54 - 1.40 (m, 4 H), 1.23 (t, J = 7.4 Hz, 3 H), 1.09 (t, J = 7.4 Hz, 3 H), 1.00 - 0.92 (m, 3 H) ppm

The DEPT 135 experiment shows the expected signals and the separation between the peaks β and γ is like in the

previous derivative C’ (Spectrum 24).

A

D

G

B

H C

E F

I

L

M

C

D

H

A

F

G

B

E

I

L

M

Page 69: epub.ub.uni-greifswald.de · Table of contents 1. Pteridines and pterins

65

110 105 100 95 90 85 80 75 70 65 60 55 50 45 40 35 30 25 20 15 10

Chemical Shift (ppm)

0

0.05

0.10

0.15

0.20

0.25

0.30

0.35

0.40

0.45

0.50

0.55

0.60

0.65

0.70

0.75

0.80

0.85

0.90

0.95

1.00

No

rma

lize

d In

ten

sity

7.9

0

13

.75

13

.98

21

.33

22

.33

28

.06

28

.10

31

.58

32

.32

68

.41

110 105 100 95 90 85 80 75 70 65 60 55 50 45 40 35 30 25 20 15 10

Chemical Shift (ppm)

-1.0

-0.9

-0.8

-0.7

-0.6

-0.5

-0.4

-0.3

-0.2

-0.1

0

0.1

0.2

0.3

0.4

0.5

0.6

0.7

No

rma

lize

d In

ten

sity

7.8

813

.74

13

.96

21

.33

22

.33

28

.05

28

.09

31

.58

32

.31

68

.41

Spectrum 24 – 13C NMR, DEPT 135, product 16 13C NMR (75 MHz): δ = 7.9 (CH3), 13.7 (CH3), 14.0 (CH3), 21.3 (CH2), 22.3 (CH2), 28.1 (CH2, CH2), 31.6 (CH2), 32.3 (CH2), 68.4 (CH2) ppm

β

δ

α

γ

ε

η ζ

ϑ

ι

κ

ε

κ

η

δ

β

γ α

ζ ι

ϑ

Page 70: epub.ub.uni-greifswald.de · Table of contents 1. Pteridines and pterins

66

205 200 195 190 185 180 175 170 165 160 155 150 145 140 135 130 125 120 115

Chemical Shift (ppm)

0

0.05

0.10

0.15

0.20

0.25

0.30

0.35

0.40

0.45

0.50

0.55

0.60

0.65

0.70

0.75

0.80

0.85

0.90

0.95

1.00

No

rma

lize

d In

ten

sity

11

7.6

2

13

9.9

0

15

6.4

8

16

3.2

3

16

5.4

616

8.1

5

20

1.4

2

Spectrum 25 – 13C NMR product 16, quaternary carbons 13C NMR (75MHz ,CHLOROFORM-d) δ = 201.4, 168.2, 165.5, 163.2, 156.5, 139.9, 117.6 ppm

C=O

C-2 C-8A

C-4

C-6 C-7

C-4a

C=O

Page 71: epub.ub.uni-greifswald.de · Table of contents 1. Pteridines and pterins

67

The NMR spectra of product 19 are in accordance with the proposed structures (Spectra 26, 27, 28).

7.5 7.0 6.5 6.0 5.5 5.0 4.5 4.0 3.5 3.0 2.5 2.0 1.5 1.0 0.5

Chemical Shift (ppm)

0

0.05

0.10

0.15

0.20

0.25

0.30

0.35

0.40

0.45

0.50

0.55

0.60

0.65

0.70

0.75

0.80

0.85

0.90

0.95

1.00

No

rma

lize

d In

ten

sity

0.003.003.136.004.123.612.092.142.162.011.391.34

0.9

30

.96

1.0

61

.08

1.1

11

.29

1.3

11

.40

1.4

51.4

71

.49

1.5

31

.63

1.6

61

.75

1.7

81

.80

1.8

71

.91

1.9

41

.96

3.2

43

.27

3.2

9

4.4

94

.51

4.5

14

.53

4.5

44

.55

4.5

64

.57

5.5

95.6

15

.635

.65

5.6

75

.81

5.8

35

.86

5.8

8

Spectrum 26 – 1H NMR product 19 1H NMR (300MHz ,CHLOROFORM-d) δ = 5.84 (q, J = 7.2 Hz, 1 H), 5.65 (td, J = 6.2, 12.5 Hz, 1 H), 4.53 (dt, J = 2.0, 6.7 Hz, 2 H), 3.27 (t, J

= 7.2 Hz, 2 H), 1.91 (quin, J = 6.9 Hz, 2 H), 1.84 - 1.72 (m, 2 H), 1.65 (d, J = 7.2 Hz, 3 H), 1.56 - 1.37 (m, 4 H), 1.30 (d, J = 6.2 Hz, 6 H),

1.08 (t, J = 7.3 Hz, 3 H), 1.00 - 0.90 (m, 3 H) ppm

210 205 200 195 190 185 180 175 170 165 160 155 150 145 140 135 130 125 120 115 110 105 100

Chemical Shift (ppm)

0

0.05

0.10

0.15

0.20

0.25

0.30

0.35

0.40

0.45

0.50

0.55

0.60

0.65

0.70

0.75

0.80

0.85

0.90

0.95

1.00

No

rma

lize

d In

ten

sity

11

7.7

6

13

8.2

7

15

6.5

916

3.4

6

16

6.1

9

16

8.2

2

19

6.2

5

21

1.8

1

Spectrum 27 – 13C NMR of product 19, quaternary carbons 13C NMR (75MHz ,CHLOROFORM-d) δ = 211.8, 196.3, 168.2, 166.2, 163.5, 156.6, 138.3, 117.8 ppm

A

C’

B

D

F

A

B

H’ E

G

C’ G

F

I

I

L

L

H’

M

M

N

N

E

D

R

C=O

C-2

C-8A C-4

C-6 C-7

C-4a

C=O C=S

C=S

Page 72: epub.ub.uni-greifswald.de · Table of contents 1. Pteridines and pterins

68

95 90 85 80 75 70 65 60 55 50 45 40 35 30 25 20 15

Chemical Shift (ppm)

0

0.05

0.10

0.15

0.20

0.25

0.30

0.35

0.40

0.45

0.50

0.55

0.60

0.65

0.70

0.75

0.80

0.85

0.90

0.95

1.00

No

rma

lize

d In

ten

sity

13

.72

14

.00

16

.60

21

.09

21

.1921

.31

22

.35

28

.11

28

.18

32

.28

49

.21

68

.43

78

.18

95 90 85 80 75 70 65 60 55 50 45 40 35 30 25 20 15

Chemical Shift (ppm)

-1.0

-0.9

-0.8

-0.7

-0.6

-0.5

-0.4

-0.3

-0.2

-0.1

0

0.1

0.2

0.3

0.4

0.5

0.6

0.7

0.8

0.9

1.0

No

rma

lize

d In

ten

sity

13

.76

14

.04

16

.64

21

.13

21

.23

21

.35

22

.392

8.1

42

8.2

232

.32

49

.25

68

.47

78

.22

Spectrum 28 – 13C-NMR DEPT 135, product 19

13C NMR (75 MHz) δ = 13.8 (CH3), 14.0 (CH3), 16.6 (CH3), 21.1 (CH3), 21.2 (CH3), 21.3 (CH2), 22.4 (CH2), 28.1 (CH2), 28.2 (CH2), 32.3

(CH2), 49.2 (CH), 68.5 (CH2), 78.2 (CH) ppm

The signals for the two μ are overlapped at 21.13 and 21.23 and the typical chemical shift of the signal for CH λ is proof

of the attached xanthogenate at the α-position of the carbonyl of the 6-acyl pterin. The carbon of C=S has a chemical

shift of 211.3, exactly as in the structure of B’, meaning any electronic differences between the positions 6 and 7 and

the carbonyl or alkyl chain are negligible. Also the C-7 carbon this time does not exhibit any particular shifting with

respect to the starting material 16 (Spectrum 25).

β

γ α

ϑ

ζ

δ

ι

ε

μ

λ

β γ λ

μ α

ϑ

ι'

κ'

ε δ

ζ η

κ'

ι'

Page 73: epub.ub.uni-greifswald.de · Table of contents 1. Pteridines and pterins

69

The protocol described by Garner at al. for the ring closing step worked properly and product 21 was synthesized

(Scheme 37) [75]. The employment of concentrated sulfuric acid permitted, at the same time, the cleavage of the

protecting group.

SCHEME 37 – RING CLOSING STEP AND SIMULTANEOUS DEPROTECTION BY SULFURIC ACID: FORMATION OF PRODUCT 21

The amidic proton O at lower field relative to the normal amine function confirms the cleavage of the protection

together with the disappearing of the pentyloxy chain (Spectrum 29). The good solubility is probably due to the thioether

chain which is still attached.

7.5 7.0 6.5 6.0 5.5 5.0 4.5 4.0 3.5 3.0 2.5 2.0 1.5 1.0

Chemical Shift (ppm)

0

0.05

0.10

0.15

0.20

0.25

0.30

0.35

0.40

0.45

0.50

0.55

0.60

0.65

0.70

0.75

0.80

0.85

No

rma

lize

d In

ten

sity

3.002.002.872.191.40

DMSO

0.9

50

.97

1.0

0

1.6

51

.67

1.7

01

.72

1.7

5

2.1

2

3.1

43

.17

3.1

9

3.6

1

7.3

4

Spectrum 29 – 1H NMR of product 21

1H NMR (300MHz ,DMSO-d6) δ = 7.34 (br. s., 1 H), 3.17 (t, J = 7.2 Hz, 2 H), 2.12 (s, 3 H), 1.69 (sxt, J = 7.3 Hz, 2 H), 0.97 (t, J = 7.3 Hz, 3

H) ppm

C’’

C’’

B

I

F

B

F

I

O

O

Page 74: epub.ub.uni-greifswald.de · Table of contents 1. Pteridines and pterins

70

Also the DEPT 135 NMR spectrum confirms the preparation of compound 21 (Spectrum 30).

110 105 100 95 90 85 80 75 70 65 60 55 50 45 40 35 30 25 20 15 10

Chemical Shift (ppm)

0

0.01

0.02

0.03

0.04

0.05

0.06

0.07

0.08

0.09

0.10

0.11

0.12

0.13

0.14

0.15

0.16

0.17

0.18

0.19

No

rma

lize

d In

ten

sity

13

.26

15

.35

21

.77

31

.40

115 110 105 100 95 90 85 80 75 70 65 60 55 50 45 40 35 30 25 20 15 10

Chemical Shift (ppm)

-0.9

-0.8

-0.7

-0.6

-0.5

-0.4

-0.3

-0.2

-0.1

0

0.1

0.2

0.3

0.4

0.5

0.6

0.7

0.8

0.9

1.0

No

rma

lize

d In

ten

sity

13

.26

15

.34

21

.76

31

.39

Spectrum 30 – 13C NMR DEPT 135, product 21

13C NMR (75 MHz): δ = 13.3 (CH3), 15.3 (CH3), 21.8 (CH2), 31.4 (CH2) ppm

ϑ

κ

κ'’

ϑ

ζ

κ'’

ζ

η

η

Page 75: epub.ub.uni-greifswald.de · Table of contents 1. Pteridines and pterins

71

195 190 185 180 175 170 165 160 155 150 145 140 135 130 125 120 115

Chemical Shift (ppm)

-0.01

0

0.01

0.02

0.03

0.04

0.05

0.06

0.07

0.08

0.09

0.10

0.11

0.12

0.13

0.14

0.15

0.16

0.17

0.18

No

rma

lize

d In

ten

sity

12

2.1

9

12

4.1

613

1.5

9

13

6.1

1

15

4.5

51

55

.46

16

0.1

5

16

1.8

6

19

0.5

5

Spectrum 31 – 13C NMR of product 21, quaternary carbons 13C NMR (75MHz ,DMSO-d6) δ = 190.5, 161.9, 160.1, 155.5, 154.5, 136.1, 131.6, 124.2, 122.2 ppm

C-2

C-8A C-4

C-6

C-7

C-4a

S2C=O

S2C=O C=C

C=C

Page 76: epub.ub.uni-greifswald.de · Table of contents 1. Pteridines and pterins

72

SCHEME 38 – COMPLEXATION REACTION AND THE ELEMENTAL ANALYSIS OF THE BROWNISH POWDER OBTAINED

The reaction of product 21 with the molybdenum precursor NaK3[MoO2(CN)4]6 *H20 [108] has given a dark-red powder

product, i.e. exhibiting a coloration typical of a molybdenum-(IV) complex, but a complete and unambiguous

characterization of the complex was not achieved yet (Scheme 38). The elemental analysis reports a lower content for

most of the elements probably because of salt impurities, which are side product of the reaction. Neither the

precipitation by various counter cations nor the extraction by acetonitrile did improve the analysis.

The possible formation of a product with a 1:1 ratio molybdenum/pterin by concomitant direct coordination with the

carbonyl and nitrogen donors as anchor, which has been already reported in the literature [109] cannot be excluded

without crystallographic data; nevertheless the metal in oxidation state IV should prefer the dithiolene ligand [110, 111].

Mass spectrometry was also inconclusive, most probably by the interference of salt impurities. It is in fact mentioned

by the device’s manual that this kind of impurities are problematic. The only low resolution peak obtained carrying a

molybdenum isotopic pattern has a molecular weight which is almost half of the expected complex including the counter

cations (Spectrum 32).

TABLE 5 – ELEMENTAL ANALYSIS OF THE COMPLEXATION REACTION

Complexation Product

N C H S

theoretical with 2 Na+

16.78 34.53 2.90 23.05

theoretical with 2 K+

16.15

33.25

2.79

22.19

theoretical with Na+, K+

16.46 33.88 2.84 22.61

experimental 11.23 23.98 2.92 10.99

Page 77: epub.ub.uni-greifswald.de · Table of contents 1. Pteridines and pterins

73

SPECTRUM 32 – ESI MASS PEAK WITH MOLYBDENUM ISOTOPIC PATTERN

The basic solution condition of the mass measurement may cause the formation of a bi-charged species as reported

below in Figure 33 obtained by deprotonation of the OH involved in the keto-enol tautomerism (Table 6).

TABLE 6 – POSSIBLE INTERPRETATION OF THE MASS PEAK

The signal obtained by ESI-MASS in a diluted methanol solution does not clearly demonstrate the formation of the target

complex. The 95Mo NMR of the product was measured showing a signal at -2.22 ppm which lies inside of the range of a

molybdenum (IV) [110]. Unfortunately no other related example is reported in the literature and it is not possible to

reference it (Spectrum 33).

Notably, the complexation reaction with another pterin-dithiolene ligand synthesized by the ERC-project team’s

member Nicolas Chrysochos, obtained also with the acylation protocol, reveals a similar chemical shift on the 95Mo NMR

(see Nicolas Chrysochos thesis).

Possible fragments

K Na K, Na

Complex 867.87 835.92 853.91

Complex1- 828.9 812.93

Complex2- 394.97 394.97

[M] =433.7 867.87/2 = 433.93

[M] =433.7

FIGURE 33 – POSSIBLE FRAGMENTATIONS AND STRUCTURE

Page 78: epub.ub.uni-greifswald.de · Table of contents 1. Pteridines and pterins

74

8 6 4 2 0 -2 -4 -6 -8 -10 -12 -14 -16 -18

Chemical Shift (ppm)

-0.3

-0.2

-0.1

0

0.1

0.2

0.3

0.4

0.5

0.6

0.7

0.8

0.9

1.0

No

rma

lize

d In

ten

sity

-2.2

2

SPECTRUM 33 – 95MO NMR, COMPLEXATION PRODUCT 95Mo NMR (20MHz ,Deuterium Oxide) δ = -2.22 ppm

Page 79: epub.ub.uni-greifswald.de · Table of contents 1. Pteridines and pterins

75

The carbamate derivative was not completely characterized in the course of this dissertation because of the difficulties

in isolating the pure product. The final compound is a salt (Scheme 39) and it was not possible to be purified by column

chromatography. The yield of reaction was only estimated to be around 40%.

Some interesting NMR spectra of 20 were obtained after deprotection with the mixture DCM/Et2O/H2SO4 and are

reported below (Spectra 34, 35, 36).

The NMR spectra of product 22 (Spectra 37, 38, 39) were measured from the same NMR tube of product 20 after around

24 hrs. The ring closing reaction was probably catalyzed by the presence of traces of sulfuric acid from the previous

reaction step (from 19 to 20).

SCHEME 39 – DEPROTECTION AND SPONTANEOUS RING CLOSING STEP

Page 80: epub.ub.uni-greifswald.de · Table of contents 1. Pteridines and pterins

76

6.5 6.0 5.5 5.0 4.5 4.0 3.5 3.0 2.5 2.0 1.5 1.0

Chemical Shift (ppm)

0

0.05

0.10

0.15

0.20

0.25

0.30

0.35

0.40

0.45

0.50

0.55

0.60

0.65

0.70

0.75

0.80

0.85

0.90

0.95

1.00

No

rma

lize

d In

ten

sity

3.002.973.082.622.222.164.100.84

DMSO

Water

0.9

50

.98

1.0

01

.10

1.1

21

.14

1.2

51

.27

1.5

01

.53

1.6

01

.62

1.6

41

.67

1.6

91

.72

3.0

43

.06

3.0

9

3.7

13

.73

3.7

53

.82

3.8

73

.89

3.9

13

.94

3.9

65.8

25

.84

5.8

75

.89

Spectrum 34 – 1H NMR of product 20

1H NMR (300MHz ,DMSO-d6) δ = 5.96 - 5.74 (m, 1 H), 4.03 - 3.70 (m, 4 H), 3.06 (t, J = 7.2 Hz, 2 H), 1.66 (sxt, J = 7.3 Hz, 2 H), 1.51 (d, J

= 7.3 Hz, 3 H), 1.25 (t, J = 7.0 Hz, 3 H), 1.12 (t, J = 6.9 Hz, 3 H), 0.98 (t, J = 7.3 Hz, 3 H) ppm

195 190 185 180 175 170 165 160 155 150 145 140 135 130 125 120 115 110 105 100

Chemical Shift (ppm)

0

0.05

0.10

0.15

0.20

0.25

0.30

0.35

0.40

0.45

No

rma

lize

d In

ten

sity

12

2.8

3

13

6.7

2

15

5.9

01

56

.84

16

0.1

0

16

3.3

6

19

2.4

8

19

5.9

2

Spectrum 35 – 13C-NMR product 20, quaternary carbons

13C NMR (75MHz ,DMSO-d6) δ = 195.9, 192.5, 163.4, 160.1, 156.8, 155.9, 136.7, 122.8 ppm

B

B

C’

F I

H’ M

N

M

N

H’

C’

F

I

C=O C-2

C-8A

C-4 C-6

C-7

C-4a

C=S

C=O

C=S

Page 81: epub.ub.uni-greifswald.de · Table of contents 1. Pteridines and pterins

77

90 85 80 75 70 65 60 55 50 45 40 35 30 25 20 15 10 5

Chemical Shift (ppm)

0

0.05

0.10

0.15

0.20

0.25

0.30

0.35

0.40

0.45

0.50

0.55

0.60

0.65

0.70

0.75

0.80

0.85

0.90

0.95

1.00

No

rma

lize

d In

ten

sity

11

.30

12

.42

13

.50

16

.4521

.45

31

.16

47

.11

48

.81

50

.62

90 85 80 75 70 65 60 55 50 45 40 35 30 25 20 15 10

Chemical Shift (ppm)

-1.0

-0.9

-0.8

-0.7

-0.6

-0.5

-0.4

-0.3

-0.2

-0.1

0

0.1

0.2

0.3

0.4

0.5

0.6

0.7

0.8

0.9

1.0

No

rma

lize

d In

ten

sity

11

.29

12

.42

13

.50

16

.45

21

.45

31

.16

47

.10

48

.80

50

.62

Spectrum 36 – 13C NMR, DEPT 135, product 20

13C NMR (75 MHz): δ = 11.3 (CH3), 12.4 (CH3), 13.5 (CH3), 16.5 (CH3), 21.5 (CH2), 31.2 (CH2), 47.1 (CH2), 48.8 (CH2), 50.6 (CH) ppm

λ

μ

ϑ

ι'

κ'

η ζ

ι'

λ

ζ η

ϑ

κ'

μ

Page 82: epub.ub.uni-greifswald.de · Table of contents 1. Pteridines and pterins

78

The signal C’’ in product 22 is unfortunately exactly overlapped with the DMSO signal; the product showed the best

solubility in this solvent (Spectrum 37).

8.5 8.0 7.5 7.0 6.5 6.0 5.5 5.0 4.5 4.0 3.5 3.0 2.5 2.0 1.5 1.0

Chemical Shift (ppm)

0

0.05

0.10

0.15

0.20

0.25

0.30

0.35

0.40

0.45

0.50

0.55

0.60

0.65

0.70

0.75

0.80

0.85

0.90

0.95

1.00

No

rma

lize

d In

ten

sity

3.006.062.001.883.44

DMSO

0.9

70

.99

1.0

21.2

31

.26

1.2

81

.631.6

51.6

71

.70

1.7

21

.75

3.1

23.1

53

.17

3.6

13

.63

3.6

53

.68

4.8

8

7.2

1

8.4

6

Spectrum 37 – 1H NMR product 22

1H NMR (300MHz ,DMSO-d6) δ = 3.64 (q, J = 7.2 Hz, 4 H), 3.15 (t, J = 7.2 Hz, 2 H), 1.69 (sxt, J = 7.3 Hz, 2 H), 1.26 (t, J = 7.2 Hz, 6 H),

0.99 (t, J = 7.4 Hz, 3 H) ppm

190 185 180 175 170 165 160 155 150 145 140 135 130 125 120 115 110 105 100

Chemical Shift (ppm)

0

0.05

0.10

0.15

0.20

0.25

0.30

No

rma

lize

d In

ten

sity

12

2.5

1

13

1.5

8

15

6.5

31

57

.13

15

9.0

3

16

4.8

7

18

0.3

7

18

8.3

9

Spectrum 38 – 13C NMR of product 22, quaternary carbons

13C NMR (75MHz ,DMSO-d6) δ = 188.4, 180.4, 164.9, 159.0, 157.1, 156.5, 131.6, 122.5 ppm

C’’

B

I

F

C’’ M’

N’

N’

M’

I

F

B

2.65 2.60 2.55 2.50 2.45 2.40 2.35 2.30

Chemical Shift (ppm)

0

0.05

0.10

0.15

0.20

0.25

0.30

0.35

0.40

0.45

0.50

0.55

0.60

0.65

0.70

0.75

0.80

0.85

0.90

0.95

1.00

No

rma

lize

d In

ten

sity

DMSO

C-2

C-8A

C-4 C-6 C-7

C-4a C=N C=C

Page 83: epub.ub.uni-greifswald.de · Table of contents 1. Pteridines and pterins

79

The signal ν at 55 ppm could not be assigned and it may belong to some impurities. The presence of a positive charge

influences the entire molecule. In fact, a difference in the chemical shifts in comparison with the previous product 21

can be observed. For example, the chemical shift of C-4 appeared at lower field.

95 90 85 80 75 70 65 60 55 50 45 40 35 30 25 20 15 10

Chemical Shift (ppm)

0

0.05

0.10

0.15

0.20

0.25

0.30

0.35

0.40

0.45

0.50

0.55

0.60

0.65

0.70

No

rma

lize

d In

ten

sity

11

.66

13

.40

21

.52

28

.27

31

.05

47

.70

55

.00

95 90 85 80 75 70 65 60 55 50 45 40 35 30 25 20 15 10

Chemical Shift (ppm)

-1.0

-0.9

-0.8

-0.7

-0.6

-0.5

-0.4

-0.3

-0.2

-0.1

0

0.1

0.2

0.3

0.4

0.5

0.6

0.7

0.8

0.9

No

rma

lize

d In

ten

sity

11

.41

13

.16

21

.27

28

.02

30

.80

47

.44

Spectrum 39 – 13C-NMR DEPT 135, product 22

13C NMR (75 MHz) δ = 11.4 (CH3, CH3), 13.2 (CH3), 21.3 (CH2), 28.0 (CH3), 30.8 (CH2), 47.4 (CH2, CH2) ppm

ϑ

κ'’

ζ η

ϑ

κ'’

ζ

η λ'

μ'

ν ?

λ' μ'

Page 84: epub.ub.uni-greifswald.de · Table of contents 1. Pteridines and pterins

80

The formation of the dithiolene precursor was also confirmed by the mass spectrum reported below. In particular, the

peak appeared in the positive ESI-MASS mode since, as salt, the target molecule is already positively charged (Spectrum

40)

SPECTRUM 40 – ESI MS OF CARBAMATE DERIVATIVE 22

[M] =

[M]

Page 85: epub.ub.uni-greifswald.de · Table of contents 1. Pteridines and pterins

81

3.5 Acylation protocol variations, targeting MPT

The initial good results obtained with the acylation protocol encouraged us to include further subunits of MPT. In

particular through collaboration with other members of the project two variations were pursued — the “pyrane ring

variation” and the “northern/-southern phosphate variation”. The results , which are from joint work combining the

different expertise and strategies from coplementary approaches, are reported partly in this text as well as in the

dissertations of the other two group members Nicolas Chrysochos and Mohsen Ahmadi.

3.5.1 Pyran ring variation

In this mutual work the substrate 15 and 15’ were prepared in order to allow for a synthetic comparison between the

substituents at position 7 of the pterin scaffold (Scheme 40). These alkyl-thio and alkyl-oxo chains protect this position

during the acylation reaction with the aldehydes (H, I). In later steps they work as potential leaving group during the

ring closing mechanism or can be removed as described in the following for pterin 15 (Schema 41). The derivatization

of 15 is described in this dissertation while the work on substrate 15’ is reported by Nicolas Chrysochos. Preparation of

the aldehyde I was carried out by Mohsen Ahmadi as the project member with more expertise in the handling of

phosphate/phosphnate functionalities; in fact, the preparation of the 6-alky pterin 24 precedes not only upon

deprotection, the formation of the pyran ring but also adds a functionality ready for O-P bond formation with a

phosphate moiety as described by Taylor in 2001 [65]. The deveplopment of new pterin-dithiolene ligand precursors

from the 6-acyl pterins 23 and 24 requires further synthetic work, which was not possible to carry out in the course of

this dissertation.

SCHEME 40 – PYRAN VARIATION

Product 23 was isolated after acylation with the aldehyde H; a coordinated molecule of acetic acid was revealed by the

NMR spectra (signal P,Spectrum 41) and the elemental analysis (experimental section). This phenomenon was observed

also in the molecular structure of product 24 reported in the following (Figure 34).

Page 86: epub.ub.uni-greifswald.de · Table of contents 1. Pteridines and pterins

82

Upon prolonged exposure of product to acetic acid the cleavage of the silane protection was observed. In particular a

complete loss of solubility in n-hexane is a clear signal of such side reaction. Al the signals were assigned to the targeted

structure, except for the amino function which could not be integrated (Spectrum 41).

7.0 6.5 6.0 5.5 5.0 4.5 4.0 3.5 3.0 2.5 2.0 1.5 1.0 0.5 0 -0.5

Chemical Shift (ppm)

0

0.05

0.10

0.15

0.20

0.25

0.30

0.35

0.40

0.45

0.50

0.55

0.60

0.65

0.70

0.75

0.80

0.85

0.90

0.95

1.00

No

rma

lize

d In

ten

sity

6.099.133.083.124.152.102.022.872.022.002.002.00

0.0

5

0.8

5

0.9

61

.06

1.0

81

.11

1.4

51

.46

1.4

71

.48

1.7

41.7

61

.79

1.8

91

.91

1.9

42

.11

3.1

83

.20

3.2

33

.41

3.4

33

.45

4.0

64

.08

4.1

0

4.5

24

.54

4.5

6

7.2

7

Spectrum 41 – 1H NMR of product 23 1H NMR (300MHz ,CHLOROFORM-d) δ = 4.54 (t, J = 6.6 Hz, 2 H), 4.08 (t, J = 6.8 Hz, 2 H), 3.43 (t, J = 6.6 Hz, 2 H), 3.25 - 3.16 (m, 2H),

2.11 (s, 3H), 1.91 (quin, J = 7.0 Hz, 2 H), 1.77 (sxt, J = 7.4 Hz, 2 H), 1.56 - 1.36 (m, 4 H), 1.08 (t, J = 7.4 Hz, 3 H), 0.99 - 0.92 (m, 3 H),

0.85 (s, 9 H), 0.05 (s, 6 H) ppm

205 200 195 190 185 180 175 170 165 160 155 150 145 140 135 130 125 120 115 110 105 100

Chemical Shift (ppm)

0

0.05

0.10

0.15

0.20

0.25

0.30

0.35

0.40

0.45

0.50

0.55

0.60

0.65

0.70

0.75

0.80

0.85

0.90

0.95

1.00

No

rma

lize

d In

ten

sity

11

7.3

8

13

9.9

5

15

5.7

716

3.2

1

16

5.6

9

16

8.1

7

17

6.3

319

9.0

8

Spectrum 42 – 13C NMR product 23, quaternary carbons 13C NMR (75MHz ,CHLOROFORM-d) δ = 199.1, 176.3, 168.2, 165.7, 163.2, 155.8, 140.0, 117 ppm

N

D

P B

D

C

F

N O

L

E

G

G

A

1

B

P

A

I C H

O

F

G

L

E

I

H

C

C=O

C-2 C-8A C-4 C-6 C-7

C-4a CH3-C=O

CH3-C=O

Page 87: epub.ub.uni-greifswald.de · Table of contents 1. Pteridines and pterins

83

95 90 85 80 75 70 65 60 55 50 45 40 35 30 25 20 15 10 5 0 -5 -10

Chemical Shift (ppm)

0

0.05

0.10

0.15

0.20

0.25

0.30

0.35

0.40

0.45

0.50

0.55

0.60

0.65

0.70

0.75

0.80

0.85

0.90

0.95

1.00N

orm

aliz

ed

Inte

nsi

ty

-5.4

0

13

.66

13

.98

18

.22

21

.08

21

.752

2.3

3

25

.83

28

.06

28

.11

32

.26

41

.495

9.2

9

68

.57

95 90 85 80 75 70 65 60 55 50 45 40 35 30 25 20 15 10 5 0 -5 -10

Chemical Shift (ppm)

-0.8

-0.7

-0.6

-0.5

-0.4

-0.3

-0.2

-0.1

0

0.1

0.2

0.3

0.4

0.5

0.6

0.7

0.8

0.9

1.0

No

rma

lize

d In

ten

sity

-5.3

9

13

.66

13

.97

21

.07

21

.74

22

.33

25

.82

28

.06

28

.11

32

.25

41

.50

59

.29

68

.56

Spectrum 43 – 13C NMR DEPT 135, product 23 and quaternary carbon C-9 13C NMR (75 MHz) δ = -5.4 (CH3), 13.7 (CH3), 14.0 (CH3), 21.1 (CH3), 21.7 (CH2), 22.3 (CH2), 25.8 (CH3), 28.1 (CH2), 28.1

(CH2), 32.3 (CH2), 41.5 (CH2), 59.3 (CH2), 68.6 (CH2) ppm

The signal of the carbon C-9 appears at high field in accordance with the literature references [112]. The presence of

the silicon atom permitted the measurement of the 29Si NMR (Spectrum 44).

β

δ

γ

ζ

α

ε

ϑ

κ

ι

λ

μ

η

λ

μ

ϑ

ε

β

γ α ζ

η δ κ ι

ν

ν

λ

C-9

Page 88: epub.ub.uni-greifswald.de · Table of contents 1. Pteridines and pterins

84

27 26 25 24 23 22 21 20 19 18 17 16 15 14 13 12 11 10 9

Chemical Shift (ppm)

-0.4

-0.3

-0.2

-0.1

0

0.1

0.2

0.3

0.4

0.5

0.6

0.7

0.8

0.9

1.0

No

rma

lize

d In

ten

sity

19

.88

Spectrum 44 – 29Si NMR of product 23 29Si NMR (60MHz ,CHLOROFORM-d) δ = 19.88 ppm

Page 89: epub.ub.uni-greifswald.de · Table of contents 1. Pteridines and pterins

85

The synthesis of product 24 was carried out with a racemic mixture of aldehyde H with the intention to test the stability

of the acetone protection during the acylation reaction using a cheaper reagent. The product was isolated with a

coordinated molecule of acetic acid (Spectrum 45, H), as confirmed by its molecular structure shown in Figure 34.

6.0 5.5 5.0 4.5 4.0 3.5 3.0 2.5 2.0 1.5 1.0 0.5

Chemical Shift (ppm)

0

0.05

0.10

0.15

0.20

0.25

0.30

0.35

0.40

0.45

0.50

0.55

0.60

0.65

0.70

0.75

0.80

0.85

0.90

0.95

1.00

No

rma

lize

d In

ten

sity

3.803.9811.492.492.263.012.001.131.000.991.202.001.070.010.73

0.8

80

.96

1.0

9

1.2

6

1.3

91

.45

1.7

41

.76

1.7

91

.891

.91

1.9

3

2.1

1

3.1

83

.20

3.2

3

3.3

33

.363

.71

3.7

93

.83

3.8

54.2

84.3

04

.32

4.5

44

.654.6

74

.69

Spectrum 45 – 1H NMR of product 24 1H NMR (300MHz ,CHLOROFORM-d) δ = 4.75 - 4.61 (m, 1 H), 4.54 (t, J = 6.0 Hz, 2 H), 4.30 (dd, J = 6.2, 8.0 Hz, 1 H), 3.81 (dd, J = 5.6,

17.4 Hz, 1 H), 3.71 (t, J = 7.5 Hz, 1 H), 3.32 (dd, J = 7.6, 17.3 Hz, 1 H), 3.20 (t, J = 7.4 Hz, 2 H), 2.11 (s, 3 H), 1.97 - 1.85 (m, 2 H), 1.83 -

1.70 (m, 2 H), 1.34 – 1.54 (m, 10 H), 1.09 (t, J = 7.3 Hz, 3 H), 0.96 (t, J = 6.8 Hz, 3 H) ppm

FIGURE 34 – MOLECULAR STRUCTURE OF PRODUCT 24

A

F

I D

E

C

G

G

B

H

C

H

I

F

C

L

V

M

V L

V

M

V

N

V

A

B

C D

O

V

E

C

L

M

V

N

V

O

V

Page 90: epub.ub.uni-greifswald.de · Table of contents 1. Pteridines and pterins

86

The description of the carbon 13C NMR is not reported, since the assignment of the signals requires further analysis.

Nevertheless the APCI-MASS (Spectrum 46) and the crystal structure strongly support the preparation of the targeted

product (Figure 34).

SPECTRUM 46 – APCI MS OF PRODUCT 24

[M] =

450.0 [M + H]+

Page 91: epub.ub.uni-greifswald.de · Table of contents 1. Pteridines and pterins

87

The removal of the alkyl-thioether substituent from position 7 of the pterin was established following a general protocol

described in 2011 for nitrogen containing heterocycles [113]. Even though the reaction’s yield was quite low (Scheme

41), the method is milder than the one described by W. Pfleiderer and it does not provoke a partial exchange of the

pentyloxy protection.

SCHEME 41 – REDUCTIVE SCISSION

During the first step the amino group reacted also with the triethyl silane giving intermediate 25 but by simple hydrolysis

overnight in methanol the desired product 26 is obtained. Also, the unreacted starting material can be recovered by

column chromatography.

A prolonged reaction does not improve the yield and increases the risk of side reactions. In particular, the reduction of

the pteridine ring was suggested by a APCI-MASS TCL-Express measurement (Spectra 47, 48). The spectra show the

formation of two new compounds with the molecular weight of reduced derivatives 26 and 25 and different Rf on the

TLC, meaning they are different compounds and do not result from a fragmentation process.

SPECTRUM 47 – APCI MS PRODUCT 26

[M26 red +H] =291.9

[M26 red]

Page 92: epub.ub.uni-greifswald.de · Table of contents 1. Pteridines and pterins

88

SPECTRUM 48 – APCI MS OF PRODUCT 26 AND A POTENTIAL REDUCED PRODUCT

Isolation and comprehensive characterization of these species was not achieved as of yet and requires future in-depth

studies in continuity of this dissertation.

The singlet for P at 9.45 ppm in the 1H NMR confirms the free position 7 of product 26 (Spectrum 49).

9.5 9.0 8.5 8.0 7.5 7.0 6.5 6.0 5.5 5.0 4.5 4.0 3.5 3.0 2.5 2.0 1.5 1.0 0.5

Chemical Shift (ppm)

0

0.05

0.10

0.15

0.20

0.25

0.30

0.35

0.40

0.45

0.50

0.55

0.60

0.65

0.70

0.75

0.80

0.85

0.90

0.95

1.00

No

rma

lize

d In

ten

sity

3.053.384.193.472.002.012.001.800.95

0.9

40

.97

0.9

91.2

31

.25

1.2

81

.47

1.5

11

.62

1.9

01.9

31

.95

1.9

72

.003

.24

3.2

63

.28

3.3

14.5

64

.58

4.6

0

5.7

4

9.4

5

Spectrum 49 – 1H NMR of product 26 1H NMR (300MHz ,CHLOROFORM-d) δ = 9.45 (s, 1 H), 5.78 (br. s., 2 H), 4.58 (t, J = 6.8 Hz, 2 H), 3.27 (q, J = 7.3 Hz, 2 H), 2.02 -

1.89 (m, 2 H), 1.57 - 1.40 (m, 4 H), 1.25 (t, J = 7.2 Hz, 3 H), 1.01 - 0.92 (m, 3 H) ppm

A

C

D

H

P

A

L

E G

D

E

C

P

L

R

R

H

G

[M26] =

[M26+H] =289.9

[M25 red ] =

[M25 red+H] = 405.9

Page 93: epub.ub.uni-greifswald.de · Table of contents 1. Pteridines and pterins

89

Also the 13C NMR (Spectrum 51) and DEP 135 experiment (Spectrum 50) show the comparison of signal C-7 that belongs

to the CH at position 7.

144 136 128 120 112 104 96 88 80 72 64 56 48 40 32 24 16 8

Chemical Shift (ppm)

0

0.05

0.10

0.15

0.20

0.25

0.30

0.35

0.40

0.45

0.50

0.55

0.60

0.65

0.70

0.75

0.80

0.85

0.90

0.95

1.00

No

rma

lize

d In

ten

sity

7.7

4

14

.00

22

.362

8.0

62

9.6

83

0.7

9

68

.76

11

0.8

7

14

2.8

7

14

9.8

0

152 144 136 128 120 112 104 96 88 80 72 64 56 48 40 32 24 16 8

Chemical Shift (ppm)

-1.0

-0.9

-0.8

-0.7

-0.6

-0.5

-0.4

-0.3

-0.2

-0.1

0

0.1

0.2

0.3

0.4

0.5

0.6

No

rma

lize

d In

ten

sity

7.7

3

13

.98

22

.35

28

.05

29

.67

30

.78

68

.76

77

.19

14

9.7

9

Spectrum 50 – 13C NMR DEPT 135, product 26 13C NMR (75 MHz): δ = 7.7 (CH3), 14.0 (CH3), 22.4 (CH2), 28.1 (CH2, CH2), 29.7 (CH2), 30.8 (CH2), 68.8 (CH2), 149.8 (CH) ppm

β γ

ι κ

α

ε

δ

C-7 κ

ε

β γ

α

δ ι

C-7

Page 94: epub.ub.uni-greifswald.de · Table of contents 1. Pteridines and pterins

90

200 195 190 185 180 175 170 165 160 155 150 145 140 135 130 125 120 115 110

Chemical Shift (ppm)

0

0.01

0.02

0.03

0.04

0.05

0.06

0.07

0.08

0.09

0.10

0.11

0.12

0.13

No

rma

lize

d In

ten

sity

11

0.8

7

14

2.8

7

14

9.8

0

15

8.5

2

16

2.7

0

16

8.2

4

20

0.9

4

Spectrum 51 – 13C NMR product 26, quaternary carbons and C-7 13C NMR (75MHz ,CHLOROFORM-d) δ = 200.9, 168.2, 162.7, 158.5, 149.8, 142.9, 110.9 ppm

C=O

C-2 C-8A

C-4 C-6

C-4a

C-7

C=O

Page 95: epub.ub.uni-greifswald.de · Table of contents 1. Pteridines and pterins

91

Bromination and susbstitution with isopropyl xanthate gave product 27 confimed also by the collection of the crystal

structural date (Scheme 42, Figure 35).

SCHEME 42 – XANTHATE DERIVATIVE OF PRODUCT 27

Interestingly the bromination of substrate 26 was achieved in a one day reaction while for substrate 16 the complete bromination was achieved only in 3 days (experimental section). Thepresence of the substituent at position 7 has probably either a steric and/or electronic infuence on this reaction.

9.5 9.0 8.5 8.0 7.5 7.0 6.5 6.0 5.5 5.0 4.5 4.0 3.5 3.0 2.5 2.0 1.5 1.0 0.5

Chemical Shift (ppm)

0

0.05

0.10

0.15

0.20

0.25

0.30

0.35

0.40

0.45

0.50

0.55

0.60

0.65

0.70

0.75

0.80

0.85

0.90

0.95

1.00

No

rma

lize

d In

ten

sity

3.106.074.463.022.132.111.171.240.891.00

0.9

40

.96

0.9

9

1.2

81

.30

1.3

21

.43

1.4

81

.54

1.6

51

.68

1.8

91

.911

.94

1.9

61

.98

4.5

54

.57

4.6

0

5.6

25

.67

5.6

9

5.8

8

7.5

2

9.4

1

Spectrum 52 – 1H NMR of product 27

1H NMR (300MHz ,CHLOROFORM-d) δ = 9.41 (s, 1 H), 5.86 (q, J = 7.2 Hz, 1 H), 5.67 (quind, J = 6.5, 12.5 Hz, 1 H), 4.65 - 4.49 (m, 2 H), 1.94 (quin, J = 7.0 Hz, 2 H), 1.66 (d, J = 7.2 Hz, 3 H), 1.58 - 1.38 (m, 4 H), 1.35 - 1.26 (m, 6 H), 1.01 - 0.91 (m, 3 H) ppm

A

C’ G

L

H’ M

N

E

D

R

R

S

S

A

M

D

E

C’

L

G N

H’

R

Page 96: epub.ub.uni-greifswald.de · Table of contents 1. Pteridines and pterins

92

152 144 136 128 120 112 104 96 88 80 72 64 56 48 40 32 24 16

Chemical Shift (ppm)

0

0.05

0.10

0.15

0.20

0.25

0.30

0.35

0.40

0.45

0.50

0.55

0.60

0.65

0.70

0.75

0.80

0.85

0.90

0.95

1.00

No

rma

lize

d In

ten

sity

13

.98

16

.21

21

.10

22

.33

28

.08

48

.25

68

.75

78

.31

12

2.2

114

1.1

5

15

0.2

0

152 144 136 128 120 112 104 96 88 80 72 64 56 48 40 32 24 16

Chemical Shift (ppm)

-1.0

-0.9

-0.8

-0.7

-0.6

-0.5

-0.4

-0.3

-0.2

-0.1

0

0.1

0.2

0.3

0.4

0.5

0.6

0.7

0.8

No

rma

lize

d In

ten

sity

13

.99

16

.22

21

.10

22

.33

28

.07

48

.25

68

.75

78

.32

15

0.2

0

Spectrum 53 – 13C NMR DEPT 135 of product 27

13C NMR (75 MHz): δ = 14.0 (CH3), 16.2 (CH3), 21.1 (CH3), 22.3 (CH2), 28.1 (CH2, CH2), 48.3 (CH), 68.8 (CH2), 78.3 (CH), 150.2 (CH) ppm

β γ κ' ι'

α

δ

λ

μ

ε

ν

ν ι'

λ μ

β

γ δ α

κ'

Page 97: epub.ub.uni-greifswald.de · Table of contents 1. Pteridines and pterins

93

215 210 205 200 195 190 185 180 175 170 165 160 155 150 145 140 135 130 125 120 115

Chemical Shift (ppm)

0

0.05

0.10

0.15

0.20

0.25

0.30

0.35

0.40

0.45

0.50

0.55

0.60

0.65

0.70

0.75

0.80

0.85

0.90

0.95

1.00

No

rma

lize

d In

ten

sity

12

2.2

114

1.1

5

15

0.2

0

15

8.4

116

3.1

4

16

8.1

019

6.1

4

21

1.5

1

Spectrum 54 – 13C NMR of product 27, quaternary carbons and C-7

13C NMR (75MHz ,CHLOROFORM-d) δ = 211.5, 196.1, 168.1, 163.1, 158.4, 150.2, 141.1, 122.2 ppm

FIGURE 35 – MOLECULAR STRUCTURE OF PRODUCT 27

C=O

C-2 C-8A

C-4

C-4a

C=S C=O

C=S

C-7

C-6

Page 98: epub.ub.uni-greifswald.de · Table of contents 1. Pteridines and pterins

94

3.5.2 Phosphate variation, northern and southern functionalization

Investigations on the phosphate functionality by project member Mohsen Ahmadi had supported the possibility to

adopt a C-P bond instead of the O-P bond in MPT models. In particular this alternative resulted in more stable compound

which in addition were easier to prepare. Therefore it was decided to synthesize the products 28 and 29, using as

reactants the aldehyde M and the sodium salt L (Scheme 43).

The northern functionalization (nicknamed for P-functionalization on the pre-dithiolene moiety) was carried out using

substrate 15 according to the acylation protocols described above. The C-P coordination did not show any instability

during the reaction and product 29 was obtained; a partial hydrolysis of the phosphate-ethoxy protection was the only

obdserved side reaction.

The southern functionalization (nicknamed for P-functionalization on the pyrazine moiety) requires an initial first

installation of the phosphate derivative L and the subsequent acylation with acetaldehyde. Product 28 was obtained

and it represents the first example of a pterin carrying a phosphonate moiety modeling the phosphate anchor to the

peptide of MPT.

SCHEME 43– PHOSPHATE VARIATION

The complete characterization of the products, yields of the reactions and the unique crystal structure of 28 is

reported in the dissertation of Mohsen Ahmadi.

Page 99: epub.ub.uni-greifswald.de · Table of contents 1. Pteridines and pterins

95

4 .Conclusion

The synthesis of a pterin-dithiolene ligand system was achieved, in particular the employment of the acylation protocol

had largely simplified the precedent method reported by Garner [75]. Moreover, the final model bears free amino and

amide functionalities, an aspect of extreme importance for the biological application and characterization (Figure 36).

FIGURE 36 – PTERIN DITHIOLENE LIGAND WITH FREE AMINO AND AMIDE FUNCTIONS

The intensive cooperation with the project members Nicolas Chrysochos and Mohsen Ahmadi resulted in two variations

of the acylation protocol with focus on the formation of the pyran ring and the introduction of the phosphate subunit.

The synthesis of these 6-acyl pterin derivatives (Figure 38) redrafted this synthetic protocol and further approached the

real natural cofactor with the intention of modelling MPT (Figure 37) and, perhaps, in the future MoCo.

FIGURE 37 – TARGETED SUBSTRUCTURES IN MPT

Through the preparation of these structure also the stability of the alternative C-P coordination of the phosphate

anchor with respect to the acylation protocol has been tested and verified (Figure 38, product 28 and 29).

Page 100: epub.ub.uni-greifswald.de · Table of contents 1. Pteridines and pterins

96

FIGURE 38 – STRUCTURE OBTAINED BY THE DEVELOPMENT OF THE ACYLATION PROTOCOL

A method for the removal of the alkyl-thioether chain of product 16 was optimized towards milder and easier conditions

and the xanthate derivative 27 was synthesized (Scheme 44).

SCHEME 44 – REMOVAL OF THE ALKYL-THIOETHER CHAIN

A complete characterization of the molybdenum complex with the prepared ligands was not achieved, either because

of the many encountered issues during the isolation and purification steps, or because of the formation of a different

species with a 1:1 ratio molybdenum-pterin, which could not be excluded unambiguous. The biological test with G’ (IT

114) has shown a kind of specific binding with the ApoTorA compare to the BSA ( as evidenced by comparison with BSA

bovine serum albumin). Nevertheless, further in-depth studies would be necessary prior to drawing final definite

conclusions.

Even though the first synthetic approach for the synthesis of a pterin-dithiolene ligands did not give the expected result,

the side reaction occurring during the bromination has been eventually understood and was described for the benefit

of future research along these lines (Schema 39).

The final product C’ is not the desired structure, but it is a new red coloured thiazole-pterin which could be used as

model for the S-Oxidation in the metabolism of the thiazolidinedione (TZD) ring, a reaction catalysed by cytochrome

P450 [96, 97].

Page 101: epub.ub.uni-greifswald.de · Table of contents 1. Pteridines and pterins

97

FIGURE 39 – C’ AS MODEL FOR S-OXIDATION OF TZD

Page 102: epub.ub.uni-greifswald.de · Table of contents 1. Pteridines and pterins

98

Experimental section

General

All reactions and manipulations were carried out using standard Schlenk and glove box techniques under an atmosphere

of high purity nitrogen or argon. The inert gas stream was first passed through a copper catalyst tube (from company

BASF with specification R3 / 11) at a reaction temperature of 200 °C followed by P4O10 and KOH columns and finally

the stream of inert gas was dried by a molecular sieve column (3 Å and 4 Å) before entering the Schlenk line.

Elemental Analysis (C, H, N and S) were carried out with an Elementar Vario micro elemental analyzer. Calculations of

the theoretical molar masses was based on the relative atomic masses obtained from IUPAC tables.

Nuclear Magnetic Resonance NMR measurements were recorded on a Bruker Avance II-300 MHz. All samples were

dissolved in deuterated solvents and chemical shifts (δ) are given in parts per million (ppm) using solvent signals as the

reference (CDCl3 1H: δ = 7.24 ppm; 13C: δ = 77.0 ppm; d6-DMSO 1H: δ = 2.49 ppm; 13C: δ = 39.5 ppm; CD3CN 1H: δ = 1.94

ppm; 13C: δ = 1.3 ppm) related to external tetramethylsilane (δ = 0 ppm). 1H decoupled 13C C NMR spectra were recorded.

Coupling constants (J) are reported in Hertz (Hz).

Infrared spectra were recorded on a Shimadzu IR Affinity-1 FTIR-spectrophotometer in the range of 4000–400 cm-1

using KBr pellets.

Single-crystal X-ray diffraction suitable single crystals of compounds were mounted on a thin glass fibre coated with

paraffin oil. X-ray single-crystal structural data were collected using a STOEIPDS 2T diffractometer equipped with a

normal-focus, 2.4 kW, sealed-tube X-ray source with graphite-monochromated MoKα radiation (λ =0.71073 Å).

The molecular structure C’-IT 70 was measured by Dr. G. J. Palm, the crystal was mounted in a 0.1 mm loop in paraffin

oil and frozen and stored in liquid nitrogen in April 2017. Data were collected at DESY on beamline P11 on 02-

03.07.2017.The wavelength was 20 keV. 720 0.5° oscillation images were taken. The exposure time was 300 ms.

Transmission was set to 100% (full intensity beam). The crystal-detector distance was 207.7 mm and gave a maximum

resolution of 0.8 A. A 50 μm beam size was used. The detector was a Pilatus detector (2463 x 2527 pixel). Data were

collected at 100 K in a N2 stream.

2-Amino-3-cyanopyrazine 1-oxide 1

Method B as reported in a publication [114] was followed exactly: a suspension of powdered glyoxime (17.3 g, 0.196

mol) and aminomalononitrile tosylate (49.8 g, 0.19 mol) in 160 ml of water was stirred at RT for 17 hrs. During this time

all starting materials passed into solution and a new precipitate formed. The reaction product was collected by filtration,

washed with a small amount of cold water and then with ethanol. After drying, the crude product was crystallized from

a mixture of DMF/EtOH. Data code = IT 3; yield 26%; elemental analysis: calculated C-44.12, H-2.96, N-41.16; found C-

42.14, H-3.039, N-39.3 ; 1H NMR (300MHz, DMSO-d6) δ= 8.48 (d, J = 3.8 Hz, 1 H), 7.98 (s, 3 H), 7.85 (d, J = 3.8 Hz, 1 H)

ppm.

Page 103: epub.ub.uni-greifswald.de · Table of contents 1. Pteridines and pterins

99

2,4-Diaminopteridine 8-oxide 2

The procedure as reported in a publication [114] was followed exactly: guanidine hydrochloride (2.4 g, 39.2 mmol) was

added to a solution of sodium (1.5 g, 65 mmol) in 200 ml of absolute methanol, and the precipitated sodium chloride

was removed by filtration. To the filtrate was added product 1 (2.9 g, 21.3 mmol), and the resulting mixture was heated

under reflux with stirring for 16 hrs. A yellow precipitate started to separate from the orange solution after

approximately 1 hr. The mixture was cooled and filtered and the collected solid washed well with methanol and dried.

The analytical sample was prepared by crystallization from a large volume of DMF. Data code = IT 4; yield 89%; 1H NMR

(300MHz , DMSO-d6) δ = 8.42 (d, J = 3.8 Hz, 1 H), 8.05 (d, J = 3.8 Hz, 1 H), 7.85 (br. s., 2 H) ppm.

4(3H)-Pteridinone, 2-amino-, 8-oxide 3

It was followed the exactly procedure reported in the publication [83]: was followed exactly: a suspension of 1.00 g of

2 (5.6 mmol) in 100 ml of 5% NaOH was heated under reflux until completely dissolved; then for additional 5 min (total

heating time 30 min). After that, the yellow solution was brought to pH 3 with 6 N HCl and allowed to stand for 30 min

before filtering, washing thoroughly with water and drying to yield a bright yellow powder. Data code = IT 5; yield 95%;

elemental analysis: calculated C-40.23, H-2.81, N-39.10 found C-36.53, H-2.65, N-35.26; IR (KBr pellet) cm-1 : C=O

1714,74 cm-1.

2-Amino-6-chloro-4(3H)-pteridinone(6-chloropterin) 4

It was followed the exactly procedure reported in the publication [115] was followed exactly: a suspension of product

3 (0.5 g, 28 mmol) in 2.5 mL of acetyl chloride was cooled to -60°C (chloroform/N2 bath) and 2.5 mL TFA (trifluoroacetic

acid), precooled to -60°C was added. The mixture was sealed in a glass pressure bottle, and gradually warmed to RT

(overnight, the glass must be leaved in the bath). The product 3 slowly dissolved, and after about 2 min at RT a pale

yellow solid started to precipitate from the reaction mixture, It was stirred at RT for additional 45 min, the pressure

bottle opened (careful !HCl is released), the suspension diluted with dry ether, and a pale yellow solid, collected by

filtration and dried in vacuo. Product 4 was prepared by dissolution of the above hydrochloride (0.5 g) in 5 mL of 5 %

cold sodium hydroxide solution. After 15 min stirring at RT, the clear pale yellow solution was filtered and the filtrate

acidified with glacial acetic acid. After stirring for 30 min, the resulting suspension was filtered and the collected solid

washed thoroughly with water and then with acetone. Data code = IT 5; yield 90% ; as hydrochloric salt 1H NMR (300MHz

,DMSO-d6) δ = 8.90 (s, 1 H), 8.49 (br. s., 2 H) ppm ; 13C NMR (75MHz ,DMSO-d6) δ = 172.0, 158.0, 152.9, 149.1, 142.8,

128.3 ppm; 13C NMR (75 MHz, DEPT-135): δ = 149.1 (CH) ppm.

Page 104: epub.ub.uni-greifswald.de · Table of contents 1. Pteridines and pterins

100

6-Chloro-2-{[(dimethylamino)methylene]amino}pteridin-4-one 5

It was followed the exactly procedure reported in the publication [65] was followed exactly: to a stirred solution of

product 4 (3.8 g, 0.019 mol) in DMF (8 mL) at 25°C was added t-BuO(Me2N)2CH (Bredereck’s reagent) (4.7 ml, 21.7

mmol) in one portion. After 24 hrs, Et2O (30 ml) was added and the resulting precipitate collected by filtration. The

precipitate was washed with Et2O then dried by heating (100 °C) under vacuum to yield product 5 as a fine yellow

powder. Data code = IT 163.2; yield 84%; 1H NMR (300MHz , CHLOROFORM-d) δ = 8.98 (s, 1 H), 8.72 (s, 1 H), 3.27 (s, 3

H), 3.21 (s, 3 H) ppm.

2,4-Pyrimidinediamine, 5-nitroso-6-(pentyloxy) 6

A procedure as reported in publications [93, 116-118] was followed exactly : sodium (10 g, 0.43 mol, 1.5 equivalent)

was added to 600 mL of n-penthanol in a two-neck round bottom-flask and the resulting mixture was stirred and heated

at reflux for 2 hrs, than 4-chloro-2,6-diaminopyrimidine (41.4 g, 0.28.6 mol) was added and stirring with refluxing

continued for 5 hrs. After cooling, the mixture was neutralized with acetic acid and the volume reduced almost to

dryness. After adding 600 mL of 30% acetic acid solution the temperature was increased to 80°C. Sodium nitrite (24.15

g, 0.35 mol) was dissolved in a minimum amount of water and added dropwise; the formation of a deep purple

coloration was observed. The reaction was maintained under stirring at 80°C for 40 min and at the end the warm mixture

was put into a separatory funnel and the water bottom-layer removed.The oily deep purple upper-layer after 1 night at

-20°C gives a crystalline purple solid which was filtered off, washed with water and stored in a desiccator overnight.

Crystallization from chloroform/n-hexane 1:19. Data code = IT 21; X-ray code IT 21 ; yield 85%; Mass spectrometry (APCI,

positive, low t. low f.) = 226.3, 156.2 m/z; elemental analysis: calculated C-47.99, H-6.71, N-31.09; found C-47.71, H-6.5,

N-30.7; 1H NMR (300MHz , DMSO-d6) δ = 10.09 (d, J = 4.2 Hz, 6 H), 7.97 (d, J = 3.8 Hz, 6 H), 7.76 (d, J = 8.7 Hz, 13 H), 4.48

(t, J = 6.8 Hz, 13 H), 1.79 (quin, J = 6.9 Hz, 14 H), 1.46 - 1.28 (m, 27 H), 0.95 - 0.82 (m, 21 H) ppm; 13C NMR (75MHz ,

DMSO-d6) δ = 170.8, 163.5, 150.8, 139.5, 66.7, 28.0, 27.6, 21.8, 13.9 ppm; 13C NMR, (75 MHz, DEPT-135): δ = 13.9 (CH3),

21.8 (CH2), 27.6 (CH2), 28.0 (CH2), 66.7 (CH2) ppm.

Page 105: epub.ub.uni-greifswald.de · Table of contents 1. Pteridines and pterins

101

2,4,5-Pyrimidinetriamine, 6-(pentyloxy) 7

A procedure as reported in a publication [7]: was followed: a solution of ethanol and water (865 mL /251 mL) was

purged with nitrogen for 3 hrs, sodium hydrosulfide (147 g, 2.6 mol) was added and the temperature increased to 80°C.

To the yellow solution was added portionwise product 6 (under nitrogen flow, 94.35 g, 0.41 mol) until complete

dissolution. After cooling, the ethanol was removed by vacuum and the resulting a suspension. The yellow precipitate

was collected, washed with water, dried under vacuum and stored under nitrogen pressure. Data code = IT 22; yield

82.5%; mass spectrometry (APCI, positive, low t. low f.) = 212.1 m/z; elemental analysis: calculated C-51.17, H-8.11, N-

33.15; found C-45.88, H-7.23, N-29.76; 1H NMR (300MHz, DMSO-d6) δ = 5.62 (s, 2 H), 5.22 (s, 2 H), 4.12 (t, J = 6.6 Hz, 2

H), 3.11 (s, 2 H), 1.64 (quin, J = 6.9 Hz, 2 H), 1.44 - 1.22 (m, 4 H), 1.00 - 0.77 (m, 3 H); 13C NMR (75MHz, DMSO-d6) δ =

157.6, 155.5, 155.4, 100.2, 64.7, 28.5, 27.7, 21.9, 13.9; 13C NMR, (75 MHz, DEPT-135): δ = 13.9 (CH3), 21.9 (CH2), 27.7

(CH2), 28.5 (CH2), 64.7 (CH2) ppm.

2-Pteridinamine, 4-(pentyloxy) 8

A procedure as reported in a publication [93]: was followed: in a two-neck round-bottom flask 600 mL of DMF were

purged with nitrogen for 2 hrs. To this were added first glyoxal-hydrate trimer (3.3 g, 15.7mmol) and then product 7 (10

g, 47.4 mmol) giving a green-blue coloration of the mixture. The reaction was stirred at RT for 3 days under nitrogen.

Some insoluble material was filtered off and the filtrate diluted with 600 mL of water. The solution was extracted with

chloroform (three times 400 mL), then the organic layer was washed twice with water, dried using sodium sulfate,

filtered and evaporated to dryness. Crystallization from 160 mL of methanol/water 1:1 gave a yellowish crystalline

product. Data code = IT 23; yield 69%; mass spectrometry (APCI, positive, low t. low f.) = 233.9-235.0, 163.9 m/z;

elemental analysis: calculated C-56.64, H-6.48, N-30.02; found C-54.46, H-6.38, N-28.76; 1H NMR (300MHz,

CHLOROFORM-d) δ = 8.80 (d, J = 2.3 Hz, 1 H), 8.52 (d, J = 2.3 Hz, 1 H), 4.58 (t, J = 7.0 Hz, 2 H), 1.94 (quin, J = 7.3 Hz, 2 H),

1.53 - 1.33 (m, 4 H), 0.98 - 0.90 (m, 3 H) ppm; 13C NMR (75MHz ,DMSO-d6) δ = 157.6, 155.5, 155.4, 100.2, 64.7, 28.5,

27.7, 21.9, 13.9 ppm; 13C NMR (75MHz, CHLOROFORM-d) δ = 167.7, 161.5, 157.0, 150.5, 140.3, 124.4, 68.6, 28.1, 27.9,

22.3, 13.9 ppm; 13C NMR, (75 MHz, DEPT-135): δ = 13.9 (CH3), 22.3 (CH2), 27.9 (CH2), 28.1 (CH2), 68.6 (CH2), 140.3 (CH),

150.5 (CH) ppm.

2-Pteridinamine 4-(pentyloxy)- 8-oxide 9

A procedure ad reported in a publication [93] was followed: a solution of product 8 (5.0 g, 21 mmol) in 85 mL of TFA

(trifluoroacetic acid) was cooled to 6°C, and 5 mL of 30% H2O2 were slowly added with stirring. The mixture was kept at

6°C (refrigerator) and after 60 hrs further 2 mL of 30% H2O2 were added. After 30 hrs in the refrigerator at the same

temperature the mixture was concentrated in vacuum to 1/3 of its volume, diluted with 20 mL of water and the resulting

precipitate was collected. The solid was suspended in 100 mL of water and neutralized by concentrated ammonia. The

precipitate was filtered off and dried to give a yellowish chromatographically pure material. Data code = IT 28; X-ray

Page 106: epub.ub.uni-greifswald.de · Table of contents 1. Pteridines and pterins

102

code IT 27CHF; yield 40%; mass spectrometry (APCI, positive, low t. low f.) = 249.8 m/z; 1H NMR (300MHz, DMSO-d6) δ

= 8.52 (d, J = 3.8 Hz, 1 H), 8.20 (d, J = 3.8 Hz, 1 H), 4.46 (t, J = 6.6 Hz, 2 H), 1.81 (quin, J = 7.0 Hz, 2 H), 1.51 - 1.25 (m, 4

H), 0.97 - 0.83 (m, 3 H) ppm; 13C NMR (75MHz, DMSO-d6) δ = 167.4, 160.4, 152.3, 139.3, 135.0, 125.7, 67.8, 27.7, 27.5,

21.8, 13.9 ppm; 13C NMR, (75 MHz, DEPT-135): δ = 13.9 (CH3), 21.8 (CH2), 27.5 (CH2), 27.7 (CH2), 67.8 (CH2), 135.0 (CH),

139.3 (CH) ppm.

2-Pteridinamine, 6-chloro-4-(pentyloxy) 10

A procedure as reported in a publication [93] was followed exactly: at -40°C product 9 (1 g, 4 mmol) was suspended in

10 ml of freshly distilled acetyl chloride, and with stirring 3 mL of TFA (trifluoroacetic acid) were slowly added while

stirring. The solution was warmed to 0°C, stirred for 3 hrs, and then the reaction was stopped by addition of 30 g of ice.

The mixture was neutralized with concentrated ammonia to pH 4, then extracted with chloroform, (6 x 50 mL). The

organic layer was washed with water, dried with sodium sulfate and the solvent evaporated to a small volume. The

residue was purified by chromatography on a silica-gel column with chloroform. The product fraction was evaporated

to dryness and the residue crystallized from isopropanol. Data code = IT 99; X-ray code IT 99; yield 62%; mass

spectrometry (APCI, positive, low t. low f.) = 267.7-269.7 m/z; elemental analysis: calculated C-49.35, H-5.27, N-26.16;

found C-54.35, H-5.66, N-28.69; 1H NMR (300MHz , CHLOROFORM-d) δ = 8.75 (s, 1 H), 5.98 (br. s., 1 H), 4.57 (t, J = 7.0

Hz, 2 H), 1.92 (quin, J = 7.2 Hz, 3 H), 1.58 - 1.30 (m, 4 H), 1.07 - 0.82 (m, 3 H) ppm; 13C NMR (75MHz , CHLOROFORM-d)

δ = 166.9, 161.6, 155.8, 151.0, 142.5, 122.6, 68.7, 28.1, 27.9, 22.3, 13.9 ppm; 13C NMR (75 MHz, DEPT-135): δ = 13.9

(CH3), 21.8 (CH2), 27.5 (CH2), 27.7 (CH2), 67.8 (CH2), 135.0 (CH), 139.3 (CH) ppm.

Ethanone, 1-[2-amino-4-(pentyloxy)-7-methyl-6-pteridinyl] 11

A procedure as reported in a publication [119] was followed: a mixture of product 6 (4 g, 17.8 mmol) and 60 mL of 2,4-

pentanedione were heated under reflux for 7 hrs. The solvent was dried giving an oily black residue. The crude product

was first purified by silica gel column chromatography (ethyl acetate /n hexane 1:1) and then crystallized from a

water/methanol mixture 1:1 yielding a yellow microcrystalline powder. Data code = IT 29; X-ray code IT 29; yield 80%;

mass spectrometry (APCI, positive, low t. low f.) = 290.1 m/z; elemental analysis: calculated C-58.12, H-6.62, N-24.21;

found C-57.50, H-6.52, N-23.64; 1H NMR (300MHz, CHLOROFORM-d) δ = 6.01 (br. s., 1 H), 4.55 (t, J = 6.8 Hz, 2 H), 2.92

(s, 3 H), 2.76 (s, 3 H), 2.04 - 1.81 (m, 2 H), 1.64 - 1.31 (m, 4 H), 1.06 - 0.86 (m, 3 H) ppm; 13C NMR (75MHz, CHLOROFORM-

d) δ = 199.9, 167.9, 163.0, 161.4, 157.1, 142.7, 120.6, 68.5, 28.1, 27.5, 25.1, 22.3, 14.0 ppm; 13C NMR (75 MHz, DEPT-

135): δ = 14.0 (CH3), 22.3 (CH2), 25.1 (CH3), 27.5 (CH3), 28.1 (CH2, CH2), 68.5 (CH2) ppm.

Page 107: epub.ub.uni-greifswald.de · Table of contents 1. Pteridines and pterins

103

Acetic acid, 2-[[2,4-diamino-6-(pentyloxy)-5-pyrimidinyl] imino]- ethyl ester 12

A modified procedure similar to one reported in a publication [118] was followed: product 7 (73 g 0.34 mol) was

dissolved in a mixture of methanol and water (5.2 L/390 mL), the reactant (Acetic acid, 2-ethoxy-2-hydroxy-, ethyl ester,

see below) was added and the solution kept under stirring at RT for 1 hr. The yellow precipitate was filtered off and

washed with a minimum amount of diethyl ether. To the filtrate was added circa 500 mL of water and kept in a

refrigerator overnight obtaining a second fraction of product. The recrystallization procedure was repeated till a yield

of 74.3 % was achieved. Data code = IT 77 S1; yield 74,3%; mass spectrometry (APCI, positive, low t. low f.) = 296.1 m/z;

elemental analysis: calculated C-52.87, H- 7.17, N-23.71; found C-52.86, H-7.00, N-23.3; 1H NMR (300MHz,

CHLOROFORM-d) δ = 8.33 (s, 1 H), 5.81 (br. s., 1 H), 4.97 (br. s., 1 H), 4.48 - 4.19 (m, 4 H), 1.78 (quin, J = 7.0 Hz, 2 H),

1.51 - 1.27 (m, 7 H), 1.01 - 0.84 (m, 3 H) ppm; 13C NMR (75MHz, CHLOROFORM-d) δ = 165.4, 164.0, 163.2, 161.2, 141.8,

102.7, 77.2, 66.8, 60.9, 28.5, 28.2, 22.3, 14.3, 13.9 ppm; 13C NMR (75 MHz, DEPT-135): δ = 13.9 (CH3), 14.3 (CH3), 22.3

(CH2), 28.2 (CH2), 28.5 (CH2), 60.9 (CH2), 66.8 (CH2), 141.8 (CH) ppm.

Acetic acid, 2-ethoxy-2-hydroxy-, ethyl ester - reactant

A modified procedure similar to one reported in a publication [120] was followed: a solution of glyoxylic acid

monohydrate in ethanol (9.2 g in 100 mL) was heated for 114 hrs at 80°C. After reduction of volume at rotary evaporator

the crude product was used without further purification taking into account ca. 70% of yield. Data code = IT 66, yield

70%); 1H NMR (300MHz ,CHLOROFORM-d) δ = 4.94 (s, 1 H), 4.37 - 4.20 (m, 2 H), 3.93 - 3.79 (m, 1 H), 3.79 - 3.59 (m, 2

H), 1.37 - 1.23 (m, 6 H) ppm.

7(8H)-Pteridinone, 2-amino-4-(pentyloxy) 13

A modified procedure similar to one reported in a publication [118] was followed: product 12 (20 g, 67.7 mmol) was

dissolved in a mixture of 4 L of ethanol and 540 mL of sodium hydrogen carbonate solution (0.5 N). The mixture was

refluxed for 2 hrs, than another 135 mL of sodium hydrogen carbonate solution were added and the reflux was

continued for 1 hr. To the mixture were added around 10 g of active charcoal as fine powder, and the solution filtered

while still warm. After cooling, the yellow solution was brought to pH 5 with acetic acid giving a white powder which

was filtered off, washed with water in order to remove the excess acetic acid and kept in the desiccator overnight. Data

code = IT 77 S2/ CT-OXO; yield 60% ; mass spectrometry (APCI, positive, low t. low f.) = 250.0 m/z; elemental analysis:

calculated C-53.00, H-6.07, N-28.10; found C-52.53, H-5.697, N-26.97; 1H NMR (300MHz, DMSO-d6) δ = 7.42 (s, 1 H),

6.52 (br. s., 2 H), 5.75 (s, 1 H), 4.30 (t, J = 6.8 Hz, 2 H), 1.73 (t, J = 7.0 Hz, 2 H), 1.50 - 1.19 (m, 4 H), 0.97 - 0.76 (m, 3 H)

Page 108: epub.ub.uni-greifswald.de · Table of contents 1. Pteridines and pterins

104

ppm; 13C NMR (75MHz, DMSO-d6, TFA) δ = 166.4, 160.7, 150.8, 141.4, 110.9, 110.2, 67.5, 28.2, 27.9, 22.1, 13.9 ppm; 13C

NMR (75 MHz, DEPT-135, DMSO-d6, TFA): δ = 13.9 (CH3), 22.1 (CH2), 27.9 (CH2), 28.2 (CH2), 67.5 (CH2), 141.4 (CH) ppm.

2-Pteridinamine, 7-chloro-4-(pentyloxy) 14

A procedure as reported in publications [35, 121] was followed: to a mixture of phosphoryl chloride (615.76 mL, in

excess) and potassium chloride (30.71 g 0.4 mol) was added product 13 (29.2 g 0.12 mol). The flask was put in a hot oil

bath at 140°C. After heating for 12 min with stirring, the mixture was cooled with an ice-bath (the glassware has to be

in good condition in order to avoid leaking of the solvent that can react violently with water). The excess of phosphoryl

chloride was evaporated in vacuum (with two cooling traps in order to protect the vacuum pump). The residue was

treated with 600 g of ice and 500 mL of chloroform. After formation of two layers, the upper part was neutralized using

sodium hydrogen carbonate), shaken and neutralized many times until no more acidity was observed in the aqueous

layer. The organic layer was separated, the aqueous fraction extracted twice with chloroform, and the combined organic

extracts dried using sodium sulfate and evaporated. The crude product was purified by chromatography column short-

path aluminium oxide (neutral) using chloroform as eluent. The collected product precipitated in n-hexane and was

filtered off yielding a pure pale yellow powder (microcrystalline needles). Data code = IT 82; X-ray code CT CL yield 40%;

mass spectrometry (APCI, positive, low t. low f.) = 267.8-269.7 m/z; elemental analysis: calculated C-49.35, H-5.27, N-

26.16; found C-47.4, H-5.87, N-20.20; 1H NMR (300MHz, DICHLOROMETHANE-d2) δ = 8.38 (s, 1 H), 4.51 (t, J = 6.8 Hz, 2

H), 1.88 (quin, J = 7.1 Hz, 2 H), 1.51 - 1.33 (m, 4 H), 0.98 - 0.87 (m, 3 H) ppm; 13C NMR (75MHz, DICHLOROMETHANE-d2)

δ = 167.9, 163.1, 157.1, 153.7, 139.5, 122.7, 68.9, 28.5, 28.3, 22.7, 14.1 ppm; 13C NMR (75 MHz, DEPT-135): δ = 14.1

(CH3), 22.7 (CH2), 28.3 (CH2), 28.5 (CH2), 68.9 (CH2), 139.5 (CH) ppm.

2-Pteridinamine, 4-(pentyloxy)-7-(propylthio) 15

A procedure as reported in publications [35, 118]: was followed: to a mixture of phosphoryl chloride (615.76 mL, in

excess) and potassium chloride (30.71 g 0.4 mol) product 13 was added (29.2 g 0.12 mol). The flask was put in a hot oil

bath at 140°C. After heating for 12 min with stirring, the mixture was cooled with an ice-bath. The excess of phosphoryl

chloride was evaporated in vacuum (with two cooling traps in order to protect the vacuum pump). The residue was

treated with 600 g of ice and 500 mL of chloroform, after separation of the organic phase the aqueous fraction was

extracted twice with chloroform, and the combined organic extract dried using sodium sulfate and then evaporated.

The residue consisting in crude product 14 was treated with a prepared solution of 1-propanethiol (30 mL, use carefully

is extremely smelly, all residue or waste hast to be treated with a sodium hypochlorite solution 13%!) in 500 ml of 0.25

N sodium methoxide. The solution was stirred for 1 hr at RT, then neutralized with acetic acid and evaporated. The

residue was treated with 200 mL of water and the precipitate collected and purified by short-path aluminium oxide

Page 109: epub.ub.uni-greifswald.de · Table of contents 1. Pteridines and pterins

105

(neutral) chromatography eluting with gradient from chloroform to 10% methanol/chloroform. Recrystallization from

chloroform/n-hexane 1:19. Data code = IT 84; X-ray code IT 84, MADXM5; yield 70%; mass spectrometry (APCI, positive,

low t. low f.) = 307.9 m/z; elemental analysis: calculated C-54.70, H-6.89, N-22.78, S-10.43; found C-58.9, H-7.22, N-

24.26, S-10.94; 1H NMR (300MHz, CHLOROFORM-d) δ = 8.27 (s, 1 H), 5.45 (br. s., 2 H), 4.53 (t, J = 7.0 Hz, 2 H), 3.33 (t, J

= 7.2 Hz, 2 H), 1.99 - 1.86 (m, 2 H), 1.86 - 1.73 (m, 2 H), 1.53 - 1.31 (m, 4 H), 1.07 (t, J = 7.4 Hz, 3 H), 1.00 - 0.86 (m, 3 H)

ppm; 13C NMR (75MHz, CHLOROFORM-d) δ = 167.8, 164.8, 161.6, 157.0, 140.3, 119.4, 68.2, 31.5, 28.2, 27.9, 22.3, 22.0,

13.9, 13.4 ppm; 13C NMR (75 MHz, DEPT-135): δ = 13.4 (CH3), 13.9 (CH3), 22.0 (CH2), 22.3 (CH2), 27.9 (CH2), 28.2 (CH2),

31.5 (CH2), 68.2 (CH2), 140.3 (CH) ppm.

1-Propanone, 1-[2-amino-4-(pentyloxy)-7-(propylthio)-6-pteridinyl] 16

A procedure as reported in a publication [35] was followed: to a suspension of product 15 (4.5 g, 14.6 mmol) in 360 mL

of acetic acid/water 3:1 and propionaldehyde (17.8 mL, 17 equivalents) were added dropwise simultaneously 25.5 g of

FeSO4.*7H2O (25.5 g, 91.8 mmol) in 15 of mL water and 11.6 mL of tert-butyl hydroperoxide 70% aqueous solution with

vigorous stirring within around 60 seconds. After stirring for 2 minutes, 720 mL of water were added, the precipitate

was collected and washed with water and dried to give a pure pale yellow powder. Data code = IT 90; X-ray code IT 90

COL; yield 78%; mass spectrometry (APCI, positive, low t. low f.) = 364.3 m/z; elemental analysis: calculated C-56.17, H-

6.93, N-19.27, S-8.82; found C-56.22, H-6.73, N-19.22, S-8.676; 1H NMR (300MHz, CHLOROFORM-d) δ = 5.89 (br. s., 1

H), 4.54 (t, J = 6.8 Hz, 2 H), 3.52 - 3.12 (m, 4 H), 1.92 (quin, J = 7.0 Hz, 2 H), 1.86 - 1.71 (m, 2 H), 1.59 - 1.36 (m, 4 H), 1.29

- 1.16 (m, 3 H), 1.10 (d, J = 7.2 Hz, 3 H), 0.96 (t, J = 7.0 Hz, 3 H) ppm; 13C NMR (75MHz ,CHLOROFORM-d) δ = 201.4, 168.2,

165.5, 163.2, 156.5, 139.9, 117.6, 68.4, 32.3, 31.6, 28.1, 28.1, 22.3, 21.3, 14.0, 13.7, 7.9 ppm; 13C NMR (75 MHz, DEPT-

135): δ = 7.9 (CH3), 13.7 (CH3), 14.0 (CH3), 21.3 (CH2), 22.3 (CH2), 28.1 (CH2), 31.6 (CH2), 32.3 (CH2), 68.4 (CH2) ppm.

1-Acetone, 1-[2-amino-4-(pentyloxy)-7-(propylthio)-6-pteridinyl] 17

A procedure as reported in a publication [35]: was followed: to a suspension of product 15 (650 mg, 2.1 mmol ) and

acetaldehyde (2 mL, 17 equivalent) in 50 mL of acetic acid/water 3:1 were added dropwise simultaneously 3.7 g of

FeSO4.7H2O in 17 of mL water and 1.7 mL of tert-butyl hydroperoxide with vigorous stirring within around 60 sec. After

stirring for 2 minutes, 100 mL of water were added, the precipitate was collected and washed with water and dried to

give a pure pale yellow powder. Data code = CT 39 yield 85%; X-ray code CT 39; mass spectrometry (APCI, positive, low

t. low f.) = 350.0 m/z; elemental analysis: calculated C-54.99, H-6.63, N-20.04, S-9.18; found C-53.56, H-6.06, N-19.31,

S-9.192; 1H NMR (300MHz, CHLOROFORM-d) δ = 5.69 (br. s., 1 H), 4.49 (t, J = 6.8 Hz, 2 H), 3.29 - 3.10 (m, 2 H), 2.69 (s, 3

H), 1.87 (quin, J = 7.0 Hz, 2 H), 1.74 (sxt, J = 7.3 Hz, 2 H), 1.63 (s, 1 H), 1.53 - 1.30 (m, 4 H), 1.04 (t, J = 7.4 Hz, 3 H), 0.96 -

0.85 (m, 3 H) ppm; 13C NMR (75MHz, CHLOROFORM-d) δ = 198.8, 168.2, 165.5, 163.3, 156.5, 140.1, 117.7, 68.5, 32.3,

Page 110: epub.ub.uni-greifswald.de · Table of contents 1. Pteridines and pterins

106

28.1, 28.1, 26.4, 22.3, 21.3, 14.0, 13.8 ppm; 13C NMR (75 MHz, DEPT-135): δ = 13.8 (CH3), 14.0 (CH3), 21.3 (CH2), 22.3

(CH2), 26.4 (CH3), 28.1 (CH2), 28.1 (CH2), 32.3 (CH2), 68.5 (CH2) ppm.

1- isopropyloxy(thiocarbonyl)thio]propanyl, 1-[2-amino-4-(pentyloxy)-

-7-(propylthio)-6-pteridinyl] 18

To a stirred solution of product 16 (3 g, 8.2 mmol) and 2-pyrrolidone (3.5 equivalents, 2.2 mL) in dry THF at 60°C was

added slowly portion-wise PHT (pyrrolidone hydrotribromide, 14.23 g, 3.5 equivalents) within around 5 hrs. After 3 days

of heating the volume was reduced almost to dryness. The product was solubilized in 80 mL of ethyl acetate, washed

three times with water and once with brine. The crude product was suspended in 60 mL of acetone and potassium

isopropylxanthate (1.43 g, 1 equivalent) was added and the solution was kept stirring overnight. The mixture was filtered

and the mother liquor dried on a rotary evaporator and the residue dissolved in ethyl acetate. The organic solution was

washed three times with water and once with brine, and dried with sodium sulfate.

The pure product was obtained after short-path aluminium oxide (neutral) column chromatography using chloroform

as eluent. The compound was stored in a dark place in order to avoid possible photochemical side reactions.

Data code = IT 103; X-ray code IT 103 FIN; yield 65%; mass spectrometry (APCI, positive, low t. low f.) = 497.7, 363.8

m/z; elemental analysis: calculated C-50.68, H-6.28, N-14.07, S-19.33; found C-50.68, H-6.17, N-13.09, S-18.85; 1H NMR

(300MHz, CHLOROFORM-d) δ = 5.84 (q, J = 7.2 Hz, 1 H), 5.65 (quind, J = 6.2, 12.4 Hz, 1 H), 4.53 (dt, J = 1.9, 6.7 Hz, 2 H),

3.26 (t, J = 7.2 Hz, 2 H), 1.98 - 1.85 (m, 2 H), 1.85 - 1.72 (m, 2 H), 1.65 (d, J = 7.2 Hz, 4 H), 1.57 - 1.37 (m, 4 H), 1.30 (d, J =

6.2 Hz, 6 H), 1.08 (t, J = 7.4 Hz, 3 H), 1.01 - 0.91 (m, 3 H) ppm; 13C NMR (75MHz, CHLOROFORM-d) δ = 211.8, 196.3,

168.2, 166.2, 163.5, 156.6, 138.3, 117.8, 78.2, 68.4, 49.2, 32.3, 28.2, 28.1, 22.3, 21.3, 21.2, 21.1, 16.6, 14.0, 13.7 ppm; 13C NMR (75 MHz): δ = 13.8 (CH3), 14.0 (CH3), 16.6 (CH3), 21.1 (CH3), 21.2 (CH3), 21.3 (CH2), 22.4 (CH2), 28.1 (CH2), 28.2

(CH2), 32.3 (CH2), 49.2 (CH), 68.5 (CH2), 78.2 (CH) ppm.

1- diethylamino(thiocarbonyl)thio]propanyl, 1-[2-amino-4-(pentyloxy)-

-7-(propylthio)-6-pteridinyl] 19

To a stirred solution of product 16 (3 g, 8.2 mmol) and 2-pyrrolidone (3.5 equivalents, 2.18 mL) in dry 300 mL of dry THF

at 60°C was added slowly portion-wise 14.23 g (3.5 equivalents) of PHT within around 5 hrs. After 3 days the volume

was reduced almost to dryness. The product was solubilized in 80 mL of ethyl acetate and washed three times with

water and once with brine. The crude product was suspended in 60 mL of acetone, sodium diethyldithiocarbamate (1.4

g, 1 equivalents) was added and the solution kept stirring overnight. The mixture was filtered, the mother liquor dried

on a rotary evaporator and the residue dissolved in ethyl acetate. The organic solution washed three times with water

and once with brine, then dried with sodium sulfate. The pure product was obtained by short-path aluminium oxide

Page 111: epub.ub.uni-greifswald.de · Table of contents 1. Pteridines and pterins

107

(neutral) column chromatography with chloroform and crystallized from a chloroform/n-hexane mixture. The

compound was stored in a dark place in order to avoid possible photochemical side reactions.

Data code = IT 108/122; X-ray code IT 122 FIN; yield 62%; mass spectrometry IT 108 (APCI, positive, low t. low f.) = 511.5

m/z.

1-diethylamino(thiocarbonyl)thio]propanyl, 1-[2-amino-7-(propylthio)-6-pteridinyl] 20

The deprotection was achieved dissolving 0.5 g (0.97 mmol) of product 19 in 100 ml dichloromethane and diethyl ether

1:1 mixture with 10 equivalents of concentrated sulfuric acid at RT overnight under stirring. The volume of the reaction

mixture was reduced almost to dryness, 10 mL of acetone were added and the mixture kept under stirring until a

homogeneous suspension was obtained. Then 40 g of ice in 10 mL of water were added slowly until complete

precipitation of the yellowish powder. The solution was neutralized with sodium hydrogen carbonate and the product

filtered out. The precipitate was washed with water and acetone and kept in a dessicator under vacuum overnight. The

compound was stored in a dark place in order to avoid possible photochemical side reactions. Data code = IT 108 CARB.,

IT 124; 73% of 20, mass spectrometry (APCI, positive, low t. low f.) = 19-511.6; 20-364 m/z; elemental analysis: 20

calculated C-46.34, H-5.49, N-19.07, S-21.83; found C-45.45, H-5.20, N-17.88; S-21.43; 1H NMR 20 (300MHz, DMSO-d6)

δ = 5.95 - 5.77 (m, 1 H), 3.90 (d, J = 6.2 Hz, 1 H), 3.86 - 3.71 (m, 3 H), 3.06 (t, J = 7.2 Hz, 2 H), 1.66 (sxt, J = 7.3 Hz, 3 H),

1.51 (d, J = 7.3 Hz, 3 H), 1.25 (t, J = 7.0 Hz, 3 H), 1.12 (t, J = 6.9 Hz, 3 H), 0.98 (t, J = 7.3 Hz, 3 H) ppm; 13C NMR 20 (75MHz,

DMSO-d6) δ = 195.9, 192.5, 163.4, 160.1, 156.8, 155.9, 136.7, 122.8, 50.6, 48.8, 47.1, 31.2, 21.5, 16.5, 13.5, 12.4, 11.3

ppm; 13C NMR 20 (75 MHz, DEPT-135): δ = 11.3 (CH3), 12.4 (CH3), 13.5 (CH3), 16.5 (CH3), 21.5 (CH2), 31.2 (CH2), 47.1

(CH2), 48.8 (CH2), 50.6 (CH) ppm.

1,3-dithiol-2-one-4-methyl-5-(2-pteridinamine, 7-(propylthio) 21

A small flask containing product 18 (3 g, 6 mmol) was kept in an ice bath with stirring. After 10 min 3.2 mL of

concentrated sulfuric acid (10 equivalents) were added slowly and dropwise until the formation of a homogeneous

mixture was observed. The flask was kept in the bath under stirring for 3 hrs, upon which RT was reached. The viscous

mixture was poured into ice for the precipitation of a yellow powder. The suspension was neutralized with sodium

hydrogen carbonate, taking care that the strong evolution of gas did not cause a loss of product. After filtration the

product was washed with water and acetone and stored under vacuum overnight. The compound was stored in a dark

place in order to avoid possible photochemical side reactions. Data code = IT 111 DEP FIN; yield 60%; mass spectrometry

(APCI, positive, low t. low f.) = 367.7 m/z; elemental analysis: calculated C-42.49, H-3.57, N-19.06, S-26.18; found C-

38.55, H-3.59, N-17, S-26.15; 1H NMR (300MHz, DMSO-d6) δ = 7.39 (br. s., 1 H), 3.17 (t, J = 7.2 Hz, 3 H), 2.12 (s, 3 H),

1.69 (sxt, J = 7.3 Hz, 2 H), 0.97 (t, J = 7.3 Hz, 3 H) ppm; 13C NMR (75MHz, DMSO-d6) δ = 190.5, 161.9, 160.1, 154.5, 136.1,

Page 112: epub.ub.uni-greifswald.de · Table of contents 1. Pteridines and pterins

108

131.6, 124.2, 122.2, 31.4, 21.8, 15.3, 13.3 ppm; 13C NMR (75 MHz, DEPT-135): δ = 13.3 (CH3), 15.3 (CH3), 21.8 (CH2), 31.4

(CH2) ppm.

Ethanaminium, N-(1,3-dithiol-2-ylidene-4-methyl-5-(2-pteridinamine, 7-(propylthio))-

-N-ethyl, sulfate 22

A small flask containing product 20 (3 g, 6.8 mmol) was kept in an ice bath with stirring. After 10 min 3.6 mL of

concentrated sulfuric acid (10 equivalents) was added slowly and dropwise until the formation of a homogeneous

mixture was observed. The flask was kept in the bath under stirring for 3 hrs, upon which the RT was reached. The

viscous mixture was poured into ice and neutralized with sodium hydrogen carbonate. The product was partly extracted

four times with chloroform. The organic phases were collected and dried on a rotary evaporator obtaining a brownish

oil, which was kept in a dessicator overnight. The analytical data were obtained after the attempt of recrystallization in

methanol at low temperature. The compound was stored in a dark place in order to avoid possible photochemical side

reactions. Data code = IT 124, IT 125; yield estimated around 40%; mass spectrometry (APCI, positive, low t. low f.) =

423.3 m/z; 1H NMR (300MHz, DMSO-d6) δ = 8.33 (br. s., 2 H), 5.96 - 5.68 (m, 1 H), 4.05 - 3.65 (m, 4 H), 3.12 (t, J = 7.0 Hz,

2 H), ), 2.55-2.45 (s, 3 H) overlap with DMSO signal, 1.68 (sxt, J = 7.3 Hz, 2 H), 1.52 (d, J = 7.2 Hz, 3 H), 1.25 (t, J = 6.8 Hz,

3 H), 1.11 (t, J = 6.8 Hz, 3 H), 1.05 - 0.93 (m, 3 H); 13C NMR (75MHz, DMSO-d6) δ = 188.4, 180.4, 164.9, 159.0, 157.1,

156.5, 131.6, 122.5, 55.0, 47.7, 31.1, 28.3, 21.5, 13.4, 11.7 ppm; 13C NMR (75 MHz, DEPT-135): δ = 11.4 (CH3, CH3), 13.2

(CH3), 21.3 (CH2), 28.0 (CH3), 30.8 (CH2), 47.4 (CH2, CH2) ppm.

1-[[(1,1-dimethylethyl)dimethylsilyl]oxy], 1-[2-amino-4-(pentyloxy)-7-(propylthio)-6-pteridinyl] 23

To a suspension of product 15 (0.5 g, 1.6 mmol) and the aldehyde H (5.2 g, 17 equivalents) in 40 mL of acetic acid/water

3:1 were added dropwise simultaneously 2.5 g of FeSO4.7H2O (2.5 g. 9.2 mmol) in 13 mL of water and 1.3 mL of tert-

butyl hydroperoxide 70% aqueous solution with vigorous stirring within around 60 seconds. After stirring for 2 minute,

80 mL of water were added, the precipitate was collected and washed with water and dried to give a pure pale yellow

powder. Data code = CT 38; yield 70%; mass spectrometry (APCI, positive, low t. low f.) = 494.2 [(without AcO-)+H], 362

m/z; elemental analysis (with a molecule of acetic acid): calculated C-54.22, H-7.83, N-12.65, S-5.79; found C-55.868, H-

7.922, N-12.778, S-7.12;1H NMR (300MHz, CHLOROFORM-d) δ = 4.54 (t, J = 6.6 Hz, 2 H), 4.08 (t, J = 6.8 Hz, 2 H), 3.43 (t,

J = 6.6 Hz, 2 H), 3.25 - 3.16 (m, 2 H), 2.11 (s, 3 H), 1.91 (quin, J = 7.0 Hz, 2 H), 1.77 (sxt, J = 7.4 Hz, 2 H), 1.56 - 1.37 (m, 4

H), 1.08 (t, J = 7.4 Hz, 3 H), 0.99 - 0.92 (m, 3 H), 0.85 (s, 9 H), 0.05 (s, 6 H) ppm; 13C NMR (75MHz, CHLOROFORM-d) δ =

199.1, 176.3, 168.2, 165.7, 163.2, 155.8, 140.0, 117.4, 68.6, 59.3, 41.5, 32.3, 28.1, 28.1, 25.8, 22.3, 21.7, 21.1, 18.2, 14.0,

13.7, -5.4 ppm;13C NMR (75 MHz, DEPT-135): δ = -5.4 (CH3, CH3), 13.7 (CH3), 14.0 (CH3), 21.1 (CH3), 21.7 (CH2), 22.3

(CH2), 25.8 (CH3, CH3, CH3), 28.1 (CH2), 28.1 (CH3), 32.3 (CH3), 41.5 (CH3), 59.3 (CH3), 68.6 (CH3) ppm;29Si NMR (60MHz,

CHLOROFORM-d) δ = 19.88 ppm.

Page 113: epub.ub.uni-greifswald.de · Table of contents 1. Pteridines and pterins

109

1-2-[(4RS)-2,2-Dimethyl-1,3-dioxolan-4-yl]-1-,1-[2-acetamido-4-(pentyloxy)-

-7-(propylthio)-6-pteridinyl] 24

To a suspension of product 15 (0.5 g, 1.6 mmol) and the aldehyde I (3.9 g, 17 equivalents) in 40 mL of acetic acid/water

3:1 were added dropwise simultaneously 2.5 g of FeSO4.7H2O (2.5 g. 9.2 mmol) in 13 mL of water and 1.3 mL of tert-

butyl hydroperoxide 70% aqueous solution with vigorous stirring within around 60 seconds. After stirring for 1 minute,

80 mL of water were added, the precipitate was collected and washed with water and dried to give a pure pale yellow

powder. Data code = ACT 20; yield 82.5 %; X-ray MAD205F; mass spectrometry (APCI, positive, low t. low f.) = 450.0

m/z; 1H NMR (300MHz, CHLOROFORM-d) δ = 4.75 - 4.61 (m, 1 H), 4.54 (t, J = 6.0 Hz, 2 H), 4.30 (dd, J = 6.2, 8.0 Hz, 1 H),

3.81 (dd, J = 5.6, 17.4 Hz, 1 H), 3.71 (t, J = 7.5 Hz, 1 H), 3.32 (dd, J = 7.6, 17.3 Hz, 1 H), 3.20 (t, J = 7.4 Hz, 2 H), 2.11 (s, 3

H), 1.97 - 1.85 (m, 2 H), 1.83 - 1.70 (m, 2 H), 1.34 – 1.54 (m, 10 H), 1.09 (t, J = 7.3 Hz, 3 H), 0.96 (t, J = 6.8 Hz, 3 H) ppm.

1-[2-Amino-4-(pentyloxy)-6-pteridinyl]-1-propanone 26

Method B in a procedure as reported in a publication [122] was followed: a pre-dried 100 mL Schlenk with a

PTFE/silicone septum was charged with product 16 (6.48 g, 17.8) and palladium on charcoal 10% (380 mg, 0.356 mmol

). After careful (the fine powder charcoal can be lost) evacuation of the flask and installation of an nitrogen gas

atmosphere around 15 mL of THF (dry and oxygen free) were added and kept in an ice bath. Using a syringe triethylsilan

(8.5 mL, 3 equivalents) was added and the mixture kept at ice-bath temperature for 40 min (evolution of gas was

observed) and at RT for 5 hrs. The suspension was filtered through a patch of celite to remove the charcoal and the

volume was reduced. The crude mixture was dissolved in 50 mL of methanol and stirred overnight at RT in order to

remove the silane reacted with the amino function (see chapter results and discussion: product 25). On a rotary

evaporator the methanol was removed and the residue purified through aluminium oxide (neutral) short-path column

chromatography. The first fraction (eluent chloroform) was the unreacted starting material and the second (10%

methanol/chloroform eluent) was product 26. Data code = IT 127; yield 40%; mass spectrometry (APCI, positive, low t.

low f.) = 289.9 m/z; elemental analysis: calculated C-58.12, H-6.62, N-24.21; found C-57.18, H-6.324, N-23.6; 1H NMR

(300MHz, CHLOROFORM-d) δ = 9.45 (s, 1 H), 5.74 (br. s., 1 H), 4.58 (t, J = 6.8 Hz, 2 H), 3.27 (q, J = 7.3 Hz, 2 H), 1.95 (quin,

Page 114: epub.ub.uni-greifswald.de · Table of contents 1. Pteridines and pterins

110

J = 7.0 Hz, 2 H), 1.57 - 1.38 (m, 5 H), 1.25 (t, J = 7.3 Hz, 4 H), 1.03 - 0.90 (m, 3 H); 13C NMR (75MHz, CHLOROFORM-d) δ =

200.9, 168.2, 162.7, 158.5, 149.8, 142.9, 132.1, 122.2, 68.8, 64.2, 30.8, 28.1, 22.4, 14.0, 7.7, 0.0 ppm; 13C NMR (75 MHz,

DEPT-135): δ = 7.7 (CH3), 14.0 (CH3), 22.4 (CH2), 28.1 (CH2), 28.1 (CH2), 30.8 (CH2), 68.8 (CH2), 149.8 (CH) ppm.

1- isopropyloxy(thiocarbonyl)thio]propanyl, 1-[2-amino-4-(pentyloxy)-6-pteridinyl] 27

To a stirred solution of 3 g product 26 (10.3 mmol) and 2-pyrrolidone (3.5 equivalents, 2.7 mL) in dry THF at 60°C was

added slowly portion-wise PHT (pyrrolidone hydrotribromide, 17.9 g, 3.5 equivalents) within around 3 hrs and the

mixture was kept under stirring overnight. The volume was reduced almost to dryness, the product was solubilized in

80 mL of ethyl acetate and washed three times with water and once with brine. The crude product was suspended in

60 mL of acetone and 1.8 g of potassium isopropylxanthate (1 equivalent) was added. The solution was kept stirring

overnight. The mixture was filtered and the mother liquor dried on a rotary evaporator. The residue was dissolved in

ethyl acetate, the organic solution washed three times with water and once with brine, then dried with sodium sulfate.

The pure product was obtained by short-path aluminium oxide column chromatography using chloroform as eluent.

The compound was stored in a dark place in order to avoid possible photochemical side reactions. Data code = IT 128,

X-ray data IT 128f, yield 67%,Mass spectrometry (APCI, positive, low t. low f.) = 423.7, 289.9 m/z; elemental analysis:

calculated C-51.04, H-5.95, N-16.53, S-15.14; found C-46.45, H-5.084, N-23.64 , S-13.6.

1H NMR (300MHz,CHLOROFORM-d) δ = 9.41 (s, 1 H), 5.86 (q, J = 7.2 Hz, 1 H), 5.67 (td, J = 6.2, 12.5 Hz, 1 H), 4.64 - 4.51

(m, 2 H), 1.94 (quin, J = 7.0 Hz, 2 H), 1.66 (d, J = 7.2 Hz, 3 H), 1.58 - 1.39 (m, 5 H), 1.34 - 1.26 (m, 6 H), 1.00 - 0.91 (m, 3

H) ppm; 13C NMR (75MHz, CHLOROFORM-d) δ = 211.5, 196.1, 168.1, 163.1, 158.4, 150.2, 141.1, 122.2, 78.3, 68.7, 48.2,

28.1, 22.3, 21.2, 21.1, 16.2, 14.0 ppm; 13C NMR (75 MHz, DEPT-135): δ = 14.0 (CH3), 16.2 (CH3), 21.1 (CH3), 21.2 (CH3),

22.3 (CH2), 28.1 (CH2, CH2), 48.3 (CH3), 68.8 (CH2), 78.3 (CH), 150.2 (CH) ppm.

Ethanone, 1-[2-amino-4-(pentyloxy)-7-methyl(o-isopropyl dithiocarbonate)-6-pteridinyl] B’

To a stirred solution of product 11 (3 g; 8.2 mmol) and 2-pyrrolidone (3.5 equivalents, 2.2 mL) in dry THF at 55°C was

added slowly portion-wise 14.23 g (3.5 equivalents) of PHT (pyrrolidone hydrotribromide) within around 7 hrs and the

mixture was kept under stirring overnight. The volume was reduced almost to dryness. The product was solubilized in

80 mL of ethyl acetate and washed three times with water and once with brine. The crude product was suspended in

60 mL of acetone and potassium isopropylxanthate (1.43 g, 1 equivalents) was added. The solution was kept stirring

overnight. The mixture was filtered and the mother liquor dried on a rotary evaporator. The residue was dissolved in

ethyl acetate, the organic solution washed three times with water and once with brine, then dried with sodium sulfate.

The pure product was obtained by short-path aluminium oxide (neutral) column chromatography using chloroform as

eluent. The compound was stored in a dark place in order to avoid possible photochemical side reactions. Data code =

IT 32 PU, yield 72%; elemental analysis: calculated C-51.04, H-5.95, N-16.53, S-15.14; found C-50.70, H-5.77, N-16.32, S-

Page 115: epub.ub.uni-greifswald.de · Table of contents 1. Pteridines and pterins

111

15.17; 1H NMR (300MHz, DMSO-d6) δ = 7.80 (br. s., 1 H), 7.68 (br. s., 1 H), 5.60 (spt, J = 6.2 Hz, 1 H), 4.90 (s, 2 H), 4.51

(t, J = 6.6 Hz, 2 H), 2.64 (s, 3 H), 1.84 (quin, J = 6.9 Hz, 2 H), 1.53 - 1.33 (m, 4 H), 1.30 (d, J = 6.4 Hz, 6 H), 0.96 - 0.85 (m, 3

H); 13C NMR (75MHz, DMSO-d6) δ = 212.5, 198.9, 166.8, 163.4, 157.2, 156.9, 139.9, 120.4, 78.3, 67.6, 27.6, 26.8, 21.8,

20.9, 13.9; 13C NMR (75 MHz): δ = 13.9 (CH3), 20.9 (CH3, CH3), 21.8 (CH2), 26.8 (CH3), 27.6 (CH2, CH2), 40.2 (CH2), 67.6

(CH2), 78.3 (CH) ppm.

Ethanone, 1-[2-acetamido-4-(pentyloxy)-7-methyl(o-isopropyl dithiocarbonate)-6-pteridinyl]

White solid

Method as reported in a publication [92] was followed: a suspension of B’ (8 g, 18.8 mmol) in a mixture of 250 mL acetic

acid and 500 mL of acetic anhydride was refluxed for 3 hrs. Then the volume of the solution was reduced almost to

dryness. The residue dissolved in chloroform and washed with water three times, then dried with sodium sulfate and

the solvent was removed on a rotary evaporator The crude product (a dark brown solid) was purified by short-path

aluminium oxide (neutral) column chromatography with ethyl acetate/n-hexane as eluent mixture 1:1), the second

fraction is collected and crystallized in mixture of chloroform and n-hexane. The purified substance was a bright white

powder.

Data code = IT 59, yield 25%; 1H NMR (300MHz, DMSO-d6) δ = 10.92 (s, 1 H), 5.60 (quin, J = 6.2 Hz, 1 H), 4.98 (s, 2 H),

4.63 (t, J = 6.6 Hz, 2 H), 2.71 (s, 3 H), 2.35 (s, 3 H), 1.88 (quin, J = 6.9 Hz, 2 H), 1.53 - 1.35 (m, 4 H), 1.31 (d, J = 6.4 Hz, 6

H), 1.00 - 0.81 (m, 3 H) ppm.

Ethanone,1-[2-amino-4-(pentyloxy)-7-methyl(diethyldithiocarbamate)-6-pteridinyl]

Carbamate derivative

To a stirred solution of product 11 (3 g; 8.2 mmol) and 2-pyrrolidone (3.5 equivalents, 2.2 mL) in dry THF at 55°C was

added slowly portion-wise 14.23 g (3.5 equivalents) of PHT (pyrrolidone hydrotribromide) within around 7 hrs and the

mixture was kept under stirring overnight. The volume was reduced almost to dryness. The product was solubilized in

80 mL of ethyl acetate and washed three times with water and once with brine. The crude product was suspended in

60 mL of acetone and Sodium diethyldithiocarbamate (1.4 g, 1 equivalents) was added. The solution was kept stirring

overnight. The mixture was dried on a rotary evaporator and the residue was dissolved in ethyl acetate, the organic

solution washed three times with water and once with brine, then dried with sodium sulfate. The pure product was

obtained by short-path aluminium oxide (neutral) column chromatography using chloroform as eluent. The compound

was stored in a dark place in order to avoid possible photochemical side reactions.

Page 116: epub.ub.uni-greifswald.de · Table of contents 1. Pteridines and pterins

112

Data code = IT 37, yield 68%; 1H NMR (300MHz, DMSO-d6) δ = 7.91 - 7.50 (m, 2 H), 4.99 (s, 2 H), 4.50 (t, J = 6.6 Hz, 2 H),

3.91 (d, J = 6.8 Hz, 2 H), 3.76 (d, J = 7.2 Hz, 2 H), 2.64 (s, 3 H), 1.84 (t, J = 6.8 Hz, 2 H), 1.55 - 1.30 (m, 4 H), 1.28 - 1.09 (m,

7 H), 0.90 (t, J = 7.0 Hz, 3 H) ppm.

Ethanone,1-[2-amino-4-(pentyloxy)-7-methyl(diethyldithiocarbamate)-6-pteridinyl]- sulfate salt

Carbamate derivative protonated

The product was exposed to the nitrogen flow containing hydrochloric acid for around 5 minutes, as described in the

chapter “Result and Discussion”. The product was characterized without further purification. Data code = IT 39, yield

26%; 1H NMR (300MHz, CHLOROFORM-d) δ = 9.82 (br. s., 1 H), 7.72 (br. s., 1 H), 4.90 (s, 2 H), 4.72 (t, J = 6.6 Hz, 2 H),

3.86 (q, J = 6.9 Hz, 2 H), 3.73 (q, J = 6.7 Hz, 2 H), 2.82 (s, 3 H), 1.55 - 1.38 (m, 4 H), 1.32 (t, J = 7.0 Hz, 3 H), 1.18 (t, J = 7.2

Hz, 3 H), 1.01 - 0.91 (m, 3 H) ppm.

Ethanone, 1-[2-amino-4-(pentyloxy)-7,8-thiazolone)-6-pteridinyl]- C’

Method as reported in a publication [92] was followed: a suspension of B’ (8 g, 18.8 mmol) in a mixture of 250 mL acetic

acid and 500 mL of acetic anhydride was refluxed for 6 hrs. Then the volume of the solution was reduced almost to

dryness, the residue dissolved in chloroform and washed with water three times, then dried with sodium sulfate and

the solvent was removed on a rotary evaporator. The crude product (a dark brown solid) was purified by silica gel column

chromatography with ethyl acetate/n-hexane as eluent mixture 1:1 and crystallized in mixture of chloroform and n-

hexane. The purified substance was a bright red powder.

The collected powder is structure C’ but with the protected amino function (red solid, see below). The product was

dissolved in 500 mL of methanol and kept under stirring overnight, in this time the hydrolysis of the protection is

achieved. The volume of the solution reduced and the crude product recrystallized from a mixture of chloroform and n-

hexane.

Data code = IT 70, overall yield 35% (referring to the moles of product B’, comprehensive the hydrolysis of the protection

of the amino function), elemental analysis: calculated C-51.86, H-4.93, N-20.16, S-9.23; found C-51.57, H-4.68, N-19.54,

S-9.19; mass spectrometry (APCI, positive - negative, low t. low f.) = 348.3, 278.2 - 346.2 m/z; 1H NMR (300MHz,

CHLOROFORM-d) δ = 7.70 (s, 1 H), 5.69 (br. s., 2 H), 4.43 (t, J = 6.8 Hz, 2 H), 2.62 (s, 3 H), 2.04 (s, 1 H), 1.95 - 1.73 (m, 2

H), 1.56 - 1.33 (m, 4 H), 1.02 - 0.86 (m, 3 H); 13C NMR (75MHz, CHLOROFORM-d) δ = 198.0, 167.0, 166.2, 162.0, 149.4,

Page 117: epub.ub.uni-greifswald.de · Table of contents 1. Pteridines and pterins

113

141.5, 125.0, 108.3, 103.3, 67.9, 28.3, 28.1, 25.7, 22.3, 14.0; 13C NMR (75 MHz): δ = 14.0 (CH3), 22.3 (CH2), 25.7 (CH3),

28.1 (CH2), 28.2 (CH2), 67.9 (CH2), 103.3 (CH) ppm.

Ethanone, 1-[2-acetamido-4-(pentyloxy)-7,8-thiazolone)-6-pteridinyl]

Red solid

Data code = IT 59C1; 1H NMR (300MHz ,CHLOROFORM-d) δ = 8.16 (s, 1 H), 7.78 (s, 1 H), 4.50 (t, J = 6.8 Hz, 2 H), 2.68 (s,

3 H), 2.66 (s, 3 H), 1.88 (quin, J = 7.0 Hz, 2 H), 1.56 - 1.34 (m, 4 H), 1.04 - 0.86 (m, 3 H) ppm.

Ethanone, 1-[2-amino-7,8-thiazolone)-6-pteridinyl]

Product C’ without pentyloxy chain

The removal of the pentyloxy chain was achieved following the method reported in publication [93]: the product was

dissolved in a minimum amount of 0.1 N solution of hydrochloric acid and refluxed for 1 hr. After cooling, the precipitate

was collected, washed with H20 and EtOH, and then dried.

IT 83 (deprotected); yield 42% C-43.32, H-2.54, N-25.26, S-11.57; found C-41.27, H-3.22, N-21.9, S-10.06.

Complexation product (G’, IT 114)

The reaction was conducted under inert gas atmosphere (nitrogen). The product C’ (100 mg, 0.28 mmol) were added in

200 mL of methanol (oxygen free). Five mL of aqueous solution containing 0.5 equivalents of K3Na[MoO2(CN)4]*6H2O

and 10 equivalent of potassium hydroxide were added slowly (the water was first purged with nitrogen for around 30

minutes). The mixture was kept under reflux for 1 hr. The deep red colour solution was evaporated and the residue

washed with diethyl ether and acetonitrile (both solvents oxygen free). The final filtrate was washed with a small

amount of methanol (around 7 mL) and the resulting product dried under vacuum. Data code = IT 114 LAV; elemental

analysis: found C-32.11, H-2.73, N-18.91, S-4.78.

Page 118: epub.ub.uni-greifswald.de · Table of contents 1. Pteridines and pterins

114

3-(t-Butyldimethylsilyloxy)propan-1-ol G-first step

A procedure as reported in a publication [112] was followed: sodium hydride (80% suspension in oil, 7.5 g, 249 mmol)

was suspended in THF (500 mL) after being washed with n-hexane. 1,3-propanediol (15 mL, 15.8 g. 207.5 mmol) was

added to the mixture at RT and stirred for 45 min, after which time an opaque white precipitate had formed. Tert-

butyldimethylsilyl chloride (31.3 g, 207.5 mmol) was then added and vigorous stirring was continued for 45 min. The

mixture was poured into diethyl ether (500 mL), washed with 10% potassium carbonate (150 mL) and brine (150 mL),

dried with sodium sulfate and concentrated in vacuo. The resulting oil was purified by flash column chromatography

using ethyl acetate/petrol (1:4) as eluent. Data code = CT 1, yield 80%, mass spectrometry (APCI, positive, low t. low f.)

= 191.1. m/z.

3-(t-Butyldimethylsilyloxy)butanal G-second step

Method A as reported in a publication [112] was followed: a solution of oxalyl chloride (0.9 mL, 10 mmol) in

dichloromethane (100 mL) was cooled to -78°C (for cooling an ethyl acetate /N2 bath was used; -84°C) to which was

added dropwise dried dimethyl sulfoxide (1.5 mL, 20.8 mmol, dimethyl sulfoxide anhydrous ≥99.9%, sigma Aldrich).

After 15 min at -78°C a solution of product G-first step (1.75 g 8.58 mmol) in dichloromethane (20 mL) was added

dropwise and the reaction mixture stirred at -78°C for further 25-30 min. The reaction was quenched by the addition of

triethylamine (6 mL) and allowed to warm to RT. After 1 hr the reaction was poured into saturated aqueous sodium

bicarbonate solution (50 mL). The layers were separated and the aqueous layers extracted with dichloromethane. The

combined organic layers were washed with brine (50 mL), dried with sodium sulfate and concentrated in vacuo. Flash

column chromatography of the residue using 1:19 ethyl acetate: petrol as eluent, gave the desired product. Data code

= CT 4, yield 75%; 1H NMR (300MHz, CHLOROFORM-d) δ = 9.77 (t, J = 2.1 Hz, 1 H), 3.96 (t, J = 6.0 Hz, 2 H), 2.57 (dt, J =

2.1, 5.9 Hz, 2 H), 0.86 (s, 10 H) ppm; 13C NMR (75MHz, CHLOROFORM-d) δ = 201.9, 57.3, 46.5, 25.7, 18.1, -5.5 ppm; 13C

NMR (75 MHz): δ = -5.5 (CH3, CH3), 25.7 (CH3, CH3, CH3), 46.5 (CH2), 57.3 (CH2), 201.9 (CH) ppm.

2,2-Dimethyl-1,3-dioxolane-4-acetaldehyde (RS) H

The product was synthesized with the collaboration with the project member Mohsen Ahmadi and following the

procedures reported in the publications [123, 124]. The synthetic description will be reported in detail in the PhD thesis

of Mohsen Ahmadi.

Page 119: epub.ub.uni-greifswald.de · Table of contents 1. Pteridines and pterins

115

IR and UV-VIS spectra

6

7

8

Page 120: epub.ub.uni-greifswald.de · Table of contents 1. Pteridines and pterins

116

10

11

Page 121: epub.ub.uni-greifswald.de · Table of contents 1. Pteridines and pterins

117

13

14

15

Page 122: epub.ub.uni-greifswald.de · Table of contents 1. Pteridines and pterins

118

16

17

Page 123: epub.ub.uni-greifswald.de · Table of contents 1. Pteridines and pterins

119

18

20

Page 124: epub.ub.uni-greifswald.de · Table of contents 1. Pteridines and pterins

120

21

Page 125: epub.ub.uni-greifswald.de · Table of contents 1. Pteridines and pterins

121

23

26

Page 126: epub.ub.uni-greifswald.de · Table of contents 1. Pteridines and pterins

122

27

B’

C’

Page 127: epub.ub.uni-greifswald.de · Table of contents 1. Pteridines and pterins

123

Product C’ without pentyloxy chain

Page 128: epub.ub.uni-greifswald.de · Table of contents 1. Pteridines and pterins

124

Molecular structures and X-ray data

6

9

Identification code IT27CHF Empirical formula C12 H15 D Cl3 N5 O2 Formula weight 369.65 Temperature 170(2) K Wavelength 0.71073 A Crystal system, space group Monoclinic, P 21/c Unit cell dimensions a = 16.080(3) A alpha = 90 deg. b = 9.5697(19) A beta = 97.70(3) deg.

c = 10.649(2) A gamma = 90 deg. Volume 1624.0(6) A^3 Z, Calculated density 4, 1.512 Mg/m^3 Absorption coefficient 0.578 mm^-1 F(000) 760 Crystal size 0.458 x 0.228 x 0.207 mm Theta range for data collection 3.249 to 27.176 deg. Limiting indices -20<=h<=20, -12<=k<=12, -11<=l<=13 Reflections collected / unique 13736 / 3523 [R(int) = 0.0384] Completeness to theta = 25.242 99.7 % Absorption correction None Max. and min. transmission 0.9818 and 0.9447 Refinement method Full-matrix least-squares on F^2 Data / restraints / parameters 3523 / 0 / 212 Goodness-of-fit on F^2 1.061 Final R indices [I>2sigma(I)] R1 = 0.0364, wR2 = 0.0864 R indices (all data) R1 = 0.0524, wR2 = 0.0921 Extinction coefficient n/a Largest diff. peak and hole 0.440 and -0.390 e.A^-3

Identification code Shelx it 21f Empirical formula C9 H15 N5 O2 Formula weight 225.26 Temperature 170(2) K Wavelength 0.71073 A Crystal system, space group Monoclinic, C 2/c Unit cell dimensions a = 13.610(3) A alpha = 90 deg. b = 9.884(2) A beta = 111.06(3) deg.

c = 17.359(4) A gamma = 90 deg. Volume 2179.1(9) A^3 Z, Calculated density 8,1.373 Mg/m^3 Absorption coefficient 0.101 mm^-1 F(000) 960 Crystal size 0.380 x 0.279 x 0.230 mm Theta range for data collection 3.201 to 29.245 deg. Limiting indices -18<=h<=15, -13<=k<=13, -23<=l<=23 Reflections collected / unique 11888 / 2954 [R(int) = 0.0399] Completeness to theta = 25.242 99.7 % Absorption correction None Max. and min. transmission 0.9818 and 0.9447 Refinement method Full-matrix least-squares on F^2 Data / restraints / parameters 2954 / 0 / 207 Goodness-of-fit on F^2 0.524 Final R indices [I>2sigma(I)] R1 = 0.0393, wR2 = 0.1049 R indices (all data) R1 = 0.0500, wR2 = 0.1223 Extinction coefficient n/a Largest diff. peak and hole 0.326 and -0.184 e.A^-3

Page 129: epub.ub.uni-greifswald.de · Table of contents 1. Pteridines and pterins

125

11

14

Identification code CTCl Empirical formula C11 H14 Cl N5 O Formula weight 267.7 Temperature 170(2) K Wavelength 0.71073 A Crystal system, space group Monoclinic, P 21/c Unit cell dimensions a = 10.538(2) A alpha = 90 deg. b = 4.6688(9) A beta = 97.42(3) deg.

c = 26.226(5) A gamma = 90 deg. Volume 1279.5(4) A^3 Z, Calculated density 4, 1.390 Mg/m^3 Absorption coefficient 0.295 mm^-1 F(000) 560 Crystal size 0.493 x 0.452 x 0.123 mm Theta range for data collection 1.949 to 26.371 deg. Limiting indices -13<=h<=13, -5<=k<=5, -32<=l<=29 Reflections collected / unique 10316 / 2608 [R(int) = 0.0833] Completeness to theta = 25.242 99.9 % Absorption correction Numerical Max. and min. transmission 0.8180 and 0.4728 Refinement method Full-matrix least-squares on F^2 Data / restraints / parameters 2608 / 0 / 173 Goodness-of-fit on F^2 0.954 Final R indices [I>2sigma(I)] R1 = 0.0520, wR2 = 0.1181 R indices (all data) R1 = 0.1079, wR2 = 0.1476 Extinction coefficient 0.014(3) Largest diff. peak and hole 0.252 and -0.285 e.A^-3

Identification code Shelx IT 29 Empirical formula C14 H19 N5 O2 Formula weight 289.34 Temperature 293(2) K Wavelength 0.71073 A Crystal system, space group Monoclinic, P 21/n linic, P -1 Unit cell dimensions a = 13.746(3) A alpha = 90 deg. b = 7.0835(14) A beta = 104.97(3) deg.

c = 15.970(3) A gamma = 90 deg. Volume 1502.2(6) A^3 Z, Calculated density 4, 1.279 Mg/m^3 Absorption coefficient 0.089 mm^-1 F(000) 616 Crystal size 0.440 x 0.132 x 0.076 mm Theta range for data collection 3.165 to 29.270 deg. Limiting indices -17<=h<=18, -9<=k<=9, -21<=l<=21 Reflections collected / unique 15884 / 4059 [R(int) = 0.1730] Completeness to theta = 25.242 99.8 % Absorption correction None Max. and min. transmission 0.9818 and 0.9447 Refinement method Full-matrix least-squares on F^2 Data / restraints / parameters 4059 / 1 / 206 Goodness-of-fit on F^2 0.910 Final R indices [I>2sigma(I)] R1 = 0.0580, wR2 = 0.1285 R indices (all data) R1 = 0.1545, wR2 = 0.2512 Extinction coefficient 0.040(6) Largest diff. peak and hole 0.353 and -0.377 e.A^-3

Page 130: epub.ub.uni-greifswald.de · Table of contents 1. Pteridines and pterins

126

15

16

Identification code MADXM5a Empirical formula C15 H25 N5 O2 S Formula weight 339.46 Temperature 170(2) K Wavelength 0.71073 A Crystal system, space group Triclinic, P -1 Unit cell dimensions a = 9.765(2) A alpha = 71.03(3) deg. b = 12.019(2) A beta = 80.36(3) deg.

c = 16.561(3) A gamma = 79.22(3) deg. Volume 1793.9(7) A^3 Z, Calculated density 4, 1.257 Mg/m^3 Absorption coefficient 0.197 mm^-1 F(000) 728 Crystal size 0.402 x 0.181 x 0.099 mm Theta range for data collection 3.247 to 26.372 deg. Limiting indices -12<=h<=12, -14<=k<=15, -20<=l<=20 Reflections collected / unique 15005 / 7288 [R(int) = 0.0901] Completeness to theta = 25.000 99.5 % Absorption correction None Max. and min. transmission 0.9818 and 0.9447 Refinement method Full-matrix least-squares on F^2 Data / restraints / parameters 7288 / 41 / 44296 / 323 / 444 Goodness-of-fit on F^2 0.984 Final R indices [I>2sigma(I)] R1 = 0.0706, wR2 = 0.1659 R indices (all data) R1 = 0.1418, wR2 = 0.2079 Extinction coefficient n/a Largest diff. peak and hole 0.243 and -0.347 e.A^-3

Identification code Shelx, IT 90 Empirical formula C17 H25 N5 O2 S Formula weight 363.48 Temperature 293(2) K Wavelength 0.71073 A Crystal system, space group Monoclinic, P 21/c Unit cell dimensions a = 4.5429(9) A alpha = 90 deg. b = 17.490(4) A beta = 92.92(3) deg.

c = 24.684(5) A gamma = 90 deg. Volume 1958.7(7) A^3 Z, Calculated density 4, 1.233 Mg/m^3 Absorption coefficient 0.185 mm^-1 F(000) 776 Crystal size 0.445 x 0.066 x 0.045 mm Theta range for data collection 1.428 to 24.730 deg. Limiting indices -5<=h<=5, -20<=k<=20, 0<=l<=28 Reflections collected / unique 6452 / 3304 [R(int) = 0.3422] Completeness to theta = 25.000 95.5 % Absorption correction Numerical Max. and min. transmission 0.9236 and 0.5309 Refinement method Full-matrix least-squares on F^2 Data / restraints / parameters 3304 / 0 / 226 Goodness-of-fit on F^2 0.817 Final R indices [I>2sigma(I)] R1 = 0.1125, wR2 = 0.2073 R indices (all data) R1 = 0.4148, wR2 = 0.3529 Extinction coefficient n/a Largest diff. peak and hole 0.249 and -0.240 e.A^-3

Page 131: epub.ub.uni-greifswald.de · Table of contents 1. Pteridines and pterins

127

17

Identification code CT39 Empirical formula C16 H23 N5 O2 S Formula weight 349.45 Temperature 170(2) K Wavelength 0.71073 A Crystal system, space group Monoclinic, P 21/n Unit cell dimensions a = 4.2777(9) A alpha = 90 deg. b = 23.560(5) A beta = 94.44(3) deg.

c = 17.673(3) A gamma = 90 deg. Volume 1775.8(6) A^3 Z, Calculated density 4, 1.307 Mg/m^3 Absorption coefficient 0.201 mm^-1 F(000) 744 Crystal size 3.459 x 0.065 x 0.063 mm Theta range for data collection 3.459 to 25.689 deg. Limiting indices -5<=h<=5, -28<=k<=28, -21<=l<=21 Reflections collected / unique 12607 / 3359 [R(int) = 0.1173] Completeness to theta = 25.242 99.4 % Absorption correction Numerical Max. and min. transmission 0.9972 and 0.8172 Refinement method Full-matrix least-squares on F^2 Data / restraints / parameters 3359 / 1 / 228 Goodness-of-fit on F^2 0.989 Final R indices [I>2sigma(I)] R1 = 0.0569, wR2 = 0.1083 R indices (all data) R1 = 0.1410, wR2 = 0.1402 Extinction coefficient n/a Largest diff. peak and hole 0.311 and -0.345 e.A^-3

18

Identification code IT103FIN Empirical formula C21 H31 N5 O3 S3 Formula weight 497.69 Temperature 170(2) K Wavelength 0.71073 A Crystal system, space group Triclinic, P -1 Unit cell dimensions a = 7.8816(12) A alpha = 92.603(12) deg. b = 10.5953(17) A beta = 95.277(11)

deg. c = 15.836(2) A gamma = 106.688(12) deg. Volume 1257.8(3) A^3 Z, Calculated density 2, 1.314 Mg/m^3 Absorption coefficient 0.326 mm^-1 F(000) 528 Crystal size 0.481 x 0.078 x 0.050 mm Theta range for data collection 3.399 to 24.710 deg. Limiting indices -9<=h<=8, -12<=k<=12, -18<=l<=18 Reflections collected / unique 9015 / 4263 [R(int) = 0.1592] Completeness to theta = 24.710 99.5 % Absorption correction Numerical Max. and min. transmission 0.9869 and 0.8403 Refinement method Full-matrix least-squares on F^2 Data / restraints / parameters 4263 / 70 / 328 Goodness-of-fit on F^2 0.966 Final R indices [I>2sigma(I)] R1 = 0.0862, wR2 = 0.1520 R indices (all data) R1 = 0.2370, wR2 = 0.2070 Extinction coefficient n/a Largest diff. peak and hole 0.319 and -0.308 e.A^-3

Page 132: epub.ub.uni-greifswald.de · Table of contents 1. Pteridines and pterins

128

19

24

Identification code IT122 Empirical formula C22 H34 N6 O2 S3 Formula weight 510.73 Temperature 170(2) K Wavelength 0.71073 A Crystal system, space group Triclinic, P -1 Unit cell dimensions a = 6.9442(14) A alpha = 86.74(3) deg. b = 13.200(3) A beta = 89.22(3) deg.

c = 14.319(3) A gamma = 89.28(3) deg. Volume 1310.2(5) A^3 Z, Calculated density 2, 1.295 Mg/m^3 Absorption coefficient 0.313 mm^-1 F(000) 544 Crystal size 0.138 x 0.104 x 0.080 mm Theta range for data collection 3.164 to 23.256 deg. Limiting indices -6<=h<=7, -14<=k<=14, -15<=l<=15 Reflections collected / unique 7747 / 3733 [R(int) = 0.0611] Completeness to theta = 23.256 99.0 % Absorption correction None Max. and min. transmission 0.9869 and 0.8403 Refinement method Full-matrix least-squares on F^2 Data / restraints / parameters 3733 / 101 / 357 Goodness-of-fit on F^2 0.840 Final R indices [I>2sigma(I)] R1 = 0.0439, wR2 = 0.0781 R indices (all data) R1 = 0.1134, wR2 = 0.0960 Extinction coefficient n/a Largest diff. peak and hole 0.170 and -0.213 e.A^-3

Identification code MAD205F, ACT 20 Empirical formula C23 H35 N5 O6 S Formula weight 509.62 Temperature 170(2) K Wavelength 0.71073 A Crystal system, space group Triclinic, P -1 Unit cell dimensions a = 8.8777(18) A alpha = 96.70(3) deg. b = 9.2872(19) A beta = 93.09(3) deg.

c = 16.197(3) A gamma = 93.58(3) deg. Volume 1321.2(5) A^3 Z, Calculated density 2, 1.281 Mg/m^3 Absorption coefficient 0.168 mm^-1 F(000) 544 Crystal size 0.232 x 0.108 x 0.071 mm Theta range for data collection 3.160 to 25.679 deg. Limiting indices -10<=h<=10, -11<=k<=11, -19<=l<=19 Reflections collected / unique 10478 / 4996 [R(int) = 0.0606] Completeness to theta = 25.242 99.7 % Absorption correction Numerical Max. and min. transmission 0.9818 and 0.9447 Refinement method Full-matrix least-squares on F^2 Data / restraints / parameters 4996 / 323 / 444 Goodness-of-fit on F^2 0.907 Final R indices [I>2sigma(I)] R1 = 0.0580, wR2 = 0.1285 R indices (all data) R1 = 0.1392, wR2 = 0.1622 Extinction coefficient n/a Largest diff. peak and hole 0.365 and -0.240 e.A^-3

Page 133: epub.ub.uni-greifswald.de · Table of contents 1. Pteridines and pterins

129

27

E’

Identification code IT128F Empirical formula C18 H25 N5 O3 S2 Formula weight 423.55 Temperature 170(2) K Wavelength 0.71073 A Crystal system, space group Triclinic, P -1 Unit cell dimensions a = 10.845(2) A alpha = 113.34(3) deg. b = 14.928(3) A beta = 107.35(3) deg.

c = 15.236(3) A gamma = 94.38(3) deg. Volume 2107.8(9) A^3 Z, Calculated density 4, 1.335 Mg/m^3 Absorption coefficient 0.281 mm^-1 F(000) 896 Crystal size 0.239 x 0.204 x 0.092 mm Theta range for data collection 3.261 to 23.257 deg. Limiting indices -12<=h<=12, -16<=k<=16, -16<=l<=16 Reflections collected / unique 13507 / 6003 [R(int) = 0.0782] Completeness to theta = 23.257 99.3 % Absorption correction Numerical Max. and min. transmission 0.9946 and 0.9083 Refinement method Full-matrix least-squares on F^2 Data / restraints / parameters 6003 / 149 / 543 Goodness-of-fit on F^2 1.004 Final R indices [I>2sigma(I)] R1 = 0.0902, wR2 = 0.2475 R indices (all data) R1 = 0.2353, wR2 = 0.3209 Extinction coefficient n/a Largest diff. peak and hole 0.345 and -0.445 e.A^-3

Identification code IT70c-19 Empirical formula C15 H18 N5 O4 S Formula weight 364.40 Temperature 100(2) K Wavelength 0.6199 A Crystal system, space group Orthorhombic, P c a 21 Unit cell dimensions a = 30.990(6) A alpha = 90 deg. b = 3.9700(8) A beta = 90 deg.

c = 25.430(5) A gamma = 90 deg. Volume 3128.7(11) A^3 Z, Calculated density 8, 1.547 Mg/m^3 Absorption coefficient 0.241 mm^-1 F(000) 1528 Crystal size 150 x 20 x 20 um Theta range for data collection 1.146 to 18.057 deg. Limiting indices -30<=h<=30, -3<=k<=3, -25<=l<=25 Reflections collected / unique 10970 / 3190 [R(int) = 0.0725] Completeness to theta = 18.057 98.9 % Absorption correction None Absolute structure parameter 0.6(10) Refinement method Full-matrix least-squares on F^2 Data / restraints / parameters 3190 / 2 / 161 Goodness-of-fit on F^2 2.783 Final R indices [I>2sigma(I)] R1 = 0.2388, wR2 = 0.5420 R indices (all data) R1 = 0.2643, wR2 = 0.5796 Extinction coefficient n/a Largest diff. peak and hole 2.810 and -2.814 e.A^-3

Page 134: epub.ub.uni-greifswald.de · Table of contents 1. Pteridines and pterins

130

F’

Identification code shelx Empirical formula C9 H17 N5 Na2 O6 S Formula weight 369.32 Temperature 170(2) K Wavelength 0.71073 A Crystal system, space group Monoclinic, P 21/c Unit cell dimensions a = 15.111(3) A alpha = 90 deg. b = 12.922(3) A beta = 90.51(3) deg.

c = 9.0809(18) A gamma = 90 deg. Volume 1773.1(6) A^3 Z, Calculated density 4, 1.383 Mg/m^3 Absorption coefficient 0.264 mm^-1 F(000) 768 Crystal size 0.171 x 0.147 x 0.085 mm Theta range for data collection 3.046 to 24.619 deg. Limiting indices -17<=h<=15, -15<=k<=15, -10<=l<=10 Reflections collected / unique 11683 / 2967 [R(int) = 0.0747] Completeness to theta = 25.242 92.6 % Absorption correction None Absolute structure parameter 0.6(10) Refinement method Full-matrix least-squares on F^2 Data / restraints / parameters 2967 / 13 / 250 Goodness-of-fit on F^2 0.932 Final R indices [I>2sigma(I)] R1 = 0.0559, wR2 = 0.1428 R indices (all data) R1 = 0.0961, wR2 = 0.1590 Extinction coefficient n/a Largest diff. peak and hole 0.348 and -0.394 e.A^-3

Page 135: epub.ub.uni-greifswald.de · Table of contents 1. Pteridines and pterins

131

References

[1] D.J. Brown, Fused pyrimidines-Pteridines, Wiley, New York, 1988. [2] J.E. Ayling, in, Plenum Press, New York [u.a.], 1993, pp. XXVI, 825. [3] F. Wöhler, Justus Liebigs Annalen der Chemie, 103 (1857) 117-118. [4] H. Hlasiwetz, Justus Liebigs Annalen der Chemie, 103 (1857) 200-218. [5] F.G. Hopkins, Proc. R. Soc. London, Ser. B, 130 (1942) 359-379. [6] W. Pfleiderer, Chemische Berichte, 92 (1959) 2468-2477. [7] O. Kühling, Berichte der deutschen chemischen Gesellschaft, 27 (1894) 2116-2119. [8] S. Gabriel, A. Sonn, Berichte der deutschen chemischen Gesellschaft, 40 (1907) 4850-4860. [9] R. Kuhn, A.H. Cook, Berichte der deutschen chemischen Gesellschaft (A and B Series), 70 (1937) 761-768. [10] S. Gabriel, J. Colman, Berichte der deutschen chemischen Gesellschaft, 34 (1901) 1234-1257. [11] D.J. Brown, Pteridines, Wiley, New York, 1988. [12] F.G. Hopkins, Proceedings of the Chemical Society, London, 5 (1889) 105-126. [13] H. Wieland, R. Purrmann, Justus Liebigs Ann. Chem., 544 (1940) 163-182. [14] R. Purrmann, Justus Liebigs Ann. Chem., 544 (1940) 182-190. [15] H. Wieland, A. Tartter, R. Purrmann, Justus Liebigs Ann. Chem., 545 (1940) 209-219. [16] R. Purrmann, Ann., 546 (1940) 98-102. [17] W. Pfleiderer, in, De Gruyter, Chemistry and biology of pteridines : proceedings of the 5. international symposium held at the Univ. of Konstanz, West Germany, 1975, pp. XVI, 949. [18] J.A. Blair, in, De Gruyter, Berlin [u.a.], Chemistry and biology of pteridines : pteridines and folic acid derivatives; proceedings of the 7. International Symposium on Pteridines and Folic Acid Derivatives, Chemical, Biological and Clinical Aspects, 1983, pp. XXXIV, 1070. [19] E. Hückel, Zeitschrift für Physik, 70 (1931) 204-286. [20] D.T. Hurst, An introduction to the chemistry and biochemistry of pyrimidines, purines and pteridines, Wiley, Chichester [u.a.], 1980. [21] P. Basu, S.J.N. Burgmayer, Coordination chemistry reviews, 255 (2011) 1016-1038. [22] H. Wieland, C. Schöpf, Berichte der deutschen chemischen Gesellschaft (A and B Series), 58 (1925) 2178-2183. [23] M. Vignoni, N. Walalawela, S.M. Bonesi, A. Greer, A.H. Thomas, Mol. Pharmaceutics, 15 (2018) 798-807. [24] T.A. Telegina, T.A. Lyudnikova, A.A. Buglak, Y.L. Vechtomova, M.V. Biryukov, V.V. Demin, M.S. Kritsky, Journal of Photochemistry and Photobiology A: Chemistry, 354 (2018) 155-162. [25] E. Sandra, L. Carolina, K.T. S., L.E. L., T.A. H., S.M. P., Photochemistry and Photobiology, (2018) 1. [26] M. Viscontini, H. Raschig, Helvetica Chimica Acta, 41 (1958) 108-113. [27] H.S. Forrest, H.K. Mitchell, J. Am. Chem. Soc., 77 (1955) 4865-4869. [28] M. Vignoni, F.M. Cabrerizo, C. Lorente, A.H. Thomas, Photochemistry and Photobiology, 85 (2009) 365-373. [29] E.C. Taylor, W. Pfleiderer, Pteridine Chemistry: Proceedings of the Third International Symposium Held at the Institut Fur ̈Organische Chemie Der Technischen Hochschule, Stuttgart, September 1962, Pergamon Press, 1964. [30] D.J. Brown, S.F. Mason, Journal of the Chemical Society (Resumed), (1956) 3443-3453. [31] A. Albert, F. Reich, J. Chem. Soc., (1961) 127-135. [32] W. Pfleiderer, Journal of Heterocyclic Chemistry, 29 (1992) 583-605. [33] A. Michael, Journal für Praktische Chemie, 35 (1887) 349-356. [34] e. D. J. Brown, Chemistry of Heterocyclic Compounds T3-Fused Pyrimidines - Part III: Pteridines, 1988. [35] R. Baur, T. Sugimoto, W. Pfleiderer, Helv. Chim. Acta, 71 (1988) 531-543. [36] A. Albert, J.J. McCormack, J. Chem. Soc. C, (1966) 1117-1120. [37] C.E. Perez, H.B. Park, J.M. Crawford, Biochemistry, 57 (2018) 354-361.

Page 136: epub.ub.uni-greifswald.de · Table of contents 1. Pteridines and pterins

132

[38] M. Fischer, B. Thöny, S. Leimkühler, 7.17 - The Biosynthesis of Folate and Pterins and Their Enzymology, in: Comprehensive Natural Products II, Elsevier, Oxford, 2010, pp. 599-648. [39] P.L. Smedley, D.G. Kinniburgh, Applied Geochemistry, 84 (2017) 387-432. [40] C. Iobbi-Nivol, S. Leimkühler, Biochimica et Biophysica Acta (BBA) - Bioenergetics, 1827 (2013) 1086-1101. [41] S. Ramaswamy, Science (Washington, DC, U. S.), 334 (2011) 914-915. [42] T. Spatzal, M. Aksoyoglu, L. Zhang, S.L.A. Andrade, E. Schleicher, S. Weber, D.C. Rees, O. Einsle, Science (Washington, DC, U. S.), 334 (2011) 940. [43] K.M. Lancaster, M. Roemelt, P. Ettenhuber, Y. Hu, M.W. Ribbe, F. Neese, U. Bergmann, S. DeBeer, Science (Washington, DC, U. S.), 334 (2011) 974-977. [44] I. Pikaar, S. Matassa, K. Rabaey, B.L. Bodirsky, A. Popp, M. Herrero, W. Verstraete, Environmental Science & Technology, 51 (2017) 7297-7303. [45] K.V. Rajagopalan, J.L. Johnson, J. Biol. Chem., 267 (1992) 10199-10202. [46] M.K. Chan, S. Mukund, A. Kletzin, M.W. Adams, D.C. Rees, Science, 267 (1995) 1463-1469. [47] C. Iobbi-Nivol, S. Leimkuhler, Biochim. Biophys. Acta, Bioenerg., 1827 (2013) 1086-1101. [48] D.M. Pitterle, J.L. Johnson, K.V. Rajagopalan, J. Biol. Chem., 268 (1993) 13506-13509. [49] E.N.G. Marsh, D.P. Patterson, L. Li, ChemBioChem, 11 (2010) 604-621. [50] J.L. Vey, C.L. Drennan, Chem. Rev. (Washington, DC, U. S.), 111 (2011) 2487-2506. [51] B.M. Hover, A. Loksztejn, A.A. Ribeiro, K. Yokoyama, Journal of the American Chemical Society, 135 (2013) 7019-7032. [52] A.P. Mehta, J.W. Hanes, S.H. Abdelwahed, D.G. Hilmey, P. Hanzelmann, T.P. Begley, Biochemistry, 52 (2013) 1134-1136. [53] W.R. Hagen, Coordination Chemistry Reviews, 255 (2011) 1117-1128. [54] D.C. Rees, E. Johnson, O. Lewinson, Nat. Rev. Mol. Cell Biol., 10 (2009) 218-227. [55] K. Hollenstein, R.J.P. Dawson, K.P. Locher, Curr. Opin. Struct. Biol., 17 (2007) 412-418. [56] P.M. McNicholas, S.A. Rech, R.P. Gunsalus, Mol. Microbiol., 23 (1997) 515-524. [57] S. Leimkuhler, K.V. Rajagopalan, J. Biol. Chem., 276 (2001) 1837-1844. [58] O. Genest, V. Méjean, C. Iobbi‐Nivol, FEMS Microbiology Letters, 297 (2009) 1-9. [59] G. Schwarz, Current Opinion in Chemical Biology, 31 (2016) 179-187. [60] J.M. Klein, J.D. Busch, C. Potting, M.J. Baker, T. Langer, G. Schwarz, J. Biol. Chem., 287 (2012) 42795-42803. [61] A. Havemeyer, J. Lang, B. Clement, Drug Metab. Rev., 43 (2011) 524-539. [62] P.S. Atwal, F. Scaglia, Molecular Genetics and Metabolism, 117 (2016) 1-4. [63] A. Veldman, J.A. Santamaria-Araujo, S. Sollazzo, J. Pitt, R. Gianello, J. Yaplito-Lee, F. Wong, C.A. Ramsden, J. Reiss, I. Cook, J. Fairweather, G. Schwarz, Pediatrics, 125 (2010) e1249-e1254. [64] K.V. Rajagopalan, J.L. Johnson, J. Biol. Chem., 267 (1992) 10199-10202. [65] B. Bradshaw, A. Dinsmore, W. Ajana, D. Collison, C.D. Garner, J.A. Joule, J. Chem. Soc., Perkin Trans. 1, (2001) 3239-3244. [66] A. Dinsmore, C.D. Garner, J.A. Joule, Tetrahedron, 54 (1998) 9559-9568. [67] A. Dinsmore, C. David Garner, J.A. Joule, Tetrahedron, 54 (1998) 3291-3302. [68] B. Bradshaw, A. Dinsmore, D. Collison, C.D. Garner, J.A. Joule, J. Chem. Soc., Perkin Trans. 1, (2001) 3232-3238. [69] G.D. Allred, L.S. Liebeskind, J. Am. Chem. Soc., 118 (1996) 2748-2749. [70] F.W. Fowler, The Journal of Organic Chemistry, 37 (1972) 1321-1323. [71] E.R. Wijsman, O. van den Berg, E. Kuyl-Yeheskiely, G.A. van der Marel, J.H. van Boom, Recl. Trav. Chim. Pays-Bas, 113 (1994) 337-338. [72] B.R. Williams, Y. Fu, G.P.A. Yap, S.J.N. Burgmayer, Journal of the American Chemical Society, 134 (2012) 19584-19587. [73] B.R. Williams, D. Gisewhite, A. Kalinsky, A. Esmail, S.J.N. Burgmayer, Inorganic Chemistry, 54 (2015) 8214-8222. [74] D.J. Rowe, C.D. Garner, J.A. Joule, Journal of the Chemical Society, Perkin Transactions 1, (1985) 1907-1910.

Page 137: epub.ub.uni-greifswald.de · Table of contents 1. Pteridines and pterins

133

[75] E. Stephen Davies, R. L. Beddoes, D. Collison, A. Dinsmore, A. Docrat, J. A. Joule, C. R. Wilson, C. David Garner, Journal of the Chemical Society, Dalton Transactions, (1997) 3985-3996. [76] A. Dinsmore, J. H. Birks, C. David Garner, J. A. Joule, Journal of the Chemical Society, Perkin Transactions 1, (1997) 801-808. [77] U. Ryde, C. Schulzke, K. Starke, JBIC, J. Biol. Inorg. Chem., 14 (2009) 1053-1064. [78] S.J. Nieter Burgmayer, D.L. Pearsall, S.M. Blaney, E.M. Moore, C. Sauk-Schubert, JBIC, J. Biol. Inorg. Chem., 9 (2004) 59-66. [79] W. Kaim, B. Schwederski, Coordination Chemistry Reviews, 254 (2010) 1580-1588. [80] P. Basu, S.J.N. Burgmayer, Coordination Chemistry Reviews, 255 (2011) 1016-1038. [81] G.B. Rosso, SYNLETT Spotlight 156. [82] D. Mohr, Z. Kazimierczuk, W. Pfleiderer, Helv. Chim. Acta, 75 (1992) 2317-2326. [83] E.C. Taylor, R.F. Abdulla, K. Tanaka, P.A. Jacobi, The Journal of Organic Chemistry, 40 (1975) 2341-2347. [84] G.M. Timmis, Nature, 164 (1949) 139. [85] G. Heizmann, W. Pfleiderer, Helv. Chim. Acta, 90 (2007) 1856-1873. [86] D.V.C. Awang, S. Wolfe, Can. J. Chem., 47 (1969) 706-709. [87] D.V.C. Awang, S. Wolfe, Can. J. Chem., 47 (1969) 706-709. [88] U. Ewers, H. Guenther, L. Jaenicke, Chem. Ber., 107 (1974) 3275-3286. [89] U. Ewers, H. Guenther, L. Jaenicke, Chem. Ber., 107 (1974) 876-886. [90] E. Ulrich, G. Harald, J. Lothar, Chemische Berichte, 106 (1973) 3951-3961. [91] G. Mueller, W. Von Philipsborn, Helv. Chim. Acta, 56 (1973) 2680-2687. [92] D. Mohr, in, Konstanz, Synthese, "Reaktionen und Eigenschaften neuer 6- und 7-substituierter C-S- und C-C- verknüpfter Pteridine : Modellreaktionen für Naturstoffsynthesen", 1991, pp. VI, 203. [93] D. Mohr, Z. Kazimierczuk, W. Pfleiderer, Helv. Chim. Acta, 75 (1992) 2317-2326. [94] A.A. Kalinin, V.A. Mamedov, Chemistry of Heterocyclic Compounds, 40 (2004) 129-131. [95] A.H. Cook, I. Heilbron, A.L. Levy, Journal of the Chemical Society (Resumed), (1947) 1594-1598. [96] N. Taxak, V. Parmar, D.S. Patel, A. Kotasthane, P.V. Bharatam, The Journal of Physical Chemistry A, 115 (2011) 891-898. [97] N. Taxak, V.A. Dixit, P.V. Bharatam, J. Phys. Chem. A, 116 (2012) 10441-10450. [98] K.V. Rajagopalan, J.L. Johnson, J. Biol. Chem., 267 (1992) 10199-10202. [99] S.P. Kaiwar, J.K. Hsu, A. Vodacek, G. Yap, L.M. Liable-Sands, A.L. Rheingold, R.S. Pilato, Inorg. Chem., 36 (1997) 2406-2412. [100] W. Pfleiderer, 6. Winter Workshop on Pteridines, February 14 - 21, 1987, St. Christoph, Arlberg, Austria. Ed.: W. Pfleiderer, 1987. [101] F. Minisci, Synthesis, (1973) 1-24. [102] F. Minisci, A. Citterio, E. Vismara, C. Giordano, Tetrahedron, 41 (1985) 4157-4170. [103] D.J. Mackinnon, W.A. Waters, Journal of the Chemical Society (Resumed), (1953) 323-328. [104] T. Caronna, G. Fronza, F. Minisci, O. Porta, Journal of the Chemical Society, Perkin Transactions 2, (1972) 2035-2038. [105] T. Caronna, G.P. Gardini, F. Minisci, Journal of the Chemical Society D: Chemical Communications, (1969) 201-201. [106] F. Minisci, E. Vismara, F. Fontana, G. Morini, M. Serravalle, C. Giordano, J. Org. Chem., 51 (1986) 4411-4417. [107] C. Chatgilialoglu, D. Crich, M. Komatsu, I. Ryu, Chemical Reviews, 99 (1999) 1991-2070. [108] J.P. Smit, W. Purcell, A. Roodt, J.G. Leipoldt, Polyhedron, 12 (1993) 2271-2277. [109] H.L. Kaufmann, L. Liable-Sands, A.L. Rheingold, S.J.N. Burgmayer, Inorganic Chemistry, 38 (1999) 2592-2599. [110] O. Lutz, A. Nolle, P. Kroneck, Z. Naturforsch., A, 31A (1976) 454-456. [111] R.I. Maksimovskaya, G.M. Maksimov, Inorg. Chem., 46 (2007) 3688-3695. [112] J.C. Conway, P. Quayle, A.C. Regan, C.J. Urch, Tetrahedron, 61 (2005) 11910-11923. [113] T.H. Graham, W. Liu, D.-M. Shen, Org. Lett., 13 (2011) 6232-6235. [114] E.C. Taylor, K.L. Perlman, Y.-H. Kim, I.P. Sword, P.A. Jacobi, J. Amer. Chem. Soc., 95 (1973) 6413-6418. [115] E.C. Taylor, R. Kobylecki, The Journal of Organic Chemistry, 43 (1978) 680-683.

Page 138: epub.ub.uni-greifswald.de · Table of contents 1. Pteridines and pterins

134

[116] F. Marchetti, C. Cano, N.J. Curtin, B.T. Golding, R.J. Griffin, K. Haggerty, D.R. Newell, R.J. Parsons, S.L. Payne, L.-Z. Wang, I.R. Hardcastle, Org. Biomol. Chem., 8 (2010) 2397-2407. [117] V. Mesguiche, R.J. Parsons, C.E. Arris, J. Bentley, F.T. Boyle, N.J. Curtin, T.G. Davies, J.A. Endicott, A.E. Gibson, B.T. Golding, R.J. Griffin, P. Jewsbury, L.N. Johnson, D.R. Newell, M.E.M. Noble, L.Z. Wang, I.R. Hardcastle, Bioorg. Med. Chem. Lett., 13 (2003) 217-222. [118] H. Schmid, M. Schranner, W. Pfleiderer, Chem. Ber., 106 (1973) 1952-1975. [119] Y. Kuroda, K. Isarai, S. Murata, Heterocycl. Commun., 18 (2012) 117-122. [120] U. Schmidt, J. Langner, B. Kirschbaum, C. Braun, Synthesis, (1994) 1138-1140. [121] A. Heckel, W. Pfleiderer, Helv. Chim. Acta, 69 (1986) 708-717. [122] T.H. Graham, W. Liu, D.-M. Shen, Org. Lett., 13 (2011) 6232-6235. [123] P. Patel, V. Gore, W.S. Powell, J. Rokach, Bioorganic & Medicinal Chemistry Letters, 21 (2011) 1987-1990. [124] Z. Guo, R. Padmakumar, R.I. Hollingsworth, K.V. Radhakrishnan, M.V. Nandakumar, B. Fraser-Reid, Synlett, (2003) 1067-1069. [125] G.A. Ardizzoia, S. Brenna, S. Durini, B. Therrien, I. Trentin, Dalton Trans., 42 (2013) 12265-12273.

Page 139: epub.ub.uni-greifswald.de · Table of contents 1. Pteridines and pterins

135

Eigenständigkeitserklärung

Hiermit erkläre ich, dass diese Arbeit bisher von mir weder an der Mathematisch Naturwissenschaftlichen

Fakultät der Ernst-Moritz-Arndt-Universität Greifswald noch einer anderen wissenschaftlichen Einrichtung

zum Zwecke der Promotion eingereicht wurde.

Ferner erkläre ich, dass ich diese Arbeit selbstständig verfasst und keine anderen als die darin angegebenen

Hilfsmittel und Hilfen benutzt und keine Textabschnitte eines Dritten ohne Kennzeichnung übernommen habe.

Unterschrift des Promovenden

Page 140: epub.ub.uni-greifswald.de · Table of contents 1. Pteridines and pterins

136

Lebenslauf - Ivan Trentin

Schulbildung und Studium seit 10/2012 Promotion – Universität Greifswald, Institut für Biochemie MOCOMODELS‐European Research Council „Modelling the Molybdenum cofactor, synthesis of pterin-dithiolene ligand system “ Kooperation: Institut für Molekulare Enzymologie – Universität Potsdam Betreuerin: Prof. Dr. rer. nat. Carola Schulzke 03/2012‐ 10/2003 Studium der Chemie – Università degli Studi dell’ Insubria – Como – Italien 03/2012‐ 09/2007 Chemie Master of Science Masterarbeit im Fach anorganische und metallorganische Chemie „Synthese und Charakterisierung von polynukleare pyrazol-komplexen mit Kupfer(II).“Betreuer: Prof. Dr. G. Attilio Ardizzoia, Dr. Stefano Brenna 10/2010‐ 09/2010 Auslandsaufenthalt an der Sprachschule „Bridge Mills“ Galway – Irland Intensivkurs mit Zertifikat C1 10/2009‐ 10/2008 Auslandsstudium – Universität Bremen SOKRATES Stipendium ‐ ERASMUS Programm Besuch von Polymerchemie Vorlesungen und Erlernen von physikalischen Methoden 10/2007‐ 10/2003 Chemie Bachelor of Science Bachelorarbeit im Fach anorganische und metallorganische Chemie „Synthese und Charakterisierung von Koordinationspolymeren der Gruppe11 Betreuer: Prof. Dr. G. Attilio Ardizzoia, Dr. Fulvio Castelli. 10/2005‐ 08/2005 Auslandsaufenthalt in Proitzener Mühle – Uelzen – Deutschland Mühlenmitarbeiter 09/2003‐ 08/2003 Auslandsaufenthalt in Ballytoughey – Clare Island – Irland Farmmitarbeiter auf der „Macalla Farm“ 07/2003 Abitur I.T.I.S. Setificio “Paolo Carcano” – Technikum für Textilien – Como – Italien 08/2002 Auslandsaufenthalt an der Sprachschule „Regent School” Margate Pre‐intermediate – Kent – England Arbeitserfahrung im Rahmen der Chemie seit 10/2018 Wissenschaftlicher Mitarbeiter – CATALIGHT‐ CRC/Transregio Project and der Universität Ulm: Light-driven Molecular Catalysts in Hierarchically Structured Materials, Synthesis and Mechanistic Studies. 10/2018‐ 10/2012 Aktive Mitarbeit am MOCOMODELS – European Research Council Project “Synthesis of mono‐dithiolene molybdenum complexes and their evaluation as potential drugs for the treatment of human isolated sulphite oxidase deficiency.” Betreuerin: Prof. Dr. rer. nat. Carola Schulzke 07/2012‐ 03/2012 Wissenschaftlicher Mitarbeiter Synthese und Manipulation von luftempfindlichen Verbindungen bei inerter

Atmosphäre mittels Schlenktechnik Fakultät für Naturwissenschaften und Hochtechnologie – Como – Italien Laborleitung: Prof. Dr. G. Attilio Ardizzoia

Page 141: epub.ub.uni-greifswald.de · Table of contents 1. Pteridines and pterins

137

Wissenschaftliche Beiträge 13‐12/03/2018 14th Koordinationschemie‐Treffen (KTC) Heidelberg – Deutschland Posterpräsentation: “Synthesis of ligand System that mimic different features of Molybdopterin.” 23‐18/06/2018 10th Molybdenum and Tungsten Enzymes Conference (MoTEC) Santa Fe – New Mexico – USA Vortrag: “Molybdenum and Tungsten enzyme’s Active Site Models – Blueprints for mimicking Molybdopterin.” 07‐05/03/2017 13th Koordinationschemie‐Treffen (KTC) Potsdam – Deutschland Vortrag: “Mimicking the Molybdopterin System by Synthesis of Molybdenum-Pterin Complexes.” 24‐23/02/2017 26th Industrial Inorganic Chemistry‐Material and Processes (ATC) DECHEMA‐ House, Frankfurt am Main – Deutschland Posterpräsentation: “Synthesis of ligand System that mimic different features of Molybdopterin.” 01/09‐28/08/2016 13th European Biological Inorganic Chemistry Conference (EuroBIC) Budapest – Ungarn Posterpräsentation: “Mimicking the Molybdenum Cofactor by synthesis of Molybdenum-Pterin Complexes” 28‐23/08/2015 25th International Society of Heterocyclic Chemistry Congress Santa Barbara – California – USA Posterpräsentation: “Synthesis of ligand System that mimic different features of Molybdopterin.” 10‐06/09/2015 9th Molybdenum and Tungsten Enzymes Conference (MoTEC) Balaton – Ungarn Posterpräsentation: “Mimicking the Molybdopterin System by Synthesis of Molybdenum-Pterin Complexes.” 28‐24/08/2014 12th European Biological Inorganic Chemistry Conference (EuroBIC) Zürich – Schweiz Posterpräsentation: “Mimicking the Molybdopterin System by Synthesis of Molybdenum-Pterin Complexes.” 12‐11/09/2014 17th Norddeutsches Doktorandenkolloquium (NDDK) LIKAT Rostock – Deutschland Posterpräsentation: “Mimicking the Molybdopterin System by Synthesis of Molybdenum-Pterin Complexes.” 20‐19/09/2013 16th Norddeutsches Doktorandenkolloquium (NDDK) Bremen – Deutschland Posterpräsentation: “Synthesis of ligand System that mimic different features of Molybdopterin.” 20‐16/07/2013 8th Molybdenum and Tungsten Enzymes Conference (MoTEC) Sintra – Portugal Posterpräsentation: “Synthesis of ligand System that mimic different features of Molybdopterin.” Zusatzqualifikationen Wissenschaftliche Methoden Spektroskopie: NMR, GC‐MS, MS, IR, GC, UV / VIS Schlenktechnik, Chromatographie, Lösungsmittel Destillation (Wasser/Sauerstoff‐frei) Kurse 24/06/2016 „Intercultural Communication“ One‐day training program mit Zertifikat 10‐04/06/2015 Summer school on Spectroscopy

Page 142: epub.ub.uni-greifswald.de · Table of contents 1. Pteridines and pterins

138

“COST action CM1305 Spectroscopy of Spin in Catalysis, Bioinorganic and Materials Chemistry : spinning a web of theory and practice.” University of Groningen – Niederlande 17‐12/06/2016 Summer school on Organic Synthesis 41th Edition of the “A. Corbella” Gargnano – Italien 16/052017 Arbeitssicherheit und Gesundheitsschutz in Laboratorien Fortbildungsveranstaltung – Greifswald – Deutschland EDV Kenntnisse MS Office Excel, Word, PowerPoint Chemoffice ChemDraw Database Scifinder, Reaxys, Beilstein, Pubmed Wissenschaftliche Software EndNote, Mercury Sprachen Italienisch Muttersprache Deutsch B2 – fliesend Englisch C1 – fliesend, verhandlungssicher Veröffentlichungen

„Modelling the Molybdenum cofactor, synthesis of pterin-dithiolene ligand system .“

Ivan Trentin, Nicolas Chrysochos, Mohsen Ahmadi, Carola Schulzke. In Progress

im Rahmen des Projektes ERC‐ MOCOMODELS

https://cordis.europa.eu/result/rcn/179750_en.html

EU Research ‐ Journal

https://issuu.com/euresearcher/docs/eur10_digital_mag/14

„Crystal Structures of Bis[3-Methyl-1,3-Ene-Dithiol-2-One] Disulfide and Bis[3-Methyl-1,3-Ene-Dithiol-2-One] Diselenide“

Ivan Trentin, Claudia Schindler and Carola Schulzke, From Research Communications, Acta Crystallographica Section E, (2018) 74, 840‐845.

„Pd/PTABS: Catalyst for Room Temperature Amination of Heteroarenes“

Murthy Bandaru, Siva Sankar; Bhilare, Shatrughn; Chrysochos, Nicolas; Gayakhe, Vijay; Trentin, Ivan; Schulzke, Carola; Kapdi, Anant R.From Organic Letters (2018), 20(2), 473‐476.

„Engineering the Active Site of the Amine Transaminase from Vibrio fluvialis for the Asymmetric Synthesis of Aryl–Alkyl Amines and Amino Alcohols“

Nobili, Alberto; Steffen‐Munsberg, Fabian; Kohls, Hannes; Trentin, Ivan; Schulzke, Carola; Hoehne, Matthias; Bornscheuer, Uwe T.From ChemCatChem (2015), 7(5), 757‐760.

„The Goldilocks principle in action: Synthesis and structural characterization of a novel [125] cubane stabilized by monodentate ligands“

G. Attilio Ardizzoia, Stefano Brenna, Sara Durini, Bruno Therrien and Ivan Trentin, From Dalton Transactions (2013), 42(34), 12265‐12273.

Page 143: epub.ub.uni-greifswald.de · Table of contents 1. Pteridines and pterins

139

Acknowledgement

Firstly, I would like to express my sincere gratitude to my supervisor Prof. Dr. rer. nat. Carola Schulzke for

supporting me during my PhD study and related research, for her motivation, patience and immense

knowledge. Her guidance helped me in all though out my research and this thesis. She has been a great

example to me as both researcher and person.

I thank my colleagues for the stimulating discussions, for the camaraderie, and for all the fun we have had in

these years. In particular for the mutual support during the long working periods in the laboratory.

I could not have finished my research work without the help from technical and non technical staff from our

institute. Indeed I would like to thank Dr. Gottfried Palm for the measurement of my compound at the

Synchrotron.

Also I thank my friends in Como for the support gave me in these years and for visiting me in Greifswald,

despite the distance I never felt that I was away from my hometown.

I am grateful to Katharina and all the people I met in Greifswald for the help in all aspect of my life and for

making me feel at home.

Last but not the least, I would like to thank my family: my father Silvano, my mother Ornella, my sister Isabella

and Marco for supporting me throughout my studies.