Thesis AS 07 - ETH Z

244
Research Collection Doctoral Thesis Synthesis of chemical libraries based on a Bicyclo 3.1.0 hexane scaffold and studies on the synthesis of Bicyclo 3.1.0 hexane- based S-Adenosylhomocysteine and Pentostatin analogues Author(s): Schlegel, Anna Maria Publication Date: 2012 Permanent Link: https://doi.org/10.3929/ethz-a-7355965 Rights / License: In Copyright - Non-Commercial Use Permitted This page was generated automatically upon download from the ETH Zurich Research Collection . For more information please consult the Terms of use . ETH Library

Transcript of Thesis AS 07 - ETH Z

Research Collection

Doctoral Thesis

Synthesis of chemical libraries based on a Bicyclo 3.1.0 hexanescaffold and studies on the synthesis of Bicyclo 3.1.0 hexane-based S-Adenosylhomocysteine and Pentostatin analogues

Author(s): Schlegel, Anna Maria

Publication Date: 2012

Permanent Link: https://doi.org/10.3929/ethz-a-7355965

Rights / License: In Copyright - Non-Commercial Use Permitted

This page was generated automatically upon download from the ETH Zurich Research Collection. For moreinformation please consult the Terms of use.

ETH Library

Diss. ETH No.20158

Synthesis of Chemical Libraries based on a Bicyclo[3.1.0]hexane Scaffold and Studies on the Synthesis

of Bicyclo[3.1.0]hexane-based S-Adenosylhomocysteine and Pentostatin Analogues

A dissertation submitted to

ETH Zurich

for the degree of

Doctor of Sciences

presented by

Anna Maria Schlegel

Dipl. Natw. ETH Zurich

November 9, 1977

citizen of Buchs/SG, Switzerland

accepted on the recommendation of

Prof. Dr. Karl-Heinz Altmann, examiner Prof. Dr. Dario Neri, co-examiner

2011

Acknowledgments

In the first place I would like to thank Prof. Karl-Heinz Altmann for the opportunity to

accomplish my PhD thesis in his research group.

I am grateful to Prof. Dario Neri for being my co-examiner. Besides, I was allowed to use the

SpectraMaxmicroplate reader for protease activity tests in his lab, and I would also like to

thank all the people from his lab who showed me how to use it.

I would like to thank my collaborators: Dr. Doriano Fabbro and Dr. Daniel D’Orazio, Dr.

Thomas Keller, Prof. Stewart Cole and Dr. Ruben Hartkoorn, Prof. Jürg Getsch und Dr.

Andrea Chicca, Dr. Sebastian Sonntag and Dr. Paul Monaghan, Dr. Bernhard Pfeiffer, and

special thanks to Kurt Hauenstein and Tizian Herzog.

I would like to thank the whole Altmann group, especially Andrea Jantsch, Sascha Kopp,

Christian Kuzniewski, Raphael Schiess, Philipp Gersbach, Oliver Horlacher and Ana Simao,

as well as Sylvia Peleg. Heaps of gratitude go especially to Raphael (and lots of admiration).

In particular, I want to express my gratitude to my family and friends (especially Christoph

Schmid, Jeannine Lingeri, Nina Maag, Sofia Deloudi, Mike Günther, Besnik Kasumaj, and

Naomi Kirchgraber) for emotional and financial support.

Table of Contents

Abstract........................................... ...................................................................................... I

Zusammenfassung ................................... ........................................................................... V

Abbreviations, Acronyms and Symbols ............... ............................................................ IX

1 Introduction ...................................... .......................................................................... 1

1.1 Lead Finding in Drug Discovery .......................................................................... 1

1.2 Natural Products Containing the Bicyclo[3.1.0]hexane Scaffold .......................... 2

1.3 Conformational Characteristics of the Bicyclo[3.1.0]hexane Scaffold .................. 4

1.4 The Bicyclo[3.1.0]hexane Scaffold as a Rigid Sugar Analogue ........................... 8

1.4.1 Conformational Characteristics of the Sugar Part in Nucleosides............. 8

1.4.2 Discrimination between Sugar Conformations by Nucleoside-

processing Enzymes ..............................................................................11

1.4.2.1 HIV Reverse Transcriptase ....................................................................... 11

1.4.2.2 Other DNA Polymerases ........................................................................... 12

1.4.2.3 Adenosine Deaminase .............................................................................. 14

1.4.2.4 Cytidine Deaminase .................................................................................. 17

1.4.2.5 DNA Methyltransferases ........................................................................... 18

1.4.3 Receptors Discriminating between Sugar Pucker: Adenosine and

P2Y-Receptors .......................................................................................19

1.4.3.1 Adenosine Receptors ................................................................................ 19

1.4.3.2 P2Y Receptors .......................................................................................... 21

1.5 Synthesis of Bicyclo[3.1.0]hexane Systems .......................................................22

1.5.1 Natural Products .....................................................................................22

1.5.2 Sugar Analogues ....................................................................................25

1.5.3 Bicyclo[3.1.0]hexane-based Nucleoside Analogues ................................28

1.5.3.1 Northern (N)-Type Analogues ................................................................... 29

1.5.3.2 Southern (S)-Type Analogues ................................................................... 38

1.6 Conclusions .......................................................................................................42

2 Aims and Scope .................................... ....................................................................43

2.1 Chemical Library Synthesis ................................................................................43

2.2 Synthesis of Bicyclo[3.1.0]hexane-based Analogues of Pentostatin and S-

Adenosylhomocysteine ......................................................................................46

2.3 Alternative Synthesis of (+)-4 .............................................................................49

2.4 Biological Screening of the Bicyclo[3.1.0]hexane-based Chemical Library .........50

3 Results and Discussions ........................... ...............................................................51

3.1 Chemical Library Synthesis ................................................................................51

3.1.1 Synthesis of Advanced Intermediates 3, 4 and 5 ...................................51

3.1.2 Synthesis of Library Members with General Structure 1 ..........................58

3.1.2.1 Target Structures Incorporating Heterocyclic Amines ............................... 58

3.1.2.2 N-(2-((4-Methoxyphenyl)amino)ethyl)amides ............................................ 62

3.1.3 Synthesis of Library Members with General Structure 2 ..........................66

3.1.3.1 N-linked Heterocycles ............................................................................... 66

3.1.3.2 C-linked Heterocycles ............................................................................... 71

3.1.4 Final Library ............................................................................................75

3.2 Synthesis of Bicyclo[3.1.0]hexane-based 5-Alkynyl-deoxypyrimidine

Nucleoside Analogue 243 ..................................................................................76

3.3 Miscellaneous Derivatives ..................................................................................77

3.3.1 Tetrazole 250 ..........................................................................................77

3.3.2 Iodides 238 and 239 ...............................................................................78

3.3.3 C3-O-Alkyl Derivatives ............................................................................79

3.4 Structural Studies ...............................................................................................81

3.4.1 Conformational Analysis of 1aa and 1ag by Solution NMR and

Simulated Annealing (SA) ......................................................................81

3.4.2 Conformational Analysis by X-ray Crystallography ..................................85

3.5 Synthesis of Bicyclo[3.1.0]hexane-based Analogues of Pentostatin and S-

Adenosylhomocysteine ......................................................................................88

3.5.1 Bicyclo[3.1.0]hexane-based Pentostatin 6 ..............................................88

3.5.2 Bicyclo[3.1.0]hexane-based S-Adenosylhomocysteine 7 ........................92

3.6 Biological Evaluation of the Bicyclo[3.1.0]hexane-based Chemical Library ....... 100

3.6.1 Reporter Gene Assay Screening for Inhibitory Effects on Selected

Cellular Signaling Pathways ................................................................. 100

3.6.2 In Vitro Protease Inhibition .................................................................... 105

3.6.2.1 Thrombin Inhibition Assay ....................................................................... 105

3.6.2.2 Urokinase Inhibition Assay ...................................................................... 107

3.6.2.3 Cathepsin B Inhibition Assay ................................................................... 108

3.6.3 Antibiotic Activity against Mycobacteria................................................. 109

3.6.3.1 Compounds of General Structure 1 and 2 .............................................. 109

3.6.3.2 Antibiotic Activity against Mycobacterium Tuberculosis of 5-Alkynyl-

deoxypyrimidine Nucleoside Analogue 243 ............................................ 110

3.6.4 Cytotoxicity ........................................................................................... 111

3.7 Biological Evaluation of the Bicyclo[3.1.0]hexane-based S-

Adenosylhomocysteine Analogue ..................................................................... 114

3.7.1 Inhibition of AdoMet-dependant Methyltransferases by S-

Adenosylhomocysteine analogue 7 ...................................................... 114

4 Conclusions and Outlook ........................... ............................................................ 116

5 Experimental Section .............................. ................................................................ 118

5.1 General Methods .............................................................................................. 118

5.2 Experimental Procedures and Analytical Data .................................................. 120

5.2.1 Synthesis of Bicyclo[3.1.0]hexane-based Chemical Library .................. 120

5.2.1.1 Synthesis of Lactone 161 ........................................................................ 120

5.2.1.2 Synthesis of Advanced Intermediates 3, 4, and 5 ................................... 120

5.2.1.3 Synthesis of library members of general structure 1 ............................... 128

5.2.1.4 Synthesis of library members of general structure 2 ............................... 161

5.2.1.5 Crystallographic Data .............................................................................. 174

5.2.2 Biological Evaluation of Bicyclo[3.1.0]hexane-based Chemical

Library .................................................................................................. 186

5.2.3 Synthesis of Miscellaneous Derivatives and Bicyclo[3.1.0]hexane-

based 5-Alkynyl-deoxypyrimidine Nucleoside Analogue 243 ................ 187

5.2.4 Synthesis of Bicyclo[3.1.0]hexane based Pentostatin- and (S)-

Adenosylhomocysteine Analogues ....................................................... 194

5.2.4.1 Preparation of lactone (-)-44 (enantiomeric series)................................. 194

5.2.4.2 Preparation of Advanced Intermediate (+)-4 ........................................... 197

5.2.4.3 Attempted Preparation of Pentostatin Analogue 6 .................................. 202

5.2.4.4 Preparation of S-Adenosylhomocysteine Analogue 7 ............................. 205

6 Curriculum Vitae................................... ................................................................... 218

7 Bibliography ...................................... ...................................................................... 220

I

Abstract

The bicyclo[3.1.0]hexane scaffold possesses unique conformational characteristics and as

such constitutes an attractive starting point for the exploration of new types of lead structures

for drug discovery. In contrast to a simple cyclohexane ring, the bicyclo[3.1.0]hexane system

(Fig. 1) exhibits a pronounced preference for a boat-type conformation, which is the result of

the staggered arrangement of substituents/hydrogens attached to C1, C2 and C5, C4 as well

as favorable orbital interactions. Collectively, these effects outplay the adverse 1,4-

interactions of substituents/hydrogens attached to C6 and C3.

Figure 1. The boat-type conformation of bicyclo[3.1.0]hexane.

The bicyclo[3.1.0]hexane structural unit has been used as a (deoxy)ribose surrogate in

different nucleosides and nucleotides, in order to probe the conformational preferences of

various nucleoside- and nucleotide-processing enzymes, such as HIV-RT, adenosine

deaminase or DNA methyltransferases, and GPCR’s such as the adenosine or P2Yx

receptors.

One goal of this research project was to extend the exploitation of the bicyclo[3.1.0]hexane

scaffold’s special conformational properties to structures other than nucleosides and

biological targets other then nucleoside/nuceotide-interacting enzymes or receptors. To this

end, a small screening library of bicyclo[3.1.0]hexane-based molecules of the general

structures 1 and 2 was to be synthesized (Fig. 2).

Figure 2. Bicyclo[3.1.0]hexane-based library members.

II

The substituents attached to the bicyclo[3.1.0]hexane scaffold in 1 and 2 comprise various

pharmacophoric groups that were chosen based on their prevalence of occurrence in known

drugs; the heterocycles directly attached to the bridgehead carbon atom in 2 frequently occur

in biologically active nucleoside analogues or natural products. Due to the exceptional

conformational rigidity of the bicyclo[3.1.0]hexane scaffold, these groups are present in well

defined spatial orientations.

The synthesis of target structures 1 and 2 was based on racemic bicyclic lactone 161, which

could be prepared in multigram quantities starting from tetrahydrophthalic acid anhydride.

161 was elaborated into the protected bicyclo[3.1.0]hexane-based ester 185 in a highly

efficient 3-step sequence, which included the base-induced cyclization of bromo ester 184 as

the key step (Scheme 1).

Scheme 1. Preparation of key intermediate 185.

Ester 185 was then transformed into acid 3 and amine 4 with excellent efficiency. Acid 3

could be successfully elaborated into a library of bicyclo[3.1.0]hexane derivatives of type 1 by

amide formation, azide reduction to the amine, a second amide-forming step, and global

deprotection. Thus, 92 different compound of type 1 were obtained in quantities of > 1 mg

(mostly > 3 mg).

In contrast, the projected conversion of 3 and 4 into analogs of type 2 met with siginificant

diffculties and only few compounds of this type could be accessed. However, a number of 3-

benzyloxy derivatives with Het = uracil could be prepared successfully; these analogs were

obtained via intermediate 5.

III

Structual studies by x-ray crystallography and solution NMR spectroscopy confirmed the

purported boat-like conformation of the bicylic ring system in these derivatives.

The biological evaluation of the library was performed in three protease inhibition assays (for

thrombin, urokinase, and cathepsin B) and a number of reporter gene assays that reflect the

activity of different cellular signalling pathways and networks. While no activity was

detectable in any of the protease inhibition assays, 4 compounds were found to display sub-

µM inhibitory activity in reporter gene assays that measure AP-1-mediated transcription in

response to activation of the EGF or TNFα receptor, respectively.

The methodology developed for the synthesis of the above chemical library was also to be

exploited for the synthesis of deoxyuridine analog 243 (as a putative anti-mycobacterial

agent), pentostatin analog 6, and S-adenosylhomocysteine (SAH) analog 7 (Fig. 3).

N

N

N NHHO

HO

6

HO

S N

N

NN

NH2

H2N

7

OH HOOC

HO

HON

NHO

O

243

(CH2)9CH3

Figure 3. Bicyclo[3.1.0]hexane-based deoxyuridine-, pentostatin- (6) and S-adenosylhomocysteine

analogues (7).

The synthesis of racemic 243 was successfully accomplished from amine 4, but the

compound was found to be inactive against M. tuberculosis.

Analogs 6 and 7 were to be prepared as single enantiomers via the dextrorotatory

enantiomer of 4 (i. e. (+)-4). The latter was obtained through fractionate crystallization of

racemic acid (±)-175 with (-)-ephedrine; (±)-175 is an early precursor in the synthesis of

racemic 4.

IV

(±)-175

COOMe

COOH

TBSO

TBSON

NOTBS

NH2

163

N3

Amine (+)-4 could transformed into the imidazole derivative 163 in low yield; however, all

attempts at the elaboration of this intermediate into target structure 6 failed. The synthesis of

bicyclo[3.1.0]hexane-based SAH analogue 7 was successfully accomplished via

intermediates 168 and 287 (Scheme 2).

Scheme 2. Synthesis of SAH analogue 7.

While NMR-spectroscopic analysis of 7 clearly suggested the presence of a single isomer,

HPLC analysis of the material gave two distinct peaks in a 1:1 ratio. Preparative separation

of these peaks and re-analysis of the pure fractions again produced an analytical

chromatogram with two peaks. This finding indicates that 7 (under the conditions of the

HPLC experiments) exists as an equilibrium mixture of two species; the nature of this

equilibrium was not elucidated.

SAH analogue 7 did not show any inhibition of Dengue methyltransferase.

V

Zusammenfassung

Das Bicyclo[3.1.0]hexan-Gerüst besitzt einzigartige konformationelle Eigenschaften und stellt

als solches einen attraktiven Ansatzpunkt für die Erschliessung neuer Leitstrukturen für die

Wirkstoffentwicklung dar. Im Gegensatz zu einem einfachen Cyclohexanring besitzt das

Bicyclo[3.1.0]hexan-System eine ausgeprägte Präferenz für eine boot-ähnliche

Konformation, welche aus der gestaffelten Anordnung von Substituenten/H-Atomen an C1,

C2 und C5, C4, sowie vorteilhaften Orbitalwechselwirkungen resultiert. Diese Effekte

zusammen machen die ungünstige 1,4-Wechselwirkung von Substituenten/H-Atomen an C6

und C3 wett.

Abbildung 1. Die boot-ähnliche Konformation von Bicyclo[3.1.0]hexan.

Die strukturelle Einheit des Bicyclo[3.1.0]hexans wurde bisher als (Deoxy)ribose-Surrogat in

verschiedenen Nucleosiden and Nucleotiden verwendet, um die konformationellen

Präferenzen verschiedener Nukleosid- und Nukleotid-prozessierender Enzyme, wie HIV-RT,

Adenosin Deaminase oder DNA-Methyltransferasen, und GPCRs wie die Adenosin- oder

P2Yx -Rezeptoren, zu untersuchen.

Ein Ziel dieses Forschungsprojekts war es, die Nutzung der speziellen konformationellen

Eigenschaften des Bicyclo[3.1.0]hexan-Gerüsts auf andere Strukturen ausser Nukleosiden

und andere biologische Angriffsziele ausser Nucleosid-/Nucleotid-interagierende Enzyme

oder Rezeptoren auszuweiten. Zu diesem Zweck wurde eine kleine Screening-Bibliothek

Bicyclo[3.1.0]hexan-basierter Moleküle mit den allgemeinen Strukturen 1 und 2 synthetisiert

(Abbildung 2).

Abbildung 2. Bicyclo[3.1.0]hexan-basierte Bibliothekssubstanzen.

VI

Die an das Bicyclo[3.1.0]hexan-Gerüst gebundenen Substituenten der Zielstrukturen 1 und 2

umfassen verschiedenste pharmakophore Gruppen, die nach der Häufigkeit ihres

Vorkommens in bekannten Wirkstoffen ausgewählt wurden; die direkt an das

Brückenkopfatom gebundenen Heterozyklen in 2 kommen häufig in biologisch aktiven

Nukleosid-Analoga oder Naturprodukten vor. Dank der aussergewöhnlichen

konformationellen Starrheit des Bicyclo[3.1.0]hexan-Gerüsts liegen diese Gruppen in einer

definierten räumlichen Anordnung vor.

Die Synthese der Zielstrukturen 1 und 2 basierte auf dem racemischen bicyclischen Lacton

161, das von Tetrahydrophthalsäure ausgehend in mehreren Gramm hergestellt wurde. 161

wurde in einer hocheffizienten 3-Schritt-Sequenz in den geschützten bicyclo[3.1.0]hexan-

basierten Ester 185 überführt, mit der basisch induzierten Cyclisierung des Bromoesters 184

als Schlüsselschritt (Schema 1).

Schema 1. Synthese des Intermediats 185.

Der Ester 185 wurde dann mit exzellenter Effizienz in die Säure 3 und das Amin 4 überführt.

Die Säure 3 konnte durch eine Amidkopplung, eine Azidreduktion zum Amin, eine zweite

Amidkopplung, und globale Entschützung erfolgreich in eine Bibliothek von

Bicyclo[3.1.0]hexan-Derivaten vom Typ 1 überführt werden. Auf diese Weise wurden 92

verschiedene Substanzen vom Typ 1 in Mengen von > 1 mg (> 3 mg für den Grossteil)

erhalten.

VII

Im Gegensatz dazu traten bei der geplanten Konversion von 3 und 4 in Analoga vom Typ 2

signifikante Schwierigkeiten auf und nur wenige Substanzen dieses Typs konnten hergestellt

werden. Indes konnten einige 3-Benzyloxy-Derivate mit Het = Uracil erfolgreich synthetisiert

werden; diese Analoga wurden via Intermediat 5 erhalten.

Strukturelle Studien, die Kristallstrukturanalyse und NMR-Spektroskopie in Lösung

beinhalteten, bestätigten die angebliche boot-ähnliche Konformation des bicyclischen

Ringssystems in diesen Derivaten.

Die biologische Evaluation der Bibliothek wurde in Form von drei Protease-Inhibitions-

Assays (für Thrombin, Urokinase, und Cathepsin B) und einer Reihe von Reporter Gene

Assays, die die Aktivität verschiedener Zellsignalwege und Netzwerke reflektieren,

durchgeführt. Während keine Aktivität in einem der Protease-Inhibitions-Assays festgestellt

werden konnte, wurden in den Reporter Gene Assays, die die AP-1-vermittelte Transkription

als Antwort auf die Aktivierung des EGF- or TNFα−Rezeptors gemessen haben, 4

Substanzen mit inhibierender Aktivität im sub-µM Bereich identifiziert.

Die für die Synthese der chemischen Bibliothek entwickelte Methodik wurde auch für die

Synthese des Deoxyuridin Analogs 243 (als putativer anti-mycobakterieller Wirkstoff), des

Pentostatin Analogs 6, und des S-Adenosylhomocystein (SAH) Analogs 7 verwendet (Abb.

3).

N

N

N NHHO

HO

6

HO

S N

N

NN

NH2

H2N

7

OH HOOC

HO

HON

NHO

O

243

(CH2)9CH3

Abbildung 3. Bicyclo[3.1.0]hexan-basierte Deoxyuridin-(243) , Pentostatin- (6) and S-Adenosyl-

homocystein Analoga (7).

VIII

Die Synthese von racemischem 243 wurde von 4 ausgehend erfolgreich durchgeführt, aber

die Substanz war inaktiv gegen M. tuberculosis.

Analoga 6 und 7 sollten enantiomerenrein über das dextrorotatorische Enantiomer von 4 (d.

h. (+)-4) hergestellt werden. Letzteres wurde durch eine fraktionierte Kristallisation der

racemischen Säure (±)-175 mit (-)-Ephedrin hergestellt; (±)-175 ist ein früher Vorläufer in der

Synthese von racemischem 4.

(±)-175

COOMe

COOH

TBSO

TBSON

NOTBS

NH2

163

N3

Amin (+)-4 konnte in niedrigen Ausbeuten in das Imidazol-Derivat 163 überführt werden; alle

Versuche, dieses Intermediat zur Endverbindung 6 umzusetzen schlugen hingegen fehl. Die

Synthese des Bicyclo[3.1.0]hexane-basierten SAH Analogs 7 wurde erfolgreich via die

Intermediate 168 and 287 ausgeführt (Schema 2).

Schema 2. Synthese des SAH Analogs 7.

Während die Analyse mittels NMR-Spektroskopie klar auf das Vorliegen eines einzelnen

Isomers hinwies, ergab die HPLC Analyse zwei individuelle Peaks mit einem Verhältnis von

1:1. Präparative Trennung dieser Peaks und erneute Analyse der reinen Fraktionen

produzierte erneut ein analytisches Chromatogramm mit zwei Peaks. Diese Ergebnis deutet

darauf hin, dass 7 (unter den Bedingungen des HPLC-Experiments) in einer

Gleichgewichtsmischung zweier Spezies vorliegt; die Natur dieses Gleichgewichts wurde

nicht aufgeklärt.

SAH Analog 7 zeigte keine Inhibition der Dengue Methyltransferase.

IX

Abbreviations, Acronyms and Symbols

))) microwave irradiation

A

AcOH acetic acid

AgNO3 silver nitrate

AP 1 activating protein 1

ARE antoxidant responsive element

B

BH3·Me2S borane dimethylsulfide complex

Bu2NH dibutylamine

BnBr benzylbromide

BnOH benzyl alcohol

C

cAMP cyclic adenosine monophopsphate

CAN cerium ammonium nitrate

cat. catalyst

CMV cytomegalovirus

CRE cAMP response element

CSA camphor sulfonic acid

D

δ delta; chemical shift in ppm (NMR)

d doublet (NMR)

DEAD diethyl azodicarboxylate

DIPEA diisopropylethylamine

DMF dimethylformamide

DPPA diphenylphosphoryl azide

E

ee enantiomeric excess

EGF epidermal growth factor

EDAC 1-Ethyl-3-(3-dimethylaminopropyl)carbodiimid

ESI electron spay ionisation

Et2O diethylether

X

Et3N triethylamine

EtOH ethanol

F

FADD Fas-Associated protein with Death Domain

FRET fluorescence resonance energy transfer

H

h hours

HATU 2-(1H-7-azabenzotriazol-1-yl)--1,1,3,3-tetramethyl-

uronium hexafluorophosphate methanaminium

HOBt hydroxybenzotriazole

HPLC high performance liquid chromatography

HR-MS high resolution mass spectrometry

I

i-PrOH isopropanol

Im2CO carbonyldiimidazole

Im imidazole

M

Me3BH trimethylborane

min minutes

MsCl mesyl chloride

MTBSA

N

NFκB nuclear factor κB

n-BuLi butyl lithium

P

PCC pyridinium chlorochromate

Pd/C 10% palladium on carbon

Pd/CaCO3 5% palladium on calcium carbonate (Lindlar cat.)

Pd(PPh3)4 tetrakis-triphenyl palladium

PhCOOH benzoic acid

PPh3 triphenylphosphine

PPh2N3 diphenylphosphorylazide

XI

py pyridine

(PhO)2PON3 diphenylphosphorylazide

R

rt room temperature

S

SBE SMAD binding element

T

TBAI tetrabutylammonium iodide

TBAF tetrabutylammonium fluoride

TBS tert-butyldimethylsilyl

TBDMSTf tert-butyldimethylsilyl triflate

tert-BuOK potassium tert-butoxide

tert-BuOH tert-butanol

TGFβ transforming growth factor

THF tetrahydrofuran

TIPDSCl2 1,3-dichloro-1,1,3,3-tetraisopropyldisiloxane

TMSBr trimethylsilylbromide

TMSN3 trimethylsilyl azide

TNFα tumor necrosis factor alpha

TosMIC tosylmethyl isocyanide

TRADD Tumor necrosis factor receptor type 1-associated

DEATH domain protein

TsN3 p-toluenesulfonylazide

U

UV ultraviolet

Introduction

1

1 Introduction

1.1 Lead Finding in Drug Discovery

Lead finding marks a decisive phase in the drug discovery process and requires the co-

operation of several disciplines, in order to identify a structural starting point for an extensive

chemical optimization effort that is directed at the discovery of a candidate drug. The aim is

to find those substances in the vast chemical space of all possible compounds that possess

a biological profile which could be potentially useful for medicinal purposes. The disciplines

involved in lead finding include synthetic and computational chemistry, biology,

pharmacology and biopharmacy.

There are different approaches currently employed in the drug discovery process for lead

finding. One important starting point is the high-throughput screening1 (HTS) of compound

collections, in the meaning of a systematic assessment of compound effects in biochemical,

cellular, or biophysical assays. The compound collections screened can be obtained by

means of combinatorial chemistry/parallel synthesis, they can be bioextracts from plants or

microbes, or pure natural products. However, the high-throughput screening in the first place

only generates hits, i.e. non-reactive compounds of verified structure and purity with a

confirmed minimal in vitro potency. These hits have to be transformed into leads in the next

phase of the drug discovery process.2 The structure and purity of compounds reaching the

minimum biological activity threshold set for a given screen have to be confirmed; likewise,

the activity, the physico-chemical and ADME (absorption, distribution, metabolism and

excretion) properties are assessed and the possibilities for further chemical derivatization

have to be investigated. Extensive hit-to-lead chemistry can then identify the core structure

required for a specific activity of interest (inhibition of a certain target).

An alternative screening approach is fragment-based screening, but again this method does

not directly generate lead compounds.3 With this method, small (MW < 300) fragments that

possess low biological activity are screened and subsequently developed into higher potency

lead structures by combining them.

Existing drugs or clinical development compounds can be used as lead structures by

extending their targets or by their further optimization for a chosen target. Natural substrates

or known receptor ligands can serve as lead structures. In general, the exploitation of

biological information won by biochemical or cellular assays, in animal experiments, by the

detection of side effects in human clinical trials or by basic biological research can be used to

restrict oneself to certain chemical structures.

Introduction

2

There are also in silico approaches such as virtual screening4 or structure-based rational

drug design.5

High-throughput screening is the presently the most widely applicable technology to deliver

chemical starting points for drug discovery. This method enables the screening of up to 100

000 compounds a day, but it has not led to the expected abundance of suitable lead

compounds; of 58 drugs introduced in the market between 1991 and 2008 only 19 were

HTS-derived.6 Although the compounds in large libraries are by now successfully subjected

to prediction tools for physicochemical properties7 ensuring that hits with drug-like properties

are generated, the issue of covering a significant portion of chemical diversity space is not

addressed satisfactorily.1

Instead of screening huge compound libraries, the directed synthesis of smaller focused

libraries which are subsequently assessed biologically is therefore also considered an

important means of lead finding.8 These libraries are designed around a specific core

structure or a certain scaffold.

However, it is difficult to choose the scaffold that is to serve as a common structural feature

of a small chemical library. In this context, the bicyclo[3.1.0]hexane system represents a

promising scaffold that has not yet been explored widely outside the nucleoside analogue

research area. With its special conformational characteristics, it embodies an excellent

starting point for the exploration of new types of structures, opening up the possibility to

predefine the spatial orientation of pharmacophoric groups attached to it.

1.2 Natural Products Containing the Bicyclo[3.1.0]h exane Scaffold

The bicyclo[3.1.0]hexane structural unit can be found in a number of natural products;

however, overall, its occurrence in nature is relatively rare. Two prominent examples of

bicyclo[3.1.0]hexane-containing natural products are (-)-α- and (+)-β-thujone (8a and 8b,

respectively), monoterpenes that have been isolated from Artemisia absinthium or several

other plants and that act on GABA and 5HT3 receptors in the brain (Fig. 1).9

Introduction

3

Figure 1. Natural products containing the bicyclo[3.1.0]hexane scaffold.

Chlorajaponilide A (11b), very recently isolated from Chloranthus japonicus, is a

sesquiterpenoid that contains two bicyclo[3.1.0]hexane units and possesses anti-HIV-1

activity.10 From the same plant, which mainly grows in East Asia, other sesquiterpenoids with

biological activities have been isolated, such as the chlorahololides (11c, 11d);11 all of these

compounds share a lindenane scaffold as the common core structure. Chloranthalactone A

(11a) is possibly a biosynthetic precursor of 11b-d.12 Syntheses of the heptacyclic cores of

chlorahololides (11c and 11d) have been recently accomplished.13

Laurinterol (9) has been isolated from seaweeds of the genus Laurencia and constitutes an

unusual natural product with its bicyclo[3.1.0]hexane unit and its bromo functionality.14 From

Jatropha curcas L., a plant widely distributed in Asian and African countries, Jatropha factor

C1 (10a) and the related compounds 10b and 10c have been isolated.15 Compounds 10a-c

are probably biosynthetically derived from the diterpene 12-deoxy-16-hydroxyphorbol; it is

unknown, however, which reactions lead to the formation of the bicyclo[3.1.0]hexane

structural unit in these compounds.16

Introduction

4

1.3 Conformational Characteristics of the Bicyclo[3 .1.0]hexane

Scaffold

The most specific conformational feature of the bicyclo[3.1.0]hexane moiety is its exclusive

occurrence as a boat conformer. As a consequence, the cyclopentane ring embedded in the

bicyclo[3.1.0]hexane scaffold is permanently locked in a single envelope conformation; in

contrast, cyclopentane itself, while also residing in an envelope conformation, normally

undergoes a rapid pseudorotation process during which its five carbon atoms take turns in

adopting the endo position, interconverting with half-chair forms that are of equally lower

energy compared to a planar conformation.17 It has to be noted that, for simplicity, the typical

conformation of a bicyclo[3.1.0]hexane system is referred to here as “boat” conformation,

although it is more planar than the corresponding form of cyclohexane (and, hence, is also

called pseudoboat conformation). The conformational preference of the bicyclo[3.1.0]hexane

system for a boat conformation was first established experimentally by microwave18 and

combined electron-diffraction microwave studies19 of bicyclo[3.1.0]hexane (12) itself as well

as X-ray crystallographic analysis of bicyclo[3.1.0]hexane-based p-bromosulfonamide 13

(Fig. 2).20

Figure 2. Bicyclo[3.1.0]hexane and derivatives used in molecular geometry studies.

Introduction

5

Additionally, NMR studies21,22,23 of bicyclo[3.1.0]hexane derivatives such as 8b/c , 14a/b, and

15a/b confirmed this conformational preference also in solution. These experimental findings

are supported by ab initio calculations on 12 using the 4-31G or the 4-21G basis set.24

This preference of the bicyclo[3.1.0]hexane system for a boat conformation can be partly

explained by the comparison of the boat and chair conformations of bicyclo[3.1.0]hexane

with those of cyclohexane (Fig. 3). In the latter, the boat conformation is greatly disfavoured

due to an adverse 1,4 interaction and, even more importantly, the boat conformation would

result in an unfavourable eclipsed arrangement of the hydrogens or substituents attached to

C1, C2, C4 and C5 (b’ ), whereas in the chair conformation the arrangement is staggered and

1,4 interactions are non-existent (a’).

Figure 3. Comparison of boat and chair conformations of cyclohexane and bicyclo[3.1.0]hexane.

Introduction

6

In contrast, the Newman projections of bicyclo[3.1.0]hexane reveal an adverse eclipsed

arrangement of hydrogen atoms or substituents in the case of the chair conformer (a). In the

case of the boat conformer, the corresponding hydrogen atoms/substituents arrange

staggered and gauche, respectively, and this favorable arrangement outplays the 1,4

interaction between the C3 and C6 hydrogens or substituents, respectively. The argument of

replacing two 1,3 interactions by a single 1,4 interaction has also been suggested to account

for a favourable boat conformation.23 In contrast, a combined electron-diffraction microwave

study19 of 12 additionally suggested an unusually short C1-C5 bond, which would render the

1,4 interaction in the boat bicyclo[3.1.0]hexane conformer more severe; however, this finding

is neither supported by the ab inito results, nor by later studies that combine experimental

data with quantum mechanical calculations, such as MM2 and MM3 force field studies, like

the one carried out in 1992 by Allinger and co-workers on 12.25

The lengths of the C1-C2, C2-C3 and the C1-C6 bonds in bicyclo[3.1.0]hexane (12) have

been determined by Kato and co-workers by ab initio calculations using the 3.21G, 3-21G*,

and 6-31G* basis sets to be all around 1.52 Å.26 A bond length in the same range has been

obtained for the C1-C5 bond.26 The torsion angle between the planes spanned by C4, C5,

C1, C2 and C4, C3, C2 was determined to be 150° ( β), the one between planes spanned by

C4, C5, C1, C2 and C5, C6, C1 to be 110° ( α). The bond angle between the bonds C1- C2

and C2- C3 was found to be 105° ( γ, Fig. 4). These results were in good agreement with

observed values18 and with earlier calculations as well as with the molecular mechanics

study.25

Figure 4. Bond lengths, torsion angles (α, β) and bond angle (γ) in bicyclo[3.1.0]hexane.

Most importantly, Kato’s work showed that stereoelectronic effects greatly add to the purely

steric analysis, such that a stabilizing orbital interaction exists between the lowest

unoccupied molecular orbital (LUMO) of the cyclopropane ring, a Walsh-type orbital, and the

back lobes of the pseudoequatorial C-H2 and C-H4 highest occupied molecular orbitals

(HOMOs). As can be seen in Figure 5, the stabilizing interaction can only take place in the

boat conformation, where the cyclopropane LUMO is in line with the C-H2 and C-H4

pseudoequatorial bonds, which is not the case for the chair conformer.26

Introduction

7

boatchair

H4

H4

H5

H2

H2

H1

1 23

45

6

Bicyclo[3.1.0]hexane

in-phase orbital interaction

of cyclopropane LUMO with

C-H4 and C-H2 HOMOs

H4

H4H5

no stabilizing orbital interaction

H2

H2H1

1 2

345

6

ba

Figure 5. Orbital interactions in the boat (b) and chair (a) conformation of bicyclo[3.1.0]hexane.26 In

case b), there is an in-phase orbital interaction existing which is geometrically not possible in case a).

All the above steric and stereoelectronic arguments in favour of the boat conformer should

also hold for substituted bicyclo[3.1.0]hexane derivatives. As indicated above, several X-ray

crystal structures of bicyclo[3.1.0]hexane derivatives, e.g. 16, 17, 18 27 indeed prove this

point.30, 28 No other conformations for the bicyclo[3.1.0]hexane scaffold have been found in

existing compounds (based on a Cambridge Structural Database search).

Accordingly, the rigid conformation of the bicyclo[3.1.0]hexane scaffold leads to a significant

degree of structural preorganization of substituents attached to this scaffold, and this, in turn,

enables the defined spatial arrangement of pharmacophores, a feature that was to be

exploited in this thesis. So far, exploitation of the bicyclo[3.1.0]hexane scaffold’s unique

conformational properties has been largely limited to its use as a rigid sugar analogue. The

bicyclo[3.1.0]hexane scaffold’s use as a means to impart rigidity on nucleoside analogues is

shortly discussed in the next chapter.

Introduction

8

1.4 The Bicyclo[3.1.0]hexane Scaffold as a Rigid Su gar Analogue

1.4.1 Conformational Characteristics of the Sugar P art in Nucleosides

Nucleosides are N-glycosides consisting of a nucleobase (adenine, guanine, cytosine, uracil

or thymine) bound to a 5-or 6-carbon sugar via a glycosidic linkage. Most common examples

of nucleosides include cytidine, uridine, adenosine, guanosine, thymidine and inosine,

consisting of either a ribose or a deoxyribose and a base moiety (Fig. 6).

Figure 6. Common nucleosides and nucleotides.

When an enzyme or receptor interacts with its nucleoside substrate, the efficiency of the

enzymatic reaction depends on all parts of the nucleoside (ribose and base) being properly

arranged to fit the active site. The critical conformational parameter of the ribose (or the 2’-

deoxyribose) moiety that ensures that these interactions are optimal is the sugar pucker,

which is defined by the pseudorotational phase angle P.29 P can adopt values between 0° and

360° and is a function of the five endocyclic torsi on angles of the tetrahydrofuran ring (Fig. 7).

A range of P between 342° and 18° ( 2E -->3T2-->3E) by convention corresponds to a northern

conformation of the sugar in nucleosides/nucleotides. A southern conformation is defined by

a range of P between 162° and 198° ( 2E -->2T3-->3E). The radius of the pseudorotational

cycle is equal to the maximum out-of-plane pucker. In principle, the whole variety of

conformations designated in the pseudorotational cycle can be adopted by nucleosides. The

preference for any of these specific conformations in solution is largely determined by the

interplay of gauche and anomeric effects. The anomeric effect30 is characterized by the

orbital interaction between the lone pair of oxygen O4’ and the highest p-orbital component

of the anti-bonding orbital of the C1’-N1/N9 glycosyl bond, and it is strongest when the

conformation of the nucleoside is northern. Gauche effects existing between O4’-C4’-C3’-O3’

and O4’-C1’-C2’-O2’ fragments exert a stronger influence than the anomeric effect, steering

the conformation for 2’-deoxyribonucleosides towards south, whereas for ribonucleosides,

Introduction

9

these effects result in a two-state 3’-endo 2’-endo equilibrium of an approximate 1:1

ratio.31

Rapid interconversion between these two extreme forms of sugar pucker via the

pseudorotational cycle takes place constantly, as the energetic barrier is very low (in the

range of 4 kcal/mol).32

E0

North

South

2E

1E

3E

0E90°270°

180°

3T2

2T3

1T2

1T0

OBHOH2CBHOH2C

O

O

BHOH2C

O

BHOH2C

3E

2E

3'-endo

2'-endo

r = max

Figure 7. The pseudorotational cycle.

Although the energy difference between these two conformations is small, experimental

findings lead to the conclusion that the majority of nucleoside/nucleotide-interacting enzymes

accept the furanose ring only in one of the two conformations.32 These enzymes seem to

have strict conformational preferences for substrate binding, as in many cases only one of

the two conformers is present in the active site of the enzyme in structures of enzyme-

substrate complexes.33 Thus, methods have been searched for to elucidate those

conformational preferences for different enzymes.

Experimental methods to achieve that goal include a rigidification of the nucleoside in one of

the two conformations, southern or northern. In this context, the bicyclo[3.1.0]hexane scaffold

offers the opportunity of a fixation of either of the two main conformations of a nucleoside by

substituting the furanose ring part of the nucleoside with a bicyclo[3.1.0]hexane moiety (Fig.

8). Depending on the location of the fused cyclopropane ring supplanting the ribofuranoside

oxygen, either a northern or a southern nucleoside conformation can be achieved, in which

Introduction

10

the conformation is then locked. Thus, an analogue corresponding to a northern (C3’-endo)

conformation is obtained when the cyclopropane ring is positioned between C4’ and the

carbon replacing the oxygen, keeping a nucleoside-like substitution elsewhere.

Figure 8. Bicyclo[3.1.0]hexane based nucleoside analogues. Adapted from 34.

Conversely, fusion of the cyclopropane ring between carbon C1’ and the carbon supplanting

the ribofuranoside oxygen provides a locked southern or C2’-endo conformation. In fact,

these conformations are only 18° away from the idea l northern or southern conformations35

that conventional nucleosides adopt (Fig. 9).

Figure 9. Position of bicyclo[3.1.0]hexane-based nucleoside analogues in the pseudorotational cycle.

Adapted from 34.

Introduction

11

Furthermore, the replacement of the furanose oxygen by carbon also provides higher

chemical and enzymatic stability of the resulting analogues, due to the absence of a

glycosidic bond. While this is also true for simple carbocyclic nucleosides where the furanose

ring is replaced by a cyclopentane unit, these latter analogues adopt an aberrant 1‘-exo

conformation, due to the steric bulk of the nucleobase, which prefers to adopt an equatorial

orientation. Consequently, such analogues are not suitable to probe conformational

preferences of nucleoside- or nucleotide-targeting, as the orientation of the hydroxyl groups

and/or base is not similar to the original nucleoside/nucleotide conformation.

1.4.2 Discrimination between Sugar Conformations by Nucleoside-

processing Enzymes

1.4.2.1 HIV Reverse Transcriptase

Dideoxynucleoside analogues targeting viral reverse transcriptase (RT), a RNA-dependant

DNA polymerase, were the first drugs enabling an effective treatment of HIV-1 infections.36

After phosphorylation by viral and host kinases, these compounds are incorporated into the

viral DNA and subsequently act as chain terminators, thereby stopping viral transcription.

The nucleoside reverse transcriptase inhibitors (NRTI’s) such as AZT or stavudine (Fig. 10)

achieve a certain selectivity, since DNA production by reverse transcriptase is very different

from human DNA replication.37

Figure 10. Selected nucleoside analogue HIV RT inhibitors (NRTI’s) and carbocyclic analogues.

Introduction

12

For HIV-1 RT, there exists evidence that the conformation of the sugar moiety preferred by

the reverse transcriptase is similar to the northern conformation.38 When the 5’-triphosphate

of AZT (19cb) was compared with its conformationally restricted analogues 19aa and 19ba

(Fig. 10), in terms of anti-RT activity, it was found that the southern analogue 19ba was

completely inactive against HIV-1 RT, whereas the northern triphosphorylated analogue

19aa inhibited RT as effectively as the (flexible) AZT nucleotide.33a However, 19a as such

shows no anti-HIV effect, meaning that it is not recognized by cellular kinases. In contrast,

the northern-type analogue of the NRTI stavudine (20b) shows anti-HIV activity; the double

bond in the carbocyclic scaffold of 20a changes the conformation of the five-membered ring

from envelope to planar, a change that made the molecule recognizable to cellular kinases.39

These findings suggest that a southern conformation is advantageous for the interaction with

cellular kinases, whereas RT interacts preferentially with the northern conformers. AZT itself,

which is flexible, has a preference for the southern conformation, due to a gauche effect

between the 3’-azido group and the ribose O4’-oxygen. This preference is reflected in the

fact that phosphorylation of AZT by thymidine kinase (tk1) is 500-fold more efficient than the

phosphorylation of stavudine or northern analogues.40 However, once triphosporylated the

compound would bind to RT in a northern conformation.

1.4.2.2 Other DNA Polymerases

Northern-methanocarbathymidine (N-MCT, 21a, Fig. 11) is another conformationally

restricted thymidine analogue41 with high activity against Herpes simplex viruses of type I

(HSV-1) and type II (HSV-2). It exhibits its antiviral activity by interaction with type I as well

as type II kinase (HSV-1-tk, and HSV-2-tk, respectively), both key enzymes in the herpes

simplex virus life cycle.

Figure 11. Conformationally restricted thymidine analogues.

Once phosphorylated to the triphosphate level, N-MCT triphosphate inhibits viral DNA

synthesis by inhibition of DNA polymerase. In addition, N-MCT has shown good in vitro

Introduction

13

activity against Kaposi’s sarcoma-associated herpesvirus (KSHV), displaying greater potency

than the reference compounds ganciclovir and cidofovir.42

When examining the processing of this compound in the cell, similarly opposed

conformational penchants towards kinases and DNA polymerase had been expected to be

observed as in the context of anti-HIV drugs acting on reverse transcriptase. The supposedly

general preference of cellular kinases for a southern conformation and the opposing

preference of DNA polymerase for a northern sugar conformation could indeed be confirmed

with the synthesis of southern-type analogue 21b. Comparing 21a and 21b, S-MCT 21b was

readily triphosphorylated, while much lower levels of the triphosphate of N-MCT 21a were

formed. Despite the high levels of S-MCT-5’-triphosphate, only triphosphorylated 21a was

incorporated into DNA and therefore displayed anti-HSV activity, while S-MCT-5’-

triphosphate was completely ineffective. However, examining the 3 phosphorylation steps in

detail, the first step in the processing of S-MCT and N-MCT, carried out by viral thymidine

kinases (HSV-1-tk or HSV-2-tk), strikingly seemed to prefer a northern conformation of the

drug, being more effective on 21a than on 21b. The second and third phosphorylation steps,

conducted by cellular thymidine kinases (tk1), clearly preferred a southern conformation,

while DNA polymerase only recognized and incorporated tri-phosphorylated northern

analogues.43 A possible explanation for the different behavior of viral HSV-tk and cellular

kinases against S-type nucleoside analogues can be found in the orientation of the

nucleobase; the latter is described by the glycosyl torsion angle χ (= torsion angle of the C1’-

C5’-N1-C2 sequence, Fig. 12) whose value determines the syn or anti disposition of the base

relative to the sugar moiety.

Figure 12. syn-anti base conformation.

For southern thymidine analogue 21b the preferred base conformation is syn (χ = 60°),

stabilized by a hydrogen bond between the C-7’-hydroxyl group and the C-2 carbonyl of the

pyrimidine (Fig. 12).50 Northern analogues preferentially adopt an anti conformation of the

base (χ = -150°). 44 This finding is in agreement with the trend observed in conventional

Introduction

14

nucleosides, where the nucleobase conformation tends to be syn for a southern sugar

pucker and anti for a northern sugar pucker,45 with the difference of a higher syn-anti energy

barrier for bicyclo[3.1.0]hexane based analogues such as 21b, a consequence of the fusion

of the cyclopropane ring immediately adjacent to the C-N bond.44b

Viral thymidine kinase HSV-tk prefers a southern sugar (or pseudosugar-) conformation but

with the nucleobase adopting an anti conformation.46 The significance of base conformation

for the recognition of bicyclo[3.1.0]hexane-based nucleoside analogs by HSV-tk was

confirmed with thymidine analogue 21c, which mimicks a south-like conformation, but

displays the nucleobase in an anti orientation. 21c was phosphorylated by viral HSV-tk at

least twice as effectively as 21b.47

1.4.2.3 Adenosine Deaminase

Adenosine deaminase (ADA) is a key enzyme in purine metabolism, catalyzing the

deamination of adenosine (22a) and 2’-deoxy-adenosine (22b), to the corresponding

inosines 24, presumably via the tetrahedral intermediates 23a or 23b, respectively

(Scheme 1).

Scheme 1. Adenosine deamination reaction catalyzed by ADA.

ADA regulates both intra- and extracellular adenosine concentrations. By regulating

extracellular adenosine levels, it modulates signal transduction through adenosine receptors,

e.g. the A2a receptor, and is considered an important factor in the control of inflammation.

ADA is ubiquitious in almost all human tissues, and anomalous levels of ADA have been

detected in a range of diseases such as AIDS (acquired immunodeficiency syndrome),

anemia, lymphomas, and leukemias.48 The inhibition of ADA could also be useful in the

therapy of viral infections and of some types of lymphoproliferative disorders.49 Furthermore,

ADA inhibitors can modulate the immune response in B-or T-cell malignancies,50 as

adenosine plays an essential role in the differentiation and maturation of the immune system.

The use of extracellular ADA inhibitors may offer cardiovascular protection in hypertension.51

Introduction

15

ADA inhibitors are also needed in order to prevent the deamination, which would lead to their

inactivation, of antileukemic and antiviral agents containing adenine bases.52

The most potent ADA inhibitors known are the natural product coformycin (25a) and its 2’-

deoxy analogue pentostatin (25b) (Fig. 13), with Ki values of 1 x 10-11 M and 2.5 x 10-12 M,

respectively.53 Their high activity is assumed to be due to nearly irreversible binding of these

compounds to ADA, by mimicking the transition state for the deamination reaction

(Scheme 1).

Figure 13. Structures of coformycin and pentostatin.

Pentostatin is a FDA-approved drug, for the treatment of hairy cell leukemia.54

Coformycin, as a highly potent in vitro inhibitor, was clinically tested but exhibited

unacceptable toxicities.55

Mammalian adenosine deaminase preferentially interacts with its natural substrate

adenosine in a C3’-endo sugar conformation and, likewise, inhibitors of ADA have been

found to bind to the enzyme in a C3’-endo conformation exclusively.56 Interestingly, ADA

from Plasmodium falciparum, the parasite causing malaria, appears to interact with

adenosine-type compounds adopting a C2’-endo sugar pucker.57 For the parasite, ADA is

essential since it provides the fundamental building blocks for its sole purine salvage

pathway.58 Unlike the human host’s ADA, however, Plasmodium ADA also acts as an

essential enzyme in the recycling of methylthiopurines from the polyamine biosynthetic

pathway.59 In this role, Plasmodium ADA catalyzes the deamination of 5’-

methylthioadenosine to 5’-methylthioinosine (Scheme 2), although less efficiently than it

catalyzes the deamination of adenosine. Human and other mammalian ADAs do not accept

5’-methylthioadenosine as a substrate.60

Introduction

16

Scheme 2. Deamination of 5-’methylthioadenosine (26) to 5’-methylthioinosine (28).

The reason for this difference in substrate acceptance exhibited by mammalian and

plasmodial ADA remained unclear, until in 2008 Merritt and co-workers57 provided insight into

the structural basis of this substrate discrimination. They reported the crystal structures of

plasmodial ADA in complex with its natural substrate adenosine, the purine guanosine, and

the picomolar inhibitor pentostatin (25b).

In all complexes of plasmodial ADA with adenosine-type compounds as well as with

pentostatin, the ribose ring adopts a C2’-endo sugar pucker, corresponding to a southern

conformation. This is not the case with mammalian ADA, where the ribose ring in all

complexes with adenosine-type compounds adopts a C3’-endo (northern) conformation. In

plasmodial ADA, a hydrogen bond between the sugar O3’ and Asp172 stabilizes the C2’-

endo sugar conformation, which results in C-5’ being pseudoaxial to the ribose ring, in

contrast to the 3’-endo conformation where C5’ is pseudoequatorial (Fig. 14). Thus, C5’

substituents on a C2’-endo ribose are positioned to interact with the binding site at a different

angle than would be possible for substituents on a C3’-endo ribose, and they are able to

sample a convenient environment within the active site. This explains plasmodial ADA’s

acceptance of C5’-thiosubstituted compounds as substrates. Importantly, these findings

suggest the feasibility of selective targeting of plasmodial ADA with conformationally fixed

ADA inhibitors (see section 2.4).

Introduction

17

Figure 14. (a and b) Alternate sugar pucker of substrate/inhibitor induced by the plasmodial ADA Asp172: mammalian ADA Met155 sequence difference. Plasmodial ADA is cyan and its bound pentostatin in orange, while mammalian ADA is green and its bound pentostatin in pink. Plasmodial ADA Asp172 hydrogen-bonds with the ribose 3′-hydroxyl group, an interaction that mammalian Met155 is incapable of making. This causes the plasmodial ADA-bound inhibitor to adopt a C2′-endo sugar pucker, while the mammalian ADA-bound inhibitor adopts a C3′-endo pucker. The result is that the 5′-carbon of the two riboses are oriented significantly differently with respect to the ribose ring, although the 5′-hydroxyl groups occupy nearly the same location and are less than 0.4 Å apart.57

1.4.2.4 Cytidine Deaminase

Cytidine deaminase (CDA) is a zinc-dependent enzyme that catalyzes the deamination of

cytidine to uridine, analogously to ADA deaminating adenosine to inosine61 (Scheme 1).

Therapeutically, inhibition of CDA is of specific interest to improve the activity of readily

deaminated antitumor nucleosides such as cytosine arabinoside or 2’-deoxycytidine.62

Marquez and co-workers63 synthesized the two bicyclo[3.1.0]hexane based analogues of the

known CDA inhibitor 29, with fixed northern (30, Fig. 15) and southern (31) conformations.

For comparative purposes they also investigated the flexible cyclopentane-based derivative

32 - to examine CDA’s preference for a certain sugar pucker. Since 29 inhibits CDA via a

mechanism independent from the zinc-promoted hydration mechanism for which the sugar

O4’ oxygen is critical, analogues of 29 were considered suitable compounds to study the

conformational preferences of CDA. Both the southern (31) and the northern (30) analogues

were found to be competitive inhibitors of CDA; the southern conformer 31 was found to be

2-fold more potent than the northern conformer. This finding confirmed the assumed

preference of CDA for a southern conformation, first derived from the result that all crystal

structures of inhibitors bound to CDA had shown the conformation of the sugar to be in the

southern hemisphere.64 This finding stood also in sharp contrast to the similar enzyme ADA,

which prefers a northern conformation of its substrates and is inhibited by nucleoside

analogues with a 3’-endo conformation (see section 1.5.3).

Introduction

18

Figure 15. Bicyclo[3.1.0]hexane based analogues of (29).

However, the flexible analogue 32 was also a competitive inhibitor of CDA. 32 was

approximately 100-fold more potent compared to the southern diazepinone 31, and thus also

more potent than northern conformer 30, with the original nucleoside 29 still being the most

active. A likely explanation for this smaller than expected potency of the bicyclo[3.1.0]-

hexane based southern diazepinone analogue is that its glycosyl torsion angle (χ = -149°) is

clearly in the syn region as X-ray structures have shown, leading to an unfavourable

orientation in space of the base moiety, although the pseudosugar conformation itself would

enhance affinity. Supporting this assumption is the finding that the crystal structure of CDA in

complex with parent riboside 29 shows that its glycosyl torsion angle is clearly in the anti

range.44b

1.4.2.5 DNA Methyltransferases

DNA methyltransferases (DNMTs) are enzymes transferring methyl groups to DNA, using

S-adenosyl-methionine (AdoMet, 33, Fig. 16) as the methyl donor molecule. AdoMet is

converted to S-adenosylhomocysteine (SAH, 34) during the reaction.

Changes in the pattern of DNA methylation are associated with cancer, as increased or

decreased methylation has been identified in all types of cancer cells so far.65 Generally,

genomic tumor DNA is characterized by definite methylation changes that are also termed

epimutations. As such epimutations appear rarely in healthy tissue, the epigenetic approach

to cancer therapy may have high tumor specificity.66

Introduction

19

Figure 16. Structures of AdoMet (33) and SAH (34).

With structural modification of S-adenosylhomocysteine, the product inhibitor of the

methylation reaction, Lim and co-workers recently achieved selective inhibition of Dengue

virus methyltransferase.67 The development of selective inhibitors of viral methyltransferases

in general could also be of high therapeutic interest.

DNA (cytosine C5)-methyltransferase as one important representative of the DNMT enzyme

family catalyses the methylation of carbon-5 of cytosine in the DNA. The enzyme links

covalently to C-6 of cytosine, along with a protonation at N-3, thus forming an activated

enamine intermediate; C-5 is rendered nucleophilic and attacks the methyl sulfonium center

of AdoMet. Evidence for this mechanism stems from X-ray crystallographic studies of DNA

(cytosine C5)-methyltransferase linked to methylated 5-fluorocytosine in DNA. The cytosine

residue targeted for methylation was “flipped” out of the helix during the transfer reaction.68

The removal of the target cytosine base, i.e. introduction of an abasic site, enhanced binding

of DNA (cytosine C5)-methyltransferase to DNA and the conformation of the sugar-

phosphate backbone at the abasic site in the resultant complexes was the same as that of

the sugar attached to a “flipped” cytosine. Conformationally constrained sugar analogues

based on bicyclo[3.1.0]hexane templates were placed in DNA duplexes as abasic target sites

in the DNA (cytosine C5)-methyltransferase recognition sequence. Biochemical studies

demonstrated that the binding affinity of the enzyme for abasic sites increases when the

abasic target sugar is constrained to the southern conformation and decreases when it is

constrained to the northern conformation.69

1.4.3 Receptors Discriminating between Sugar Pucker : Adenosine and

P2Y-Receptors

1.4.3.1 Adenosine Receptors

The nucleoside adenosine plays an important role in many aspects of cellular metabolism. It

influences different mammalian organ systems, specifically the cardiac, nervous, and

Introduction

20

immune system.70 Biochemical and pharmacological studies have validated that most of

adenosine’s physiological actions are mediated via cell surface receptors, of which there are

four subtypes: A1, A2a, A2b, and A3-receptors (AR’s). They are G-protein-coupled receptors of

which the A1 adenosine receptors are the best studied within the AR family; agonists and

antagonists for this receptor have therapeutic potential as anti-arrhythmic, neuroprotective,

and analgesic agents.71

A3 AR agonists and antagonists are proposed for the treatment of cancer and inflammatory

diseases.72

Jacobson and co-workers examined the conformational preferences of the A1, A2A, and A3

receptors using the conformationally restricted adenosine analogues 35 and 36 (Fig. 17) and

some N6-modified derivatives.72, 73 All three receptors displayed a higher binding affinity for

northern adenosine analogues (with an affinity ratio northern to southern > 150 for the A3-

receptor).

N

N

N

N

NH2

HO

HO OH

N

N

N

N

NH2

HO

HO OH

35 36

Figure 17. Northern- (35) and southern (36) adenosine analogues.

By activating either the A1 or A3 adenosine receptors in cardiac myocytes in several species

a cardioprotective effect mimicking the one induced by ischemic preconditioning could be

achieved.74 In contrast to a single activation of either A1 or A3, the coactivation of A1 and A3

adenosine receptors seems to be cardioprotective to a higher degree than activation of either

subtype alone. In agreement with their earlier work, Jacobson and co-workers deduced a

preference of A1 and A3 receptors for a northern sugar pucker72, 75 by synthesizing and

testing conformationally restricted (N)-methanocarba 2N-disubstituted adenine nucleoside

analogues as dual acting A1 and A3 adenosine receptor agonists (Fig. 18).

Introduction

21

Figure 18. Dual acting A1 and A3-receptor agonists

The derivatives 37-41 are all dually selective agonists for A1 and A3 receptors with nearly

balanced affinities, (Ki values in the range of 4 nM – 0.1 µM).

For the A2B receptor,76 there possibly exists a preference for one particular sugar pucker as

well, but this preference has not been elucidated.

1.4.3.2 P2Y Receptors

P2Y receptors are G-protein coupled receptors of which eight mammalian subtypes

(P2Y1,2,4,6,11-14) are currently known. For the activation of the P2Y1, P2Y11-13 subtypes adenine

nucleotides are required; while uracil nucleotides activate P2Y2, P2Y4, and P2Y6 subtypes.

P2Y receptor ligands are exploited for therapeutic applications in the cardiovascular and

endocrine systems and in relation to Alzheimer’s disease, and new ligands are also needed

as pharmacological tools for more basic research regarding these receptors, since detailed

structure-activity relationship (SAR) studies have only been established for P2Y1 and P2Y12

receptors.77

For the P2Y1 receptor, a preference of the receptor for the northern conformation of the

interacting ribose ring could be identified by testing northern bicyclo[3.1.0]hexane based

analogues78 43 and 44 of the known antagonist N-methyl-2’-deoxyadenosine 3’,5’-

bisphosphate (Fig. 19, relevant functional groups shown in green).

Figure 19. P2Y1 agonist 42 (relevant functional groups shown in red) and antagonists 43, 44 (green).

Introduction

22

The presence of the bicyclo[3.1.0]hexane moiety in these nucleotide analogues both

enhanced receptor affinity and improved stability towards nucleotidases. The same

conformational constraint of the ribose moiety that enhances antagonist action also improves

the potency and selectivity of P21 nucleotide agonists. The northern bicyclo[3.1.0]hexane

based compound 42 proved to be the much more potent and selective agonist of the P2Y1

receptor (Fig. 19, functional groups shown in red) than the parent nucleotide 2-methylthio

adenosine diphosphate.79 A comparison of adenosine triphosphate analogues with a

constrained northern or southern conformation had hinted at this preference of the P2Y1

receptor for a northern sugar pucker earlier: the southern analogue was less active than the

northern one, and the same was found for the P2Y2 receptor.80

A similar preference can be assumed for the P2Y14 receptor; here a bicyclo[3.1.0]hexane-

based analogue with a constrained southern conformation of uridine diphosphate glucose, a

nucleoside agonist of P2Y14, was tested and found to have no activity at all, which leads to

the conclusion that this receptor subtype also prefers a northern conformation of the sugar

moiety.81 Using conformationally locked nucleotides, preferences for the northern

hemisphere of the pseudorotational cycle could also be deduced for the P2Y4, and P2Y11

receptors.82 The P2Y6 receptor, however, prefers a southern conformation which was

predicted by molecular modelling and confirmed by synthesis of a rigid southern

bicyclo[3.1.0]hexane-based analog of uridine diphosphate (UDP, Fig. 20).

Figure 20. Selective P2Y6 agonist with constrained southern conformation.

Southern UDP analogue 45 proved to be more potent than UDP itself, whereas a uridine 5’-

diphosphate analogue locked in the northern conformation was devoid of any activity,

supporting the unique preference of the P2Y6 receptor for a southern conformation.83

1.5 Synthesis of Bicyclo[3.1.0]hexane Systems

1.5.1 Natural Products

Liu & Nan have recently described the synthesis of the bicyclo[3.1.0]hexane–containing core

framework of lindenane-type sesquiterpenoids, such as, e.g., chlorajaponilide and

Introduction

23

chlorahololides, see section 1.2).84 Scheme 3 summarizes the key steps of the synthesis of

this tricyclic scaffold, which departed from racemic bicyclic lactone 46. Methylation and TBS-

protection gave ketone 48, which was then converted into protected alkynol 49 in 8 steps in

63% overall yield. Pd-catalyzed reductive cyclization of 49 provided the bicyclo[3.1.0]hexane

derivative 50 in high yield (97%) and as a single diastereomer. The latter was then

elaborated into 55 in 7 steps in moderate overall yield (12%).

Scheme 3. i. CH3Li, THF/Et2O, -78°C. ii. TBSCl, im, 73% (over 2 steps). iii. P d(dba)2, Et3SiH, BBEDA,

HOAc, 97%. iv. TBAF, 98%. v. Swern cond., 94%. vi. p-TsOH, 100%. vii. Swern cond., 93%. viii.

Pd(OAc)2, Et3N, DMF, 80°C, 4h, 40%. ix. TBT-SO 2Me, NaHMDS, 57%.

Lu & Peng85 have recently reported the synthesis of biyclo[3.1.0]hexane derivatives 57 and

58, which they propose could serve as precursors for chlorahololides via 56 (Scheme 4). The

synthesis of both 57 and 58 is based on the Wieland-Miescher ketone 59 as a readily

available chiral starting material (Scheme 5).

Introduction

24

Scheme 4. Retrosynthesis of Chlorahololide A according to Lu & Peng.85

Thus conversion of 59 into mesylate 60 and treatment of the latter with base triggered an

intramolecular SN2 -type nucleophilic substitution reaction that provided bicyclo[3.1.0]hexane

derivative 61 in high yield (90%). 61 was then elaborated into 57 and 58 in 2 or 3 steps,

respectively.

Scheme 5. i. KHMDS, THF, -5°C, 2h, 90%.

Introduction

25

1.5.2 Sugar Analogues

In 2008, Li and co-workers86 synthesized (±)-74, a conformationally restricted amino

analogue of an α-(D)-galactofuranose ring. Using (±)-74 as a precursor, β-(D)-

arabinofuranose ring analogue 75 could also be prepared (Scheme 6). Both the analogues

were intended to be used as mimetics for UDP-α-(D)-galactofuranose, and decaprenyl β-(D)-

arabinofuranosylphosphate, respectively, the latter both playing important roles in the

synthesis of the cell wall of Mycobacterium tuberculosis and other mycobacterial

species.87

The synthesis departed from allylic alcohol 62 which can be accessed from cyclohexane-1,4-

dione in 3 steps.88 After acetal hydrolysis and MOM-protection epoxidation of 64 led to 65 as

the substrate for a ring contraction reaction that would install the bicyclo[3.1.0]hexane moiety

that was induced by treatment with base. This transformation gave a ~1:1 mixture of (±)-66a

and (±)-66b , which were separated by chromatography; both of these isomers could be

converted by different methods to the corresponding benzoates (±)-67. (±)-66b was

converted to (±)-67 by a Mitsunobu reaction. Subsequent α-selenation under acidic

conditions, oxidation of the selenide to the selenoxide and in situ elimination gave enone (±)-

68, which was then elaborated into azide (±)-73 by standard transformations in 6 steps. From

azide (±)-73, target structure 75 was received in 6 steps, including a racemic resolution, in a

high overall yield of 62%. (±)-74 was obtained from (±)-73 by reduction of the azide moiety.

Introduction

26

Scheme 6. i. oxalic acid, acetone/H2O 1:1, rt, 3h, 92%. ii. (CH3O)2CH2, LiBr, p-TsOH, rt, 1h, 72%. iii.

m-CPBA, CH2Cl2, rt, 2h, 70%. iv. 2 M NaOH, EtOH, rf, 20min, 40% (±89) and 47% (±90). v. BzCl, py,

rt, 2h, 93%. vi. BzOH, Ph3P, DIAD, THF, 84%. vii. 1) PhSeCl, HCl, EtOAc, rt, 2h, 45%. 2) NaIO4, H2O,

THF, rt, 12h, 95%. viii. H2O2, NaOH, CH3OH, 0°C, 1h, 79%. ix. 3.5 M H 2SO4 aq., 50°C, 12h, 70%. x.

BzCl, py, 0°C--> 10°C, 2h, 90%. xi. NaBH 4, CH3OH, -30°C, 20min, 83%. xii. (PhO) 2PON3, Ph3P, DIAD,

THF, rt, 20min, 84%. xiii. NaOCH3, CH3OH, rt, 30min, 91%. xiv. H2, Pd/C, THF, 97%.

Introduction

27

Introduction

28

1.5.3 Bicyclo[3.1.0]hexane-based Nucleoside Analogu es

In general, there are two main approaches to the synthesis of carbocyclic nucleoside

analogues, the first one employing carbohydrates as starting materials to afford optically pure

compounds, the second one would be a total synthesis approach starting either from racemic

or achiral compounds or from readily available chiral starting materials other than

carbohydrates. The first possibility bears the advantage of enantiomerically pure starting

materials which also possess the necessary polyoxygenated framework, but the

interconversion of one configuration to another is often not accomplishable without the

excessive use of protecting groups.

In the synthesis of carbocyclic nucleoside analogues, there is the additional question of how

the nucleobase is introduced best. The task can be accomplished by a convergent or a linear

strategy, as exemplified in Scheme 7 for the synthesis of N-type bicyclo[3.1.0]hexane-based

nucleoside analogs from a cyclopentene precursor. In the first approach, the complete

nucleobase is coupled directly to an activated carbocycle by a Mitsunobu reaction or by

displacement of a leaving group. A variety of carbocyclic nucleoside analogues can be

prepared in this way relatively fast, as the bases are introduced as ready moieties. The

disadvantage of this approach is the undesired formation of regioisomers, as the

nucleobases possess several nucleophilic sites, and the separation of the resulting mixtures

of isomers can be a tedious task.89

NH2

RO

RO

R1O

OH

RO

RO

NHR'

Convergent approach

(Mitsunobu or SN2 rct)

Linear approach

HO

RHO RHO

OH

HO

RHO OH or H

BASE

Scheme 7. Synthesis of northern bicyclo[3.1.0]hexane nucleoside analogues via a convergent vs.

linear strategy.

In the linear approach, a prefunctionalized carbocyclic amine has to be synthesized first, and

the heterocyclic base is then built up in a step-wise fashion by modification of the carbocyclic

amine. Employing this strategy, there are no regioisomers formed, however, each nucleoside

analogue has to be prepared by a separate multistep synthesis.

A convergent approach is only applicable for northern nucleoside analogues.

Introduction

29

1.5.3.1 Northern (N)-Type Analogues

Bicyclo[3.1.0]hexane-based nucleoside analogues with a fixed northern conformation are

more readily accessible and usually require shorter routes than the southern conformer, as

will be seen in the subsequent examples.

The first synthesis of a bicyclo[3.1.0]hexane based nucleoside analogue with northern

conformation was reported in 1994 by Altmann and co-workers.44a They synthesized

bicyclo[3.1.0]hexane-based thymidine analogue 82 that was incorporated into

oligonucleotides as a conformationally constrained sugar analogue (Scheme 7). Their

synthesis started with optically active cyclopentenone 76 which was accessible from D-ribose

in 7 steps.90

82

HO

N

HO

NH

O

O

OH

BnOO

O O

BnO

O O

OHBnO

O O

76 77

i ii iii, iv, v1'2'3'

4'5'

81

O

N

O

NBOM

O

O

Si

SiO

HO

HO OH

NNH

O

O

NH2BnO

O O

vi, vii, viii

ix, x, xi, xii xiii, xiv

80

78 79

Scheme 7. i. NaBH4, CeCl3, MeOH, 0°C, 10min, 97%. ii. Et 2Zn, CH2I2, CH2Cl2, 0°C to rt, 15h, 90%.

iii. TsCl, Et3N, CH2Cl2, DMAP, rt, 48h, 77%. iv. NaN3, DMF, 70°C, 18h, 88%. v. H 2, Lindlar’s cat., 4h,

100%. vi. MeOCH=C(Me)CONCO, CH2Cl2, -78°C-->rt, 30min, 95%. vii. 0.2 N HCl, EtOH/H 2O 9:1, rf,

20h, 80%. viii. H2, Pd/C, EtOAc/MeOH 1:1 (84% ee). ix. TIPSCl2, im, DMF, 67%. x. BOMCl, DBU,

CH3CN, rt, 1h, 85%. xi. CH3C6H4OC(S)Cl, DMAP, Et3N, CH2Cl2, rt, 3h, 40°C, 18h, 90%. xii. 1) Bu 3SnH,

AIBN, DME, 80°C, 3h. 2) prep. HPLC on Chiralgel OD, 250 x 4.8 mm, hexane/isopropanol 95:5, 85%

(100% ee). xiii. TBAF, THF, rt, 4h, 99%. xiv.1) H2, Pd/C, rt, 2h. 2) NaOMe, rt, 20h, 88%.

Subsequently, an additional stereocentre at C1 was installed via selective reduction to allylic

alcohol 77 by means of sodium borohydride and cerium(III)chloride, which afforded 77 in an

optical purity of approximately 50%.

Simmons-Smith cyclopropanation of 77 using Et2Zn and CH2I2 gave bicyclo[3.1.0]hexane

alcohol 78 as a single diastereosiomer, due to the directing effect of the allylic hydroxyl

Introduction

30

group.91 78 was converted to amine 79 via tosylation, azide formation and subsequent

reduction of the azido group. The base moiety was built up in a linear fashion by reacting 79

with (E)-3-methoxy-2-methylacryloyl isocyanate, followed by acid-catalyzed cyclization of the

resulting acryloyl urea with concomitant cleavage of the acetonide, and removal of the benzyl

group to afford 80. Simultaneous re-protection of the C5- and C3- hydroxyl groups in a cyclic

fashion with a TIPS-protecting group and BOM-protection of the thymine imido group allowed

for the removal of the C2-hydroxy group via radical deoxygenation to yield 81. Upon reaction

with tolyl chlorothioformate, the C2-hydroxy group underwent conversion to the

thiocarbonate, and radical reduction of the thiocarbonate with Bu3SnH in the presence of

AIBN furnished 81 as a partially racemic compound (84% ee). Complete racemic resolution

was achieved by preparative chiral HPLC, followed by removal of the TIPS- as well as the

BOM-protecting group with TBAF and catalytic hydrogenation over Pd/C, respectively, to

yield thymidine analogue 82.

Marquez and co-workers developed a synthesis of northern adenosine analogue 88 in 1995

(Scheme 8).92 They installed the bicyclo[3.1.0]hexane moiety in the same way as Altmann

and co-workers, starting from 76 with a stereoselective reduction and a Simmons-Smith

cyclopropanation to access 78 which was converted to the mesylate 83 quantitatively.

Following a convergent approach, condensation of 83 with adenine in the presence of K2CO3

and 18-crown-6 ether gave the desired N-9 product 84 as the major component (68%)

together with a small amount of the N-7 regioisomer (8%). Deprotection of the 5’-O-benzyl

ether using palladium black and formic acid, followed by the removal of the isopropylidene

group with 80% acetic acid, gave target compound 86. The primary alcohol function in 85

was oxidized to the aldehyde 87 using tetrapropylammonium perruthenate (TPAP), and

deprotection of the acetonide function afforded a second target compound 88.

Introduction

31

Scheme 8. i. MsCl, py, 0°C, 4h, 100%. ii. Adenine, K2CO3, 18-crown-6, DMF, 100°C, 18h, 68%. iii. Pd

black, HCO2H, MeOH, rt, 24h, 75%. iv. 80% AcOH, 80°C, 5h, 80%. v. TPAP, CH3CN, CH2Cl2, rt, 30h,

60%. vi. 88% HCO2H, rt, 5h ---> aq. 29% NH4OH, rt, 10min, 90%.

Starting with the same chiral cyclopentenone 76, the 2’-deoxy analogue of 86 was

synthesized by Siddiqui et al. in 1996 in a 7-step sequence with an overall yield of 25%.93 As

outlined in Scheme 9, acetonide 76 was converted into tert-butyl ether 89 by treatment with

AlMe3. The extra hydroxyl group was removed by a Barton deoxygenation in a 2-step

protocol to give 92, followed by deprotection of the TBS-group and subsequent modified

Simmons-Smith cyclopropanation to give 94.

Introduction

32

Scheme 9. i. AlMe3, CH2Cl2, -78°C -->rt, 18h. ii. TBSCl, im, DMF. iii. CS 2, NaH, MeI, THF, 1h, 82%. iv.

n-Bu3SnH, AIBN, tol, 120°C, 1.5h, 77%. v. TBAF, THF, rt, 12h, 92%. vi. Sm, HgCl2, ClCH2I, THF, -

78°C --> rt, 12h, 96%.

The construction of the base was also elaborated in a convergent approach, by a Mitsunobu

reaction of compound 94 with 6-chloropurine. Nucleophilic aromatic substitution of the

chloride atom and global deprotection with BCl3 concluded the synthesis of 2’-deoxy-

adenosine analogue 97 (Scheme 10).

Scheme 10. i. Ph3P, DEAD, 6-chloropurine, THF, rt, 3d, 58%. ii. NH4OH conc., dioxane (sealed tube),

65°C, 14h, 76%. iii. BCl 3, CH2Cl2, -78°C, 4h, 70%.

Introduction

33

In 1997, Marquez and co-workers developed a method that circumvented the 2-step

deoxygenation protocol to remove the extra hydroxyl group and that should allow the

synthesis of both conformers, southern and northern, proceeding through the same central

intermediate, azido-selenoxide NS-I (retrosynthesis depicted in Scheme 11).94

Scheme 11. Retrosynthesis of northern and southern nucleoside analogues according to Marquez et

al.

Theoretically, syn-elimination of NS-I could proceed in two directions, giving either the allylic

azide or the vinyl azide (abstraction of Ha or Hb, respectively). Experimentally, however, the

allylic azide predominated overwhelmingly over the vinyl azide, despite the inductive

polarization associated with the azido group, which might have been expected to facilitate

removal of Hb. Therefore, this strategy turned out to give access to northern conformers only,

via nucleoside analogue amine precursor 105 (Scheme 12).

The synthesis started with cyclopentadiene, which was stereoselectively converted to 98 via

deprotonation with NaH, subsequent quenching with benzyl chloromethyl ether and then

reaction with diisopinocampheylborane as the asymmetric hydroborating reagent. Oxidative

work-up gave cyclopentenol 98 in an enantiomeric excess of 94%-97%, however with

relatively low yield.95

Introduction

34

Scheme 12. i. NaH, BOMCl, THF, -60°C, 3h. ii. (-)-( ipc)2BH, -60°C --> rt, 15 h, 45%. iii. Me 3CPh2SiCl,

im, DMF, rt, 14h, 76%. iv. PhSeCl, NaN3, DMSO, rt, 12h, 87%. v. NaIO4, MeOH/H2O 9:1, rt, 36h, 76%.

vi. Ph3P, THF (wet), rf, 4h, 90%. vii. 1) C6H6(CO)2O, py, 90°C, 2h. 2) Ac 2O, 90°C, 2h, 77%. viii.

Et3N·HF, CH3CN, rf, 12h, 90%. ix. Et2Zn/CH2I2, CH2Cl2, -20°C, 10h, 96%. x. H 2NNH2, MeOH, 50°C,

3.5h, 100%.

TBDPS-protection of 98 was followed by azido-phenylselenylation, which proceeded with

complete stereochemical control. This stereochemical outcome was possibly due to the

preferred pseudoequatorial orientation of the two substituents in the cyclopentene ring, which

led to formation of the episelenonium intermediate from the bottom face of the ring to give a

stable chair-like transition state intermediate. In situ oxidation of the phenylselenide group

triggered the anticipated syn-elimination, giving almost exclusively the allylic azide 101. An

explanation for this finding is lacking, but there is some literature precedence for the

predominance of the allylic azide under similar oxidative conditions.96 Azide 101 was reduced

under Staudinger conditions and the resulting amine was protected with a phthalimido group.

Full amine protection proved necessary since an acyl-NH group, e.g., took precedence over

the hydroxyl group in directing delivery of the methylene group in the Simmons-Smith

reaction. After deprotection of the hydroxyl group the succeeding Simmons-Smith

cyclopropanation gave exclusively 105 in 96% yield. Hydrazinolysis of the phthalimido group

afforded bicyclo[3.1.0]hexane amine 106.

Introduction

35

A similar strategy also shortened access to northern conformer building block 112 allowing

for convergent approaches by starting from the same cyclopentenol synthon 98 that

Marquez’ group had used earlier for the synthesis of southern nucleoside analogues.

Scheme 13. i. NaH, BnBr, TBAI, THF, rt, 5h, 95%. ii. PhSeCl, AgCF3CO2, DMSO, rt, 1h. iii. 5%

KOH/EtOH, 0°C, 30min, 74%. iv. NaIO 4, MeOH/H2O 9:1, rt, 1h, 73%. v. Ph3P, DEAD, PhCOOH, rt,

30min, 85%. vi. K2CO3, MeOH, rt, 3h, 80%. vii. Et2Zn/CH2I2, CH2Cl2, -20°C--> rt, 6h, 80%.

In the same manner as with the PhSeCl/NaN3 system, the double bond in 107 reacted regio-

and stereoselectively with PhSeCl/AgCF3CO2 to give the trans-2-(phenylseleno)-

cyclopentanol 108 after hydrolysis of the trifluoroacetate ester. Oxidation and in situ

selenoxide-pyrolysis gave also exclusively allylic alcohol 109. This allylic alcohol was

inverted via a Mitsunobu esterification, providing the new alcohol 111 upon hydrolysis.

In 2002, Choi and co-workers presented alternative approaches to northern nucleoside

analogues, for both the 2’-, 3’-hydroxy derivatives as well as the 2’-deoxy compounds.97 Key

steps/methods included intramolecular cyclopropanation via a carbenoid intermediate to

generate the bicyclo[3.1.0]hexane scaffold and lipase-catalyzed racemic resolution of the

corresponding 2’-deoxy scaffold. The enantiospecific synthesis of 120, as outlined in Scheme

14, started with chiral precursor D-isoascorbic acid, from which 113 was obtained in 4 steps

and 60% overall yield.98 After a stepwise oxidation of the hydroxyl group, a Claisen

condensation of the activated acid with 2-lithio ethyl acetate afforded 114 with 14% of

undesired epimerization. Diazo transfer was accomplished with tosyl azide and triethylamine

to give compound 115, which underwent metal-catalyzed thermolysis to 116 in 44% yield (+

unwanted diastereomer in 19% yield) via a carbenoid intermediate. DIBALH-reduction of the

carbonyl moieties gave the corresponding diol, which was benzylated. Cleavage of the

acetonide afforded 117 and treatment with thionyl chloride then resulted in the formation of

Introduction

36

118. Subsequent reaction with sodium azide led to 82% of 119 and 10% of the alternative

regioisomer. Palladium-catalyzed hydrogenation of the azide gave amine 120.

Scheme 14. i. 1) COCl2, Et3N, DMSO, 30min. 2) NaClO4, 67%. ii. 1) Im2CO 2) LiCH2CO2Et, 67%. iii.

TsN3, TEA, 83%. iv. CuI, tol, rf, 63%. v. DIBALH, 55%. vi. BnBr, NaH, 99%. vii. H+, 74%. viii. SOCl2,

99%. ix. NaN3, 82%. x. H2, Pd/C, 81%.

An alternative approach to an aldehyde corresponding to ester 114 has been reported by

Jacobson and co-workers.99 The latter group has also employed L-ribose as a starting

material for northern-type bicyclo[3.1.0]hexane-based nucleoside analogs.

In this approach (Scheme 15) L-ribose is converted to iodides 121 (diastereomeric mixture

5:1); subsequent reductive cleavage of the mixture with Zn in refluxing methanol100

generated aldehyde 122 which was C-2 elongated in situ to 123. Next, the same key steps

as with Choi et al. were employed to install the bicyclo[3.1.0]hexane moiety (diazotization

and intramolecular cyclopropanation over a carbenoid intermediate) to give 125, along with

the diastereomer in a ratio of 4:1 in favor of the desired diastereoisomer. 125 was isolated

chromatographically, reduced stereoselectively with NaBH4 and equilibrated with TFA to the

isomeric acetonide 127, to which different bases can be attached via a convergent approach.

Introduction

37

O

HO OH

L-ribose

iOMeO

O O

121

iiOHHO I iii

OO

OEtO

O

OO

OEtO

O

123

124

N2

OEtO

O O

O

+ diastereomerdr 4:1

iv

v OEtO

HO O

OOH

EtO

O O

O

vi vii

125 126 127

122

dr 5:1

~1:4

O

O

O

H

Scheme 15. i. 1) HCl conc., acetone, MeOH, rf, 3h. 2) Ph3P, im, tol/CH3CN, I2, 70°C, 1h, 69%

(mixture). ii. Zn, i-PrOH/CH3COOH, 30°C, 2h. iii. N 2CHCOOEt, SnCl2, CH2Cl2, 0°C -->rt, 2h, 68%. iv.

TsN3, Et3N, CH3CN, 0°C-->rt, 1h, 95%. v. CuI, tol, rf, 1h, 51% ( 101). vi. NaBH4, EtOH, rt, 1h, 69%. vii.

TFA, acetone, rt, 6h, 35%.

Most recently, a ring-closing metathesis (RCM) approach for the synthesis of northern

thymidine analogue 21a has been published by Yang and co-workers.101 Their strategy is

outlined in Scheme 16.

As chiral starting material they used D-mannitol, which was converted to aldehyde 128 in 2

steps.102 Subsequently, 128 underwent allylation, benzyl protection and acetonide cleavage

affording diol 131. This compound was selectively TBS-protected, oxidized and converted to

134 via a Grignard reaction. Ring-closing metathesis employing 2nd generation Grubbs

catalyst gave cyclopentene derivative 135. A Tsuji-Trost reaction led to compound 136,

followed by a Simmons-Smith cyclopropanation to give bicyclo[3.1.0]hexane derivative 137,

which can be converted to 21a via Mitsunobu chemistry. In this way, the central intermediate

137 was reached in 9 steps with a 25% overall yield.

Introduction

38

Scheme 16. i. CH2CHCH2Br, Zn, NH4Cl, 0°C--> rt, 4h, 90%. ii. BnBr, NaH, THF, 0°C-->r t, 5h, 92%. iii.

60% AcOH aq., rt, 12h, 80%. iv. TBSCl, im, THF, 0°C , 8h, 80%. v. NaIO4, CH2Cl2, rt, 12h, 98%. vi.

CH2CHMgBr, THF, -78°C, 94%. vii. 1) 5 mol% 2nd gen. Gr ubbs cat., CH2Cl2, rt, 12h. 2) Ac2O, Et3N,

DMAP, 85%. viii. p-benzoquinone, PdCl2(MeCN)2, THF, 86%. ix. 1) 1% NaOH, MeOH. 2) CH2I2, Et2Zn,

CH2Cl2, 82%.

1.5.3.2 Southern (S)-Type Analogues

The synthesis of bicyclo[3.1.0]hexane based nucleoside analogues with a fixed southern

conformation is in general more lengthy and elaborate than the synthesis of northern

conformers. Retrosynthetic analysis of southern derivatives clearly identifies the reason for

this: with the bases (or according analogues) attached to a bridgehead carbon atom, these

compounds have to be constructed from a suitably protected bicyclic amine, with no

possibility of introducing the bases as whole moieties by convergent approaches, as it is

possible in the case of northern conformers (cf. Scheme 4).

In 1994, Altmann and co-workers had synthesized a southern bicyclo[3.1.0]hexane based

analogue of thymidine with the aim of incorporating it into antisense oligodeoxy-

ribonucleotides.103 The corresponding synthesis will be discussed later in detail as it was

included in the experimental approaches used in this thesis for the synthesis of target

structures with bicyclo[3.1.0]hexane based architectures.

Introduction

39

In 1996, Marquez and co-workers presented an approach to the synthesis of 2’-deoxy-

bicyclo[3.1.0]hexane based southern conformers bearing all common bases.104 The

synthesis was centered around the stable carbamate derivative 145, from which all the

heterocyclic bases could be constructed (Scheme 17). Starting point for the synthesis was

cyclopentene 98, which had been used before as chiral starting material in the synthesis of

northern-type conformers.94 Hydroxyl-directed stereoselective epoxidation and protection of

the free alcohol group95a gave compound 139.

Scheme 17. i. t-BuOOH, Va(acac)2, 1,2 dichloroethane, -30°C-->rt, 2h. ii. NaH, BnBr , TBAI, THF, rt,

12h, 81%. iii. KCN, LiClO4, CH3CN, rt, 3d, 77%. iv. Im2CS, DMAP, DMF, rt, 3h-->∆, 70°C, 30min, 98%.

v. CH2N2, Et2O, 0°C, 3d, 96%. vi. h ν, COPh2, 2h, 79%. vii. 25% aq. NaOH, MeOH, rf, 24h, 99%. viii.

Et3N, DPPA, tol, rt, 2h --> 80°C, 2h --> Me 3Si(CH2)2OH, 80°C, 2h --> rt, 12h, 64%. ix. TBAF,

CH3CN/THF 4:1, 70°C, 4h, 99%.

Nucleophilic opening of the epoxide ring occurred with excellent regioselectivity to give the

cyano intermediate 140, from which the desired α,β-unsaturated nitrile 141 was obtained

following the syn-β-elimination of the intermediate thiocarbonylimidazolide. The 1,3-dipolar

cycloaddition of diazomethane with 141 to give the cis-fused pyrazoline intermediate 142

occurred with the expected regioselectivity that is typical of diazomethane additions to

electron deficient alkenes in which the carbon atom of diazomethane functions as the

negative end of the dipole. The stereofacial selectivity is controlled by the the approach of

the diazomethane from the less hindered side of the double bond. Nitrile 143 was obtained

after nitrogen extrusion from 142 by photolysis. After hydrolysis of 143 to the acid, a modified

Curtius rearrangement of the corresponding acyl azide to the isocyanate, which was trapped

with 2-trimethylsilylethanol, gave stable carbamate 145.

Introduction

40

From the central intermediate 145 the elaboration of pyrimidine or purine bases could be

carried out by reacting amine 146 with 3-methoxy-2-methylacryloyl isocyanate, 3-

ethoxyacrylyol isocyanate, 5-amino-4,6-dichloroformylpyrimidine or 2,5-diamino-4,6-

dichlorodiformylpyrimidine, respectively, followed by final benzyl deprotection procedures to

give compounds 151-155. (Scheme 18).

Removal of the benzyl ethers was accomplished either by catalytic transfer hydrogenation, or

by treatment with BCl3. In the case of the target purine derivatives 154 and 155, the

nucleophilic substitution of the chloride attached to pyrimidine would only occur if 5-

formylamino-4,6-dichloropyrimidine was used instead of the free 5-amino-4,6-

dichloropyrimidine, the same was the case for 2,5-diamino-4,6-dichlorodiformylpyrimidine as

reagent.

Scheme 18. Elaboration of the base part of S-type bicylco[3.1.0]hexane-based nucleoside analogues.

More recently, Marquez and co-workers described the synthesis of a 4’-deoxy-

bicyclo[3.1.0]hexane template (160) by an approach making use of an intramolecular variant

of the Kulinkovich reaction (Scheme 19).105 Thus, (R)-epichlorohydrin was subjected to a

Introduction

41

copper bromide-promoted addition with vinylmagnesium chloride to produce alcohol 156.

Nucleophilic exchange of the chloride with cyanide afforded nitrile 157, which was hydrolyzed

to the acid. Without isolation, activation of the acid with EDAC and hydroxybenzotriazole

(HOBt) gave the key δ-vinyldibenzylamide 158 upon reaction with dibenzylamine.

Scheme 19. i. CuBr, vinyl-MgBr, Et2O, -70°C--> -40°C, 2d, 84%. ii. KCN, MeOH, 70°C, 24 h, 84%. iii.

1) 25% NaOH aq., MeOH, 90°C, 24h. 2) Bn 2NH, EDAC, HOBt, DMF, rt, 16h, 65%. iv. MOMCl, DIPEA,

CH2Cl2, 0°C--> rt, 16h, 99%. v.Ti(O- i-Pr)4, i-PrMgCl, THF, rt, 1h. vi. 3% HCl, MeOH, 60°C, 4h, 4 7%.

δ-vinyldibenzylamide 158 was MOM-protected, and subsequent Kulinkovich reaction106 of

159 with stoichiometric amounts of Ti(O-i-Pr)4 in the presence of i-PrMgCl in THF afforded a

1.5:1 mixture of diastereomers which after hydrolysis of the MOM group were separated by

column chromatography to give 160 (and the corresponding diastereomer, not shown).

Introduction

42

1.6 Conclusions

Exploiting the unique conformational characteristics of the bicyclo[3.1.0]hexane scaffold, the

conformational preferences of a number of nucleoside- and nucleotide-interacting proteins

could be elucidated. This includes viral RT enzymes, viral and host kinases, and also

GPCR’s such as the adenosine or P2Yx receptors. The bicyclo[3.1.0]hexane moiety’s special

feature, the preference for a boat-like conformation of the six-membered ring, leads to a

valuable degree of structural preorganization of substituents attached to this scaffold. This

feature might also be exploited for the presentation of pharmacophoric groups in a well-

defined arrangement.

Aims and Scope

43

2 Aims and Scope

In light of its special conformational properties, we wanted to make use of the

bicyclo[3.1.0]hexane scaffold in this Ph. D. project in two ways: First by the synthesis of a

small screening library of bicyclo[3.1.0]hexane based small molecules that would contain

various pharmacophoric groups in pre-defined orientations. The compounds should then be

submitted to biological screening in a variety of assays. As pointed out above, the

bicyclo[3.1.0]hexane scaffold has not been widely explored in drug discovery outside the

area of modified nucleosides and any hits from these screens would represent new types of

entry points into inhibitor (antagonist, agonist) development. Secondly, the effects of the

replacement of the ribose or 2’-deoxyribose moiety in S-adenosylhomocysteine and

pentostatin, respectively, on biological activity were to be investigated.

2.1 Chemical Library Synthesis

For the first objective of this research project we envisaged the construction of a chemical

library of 150-200 compounds based on a bicyclo[3.1.0]hexane scaffold with general

structures 1 and 2 (Fig. 21).

Figure 21. Target structures for the chemical library.

As discussed in the introduction, the bicyclo[3.1.0]hexane system exhibits a strong

preference for a boat-like conformation of the six-membered ring, thus leading to a significant

degree of structural preorganization of substituents attached to this scaffold. This feature

should be exploited by the attachment of a range of chemical entities as possible

pharmacophores with defined exit vectors from the core structure. These compounds should

then be made available for biological screening. The various substituent groups present in 1

and 2 were chosen according to their prevalence of occurrence in known drugs and such as

to ensure adequate physico-chemical properties.107 The heterocycles directly attached to the

bridgehead carbon atom in 2 were also selected based on their frequent occurrence in

Aims and Scope

44

biologically active nucleoside analogues108 (uracil, hypoxanthin) or a number of bioactive

natural products (oxazole, thiazole, imidazole), including anti-tumor, anti-bacterial, anti-

malarial, and immunosuppressive agents.109

Target structures 1 were envisaged to be synthesized via intermediate 3, which was to be

coupled to heterocyclic amines (ca. 10 building blocks) under standard amide coupling

conditions; reduction of the azide moiety would then be followed by coupling with a diverse

range of acids to the 7-amino group (15-20 building blocks) and global deprotection (Scheme

20).

Scheme 20. Strategy for the synthesis of chemical library target structures 1 and 2. PG = protecting

group.

Azido acid 3 was also planned to serve as a precursor for target structures 2, in the case of

C-linked heterocycles (i.e. for compounds with a C-C bond between the bridgehead C-atom

and the heterocycle). The heterocycle would be elaborated from the carboxyl group directly

Aims and Scope

45

or from a functional group that can be derived from it by simple transformations, such as,

e.g., an aldehyde or a cyano group.

In contrast, target structures 2 with N-linked heterocycles (i.e. compounds with a C-N bond

between the bridgehead C-atom and the heterocycle) were to be obtained from differently

functionalized bicyclic amines 4 or 5, depending on the specific family of target structures to

be addressed. Heterocycle construction from the bridgdehead amino group would be

followed either by selective cleavage of the protecting group on C7-O, conversion of the free

hydroxyl group into an azide substituent, and reduction (X = PGO) or by reduction only (X =

N3). Coupling of the resulting C3-O protected 7-amino intermediates with carboxylic acids

and global deprotection would then lead to target structures 2. All compounds were to be

prepared as racemates, in order to provide both enantiomers for biological testing (as

racemic mixtures).

The free hydroxyl group at position 3 that is generated in the global deprotection step could

serve as a handle for biotinylation or the attachment of fluorescent probes in compounds of

biological interest, should this be desired.

As illustrated in Scheme 21, key intermediates 3, 4 and 5 can all be derived from known

lactone 161.

Scheme 21. Retrosynthesis of key intermediates 3, 4 and 5.

Aims and Scope

46

The synthesis diverges at the stage of ester R-I, which can be elaborated into 3 by selective

deprotection of the primary 7-hydroxyl group, conversion of the free hydroxyl group into an

azide moiety, and ester saponification. Intermediate 4 is obtained by ester saponification and

subsequent Curtius rearrangement; depending on the exact conditions for the Curtius

rearrangement, the reaction may lead to a protected amino group and would thus have to be

followed by a deprotection step.

5 can be obtained from 3 by way of a Curtius rearrangement.

Intermediate R-I with PG = TBS has been described previously in the context of the

synthesis of a southern bicyclo[3.1.0]hexane-based thymidine analogue.103 The synthesis of

R-I proceeds through bromo ester R-II; treatment of the latter with base leads to

cyclopropane ring formation. R-II in turn is obtained from lactone 161, which is ultimately

derived from cis-tetrahydrophthahlic anhydride.110

2.2 Synthesis of Bicyclo[3.1.0]hexane-based Analogu es of

Pentostatin and S-Adenosylhomocysteine

A second objective of this thesis project was the synthesis of bicyclo[3.1.0]hexane-based

analogues of pentostatin (6) and of S-adenosylhomocysteine (7) (Fig. 22).

Fig. 22. Target structures pentostatin analogue 6 and (SAH) analogue 7.

As outlined in the Introduction, pentostatin is a picomolar inhibitor of adenosine deaminase

(ADA), the enzyme catalyzing the deamination of adenosine and 2’-deoxyadenosine,

respectively, to the corresponding inosines. Plasmodial ADA additionally accepts 5’-

methylthioadenosine as a substrate and the structural basis for this discrimination remained

unclear until the crystal structures of plasmodial ADA in complex with its natural substrate

adenosine, but also with guanosine, and the picomolar inhibitor pentostatin were reported by

Merritt and co-workers in 2008.57 Their data showed that in all complexes, including the one

with pentostatin, the (deoxy-)ribose ring clearly adopts a C2’-endo conformation, while

mammalian adenosine deaminase preferentially interacts with C3’-endo puckered

nucleosides.57 Thus, a southern bicyclo[3.1.0]hexane-based pentostatin analogue, in

Aims and Scope

47

principle, should preferentially inhibit plasmodial adenosine deaminase, which would enable

the selective inhibition of Plasmodium falciparum growth and survival.

The projected synthesis of 6 is outlined in Scheme 22. The synthesis would proceed through

enantiopure intermediate (+)-4 which would be reacted with imine (+)-162. Enantiopure (+)-4

can be obtained from enantiopure lactone (-)-161, which is in turn accessible by chiral

separation of an early racemic precursor (see section 3.5, Scheme 55). (+)-162 can be

prepared according to a literature procedure, starting from N-Cbz-L-vinylglycine methyl

ester.111 After addition of (+)-4 to the imine of (+)-162, ring formation to the imidazole

derivative 163 should take place upon heating. In the next step the amidine 165 would be

formed by reaction with dimethylformamide dimethyl acetal 164. Reduction of the azide and

in situ formation of the 7-membered diazepine ring should then afford compound 166. Finally,

removing all three TBS protecting groups should provide target structure 6.

Scheme 22. Envisaged synthesis of a bicyclo[3.1.0]hexane based pentostatin analogue 6.

S-Adenosylhomocysteine (SAH) is an inhibitor of various methyltransferases,112 which are

potential drug targets in cancer113 and also viral diseases.114 The corresponding 2’-deoxy

analogue S-2’-deoxyadenosyl homocysteine has been reported to be less potent, but for

some methyltransferases still shows significant inhibition.115 SAH analogue 7 serves to

investigate the question whether the loss in potency associated with the removal of the 2’-OH

group might be compensated for by the conformational fixation of the pseudosugar moiety;

Aims and Scope

48

alternatively, however, locking the sugar moiety in a 2’-endo conformation, as in 7, might

even exacerbate the loss in potency, if the enzyme preferentially interacted with the

nucleoside in a 3’-endo conformation.

Analogue 7 was envisioned to be synthesized from protected bicyclo[3.1.0]hexane based

adenosine analogue 167 (Scheme 23). The amino function of the adenine base would be

suitably protected (possibly doubly); the primary hydroxyl group would be selectively

deprotected and converted to a leaving group via an Appel reaction, a mesylation, or

tosylation.

TBSO

TBSON

N

NN

NH2

TBSO

TBSON

N

NN

NPG2

TBSO

HON

N

NN

NPG2

TBSO

TsON

N

NN

NPG2

TBSO

SN

N

NN

NPG2

BocHN

t-BuOOC

167 168

169170

172

double protectionselectivedeprotection

Appel reaction ortosylation ormesylation

SN2 reaction

HO

SN

N

NN

NH2

H2N

HOOC

7

globaldeprotection

SH

NHBoc

O

t-BuO

(+)-171

TBSO

SN

N

NN

NPG2

BocHN

HOOC

173

saponification

Scheme 23. Envisaged synthesis of bicyclo[3.1.0]hexane based S-adenosylhomocysteine analogue 7.

PG = protecting group.

The side chain would then be inserted via a nucleophilic substitution reaction with protected

homocysteine 171 to afford compound 172. Saponification and subsequent global

deprotection would furnish SAH-analogue 7.

Aims and Scope

49

2.3 Alternative Synthesis of (+)-4

A possible alternative strategy to (+)-4 is outlined in Scheme 24 and is based on

cyclopentadiene as a possible starting material (cf. section 1.6). The stereoselective

conversion of cyclopentadiene into 98 via deprotonation with NaH, quenching of the

cyclopentadienyl anion with benzyl chloromethyl ether and in situ asymmetric hydroboration

with diisopinocamphenylborane had been described.116 Directed epoxidation of 98 then leads

to intermediate R-VI.95a, 95b An epoxide opening reaction with cyanide anion would result in

compound R-V, which would then be dehydrated to R-IV. A Simmons-Smith

cyclopropanation or other types of cyclopropanation reactions should then provide R-III

which would be converted into (+)-4 by nitrile hydrolysis and Curtius rearrangement (vide

supra).

Scheme 24. Retrosynthesis of (+)-4 starting from cyclopentadiene.

Aims and Scope

50

2.4 Biological Screening of the Bicyclo[3.1.0]hexan e-based

Chemical Library

The library of compounds 1 and 2 was intended to be screened for activity against discrete

kinases and proteases; in addition, whole cell screens were foreseen, including simple

cytotoxicity testing against cancer cells, screening against Mycobacterium tuberculosis, and

the assessment of activity in a series of pathway assays with specifically manipulated cancer

cell lines. This work would be carried out in collaboration. Additionally, the library would be

made available for testing by other interested research groups.

Results and Discussions

51

3 Results and Discussions

3.1 Chemical Library Synthesis

3.1.1 Synthesis of Advanced Intermediates 3, 4 and 5

As outlined in the section 2.1, the synthesis of advanced intermediates 3, 4 and 5 was to

proceed through lactone 161, for which a synthesis had been described in the literature

starting from cis-tetrahydrophthalic anhydride (174).117 The synthesis of 161 is summarized

in Scheme 25. Since 3, 4 and 5 were needed in gram quantities, the synthesis of 161 was

performed on the largest scale possible with conventional laboratory equipment; this involved

starting with approximately 100 g of 174 and the synthesis was carried out multiple times.

The opening of anhydride 174 with MeOH in the presence of catalytic amounts of p-toluene

sulphonic acid worked well to afford a racemic half ester 175 in quantitative yield. Selective

reduction of the acid moiety was possible through conversion to the acid chloride with oxalyl

chloride followed by treatment with NaBH4 to produce alcohol 176. As has been shown by

others, this reaction, when carefully controlled, proceeds without epimerization.117a Alcohol

176 was not isolated, but directly treated with acid to induce cyclization to lactone 177. The

latter was obtained in an excellent 93% yield from 175. Oxidative cleavage of the double

bond resulted in diacid 178, which could be isolated in 73% yield. Treatment of 178 with

MeOH/sulfuric acid not only produced the expected diester 179a, but also its rearranged

isomer 179b. Fortunately, both isomers 179a and 179b upon treatment with tert-BuOK

produced only a single Dieckmann cyclization product, namely enol 180. However, the

outcome of the Dieckmann cyclization depended to a great extent on a very careful work-up.

The formation of a brown residue at the bottom of the flask during quenching remained

undetected despite temperature monitoring in the first trials, as quenching resulted (upon

formation of KCl) in a thick suspension. The formation of this brown residue, probably the

result of local overheating of the mixture, could be avoided by intense mechanical stirring

and a very slow addition of the quenching acid. Thus, the desired product could be isolated

after work-up by recrystallization as colorless crystals in a yield of 72%. Assuming first the

water content of the used sulphuric acid to be problematic, we had switched to the use of dry

hydrochloric acid, but this had not given better results. The enolate double bond was next

reduced with sodium borohydride, which gave hydroxy ester 181 selectively (73%), with a

trans arrangement of the ester and the hydroxyl group. The selectivity of the reaction is the

result of the attack of the reducing agent from the exo side of the bicylic ring system in 180,

Results and Discussions

52

and the ensuing enolate reacting with a proton from the endo side, as the resulting trans

arrangement of the two ring substituents is preferred for reasons of diminishing the

unfavourable 1,2-interaction. Ester saponification followed by borane reduction then

produced the desired lactone 161 in 58% yield for the two steps from hydroxy ester 181.

Lactone 161 was prepared in multigram quantities.

Scheme 25. i. K2CO3, MeOH, 100%. ii.1) (COCl)2, CH2Cl2, 0°C-->rt, 12h. 2) NaBH 4, EtOH, -40°C, 2h.

iii. CSA, tol, rf, 3h, 93% (over 3 steps). iv. KMnO4, H2O, Adogen 464, 5°C, 12h, 73%. v. H 2SO4, MeOH,

rf, 2d, 86%. vi. tert-BuOK, tol, 0°C-->rt, 22h, 72%. vii. NaBH 4, MeOH, -70°C, 2h, 73%. viii. KOH,

H2O/THF, rt, 12h, 83%. ix. BH3·Me2S, Me3BH, 5°C--> 15°C, 1h, 83%.

With lactone 161 in hand, the next phase of the synthesis then called for the elaboration of

this central intermediate into ester 185, which serves as a common precursor for both azido

acid 3 and amine 4. The ultimate optimized three-step sequence is shown in Scheme 26 and

involves lactone opening with trimethylsilylbromide in MeOH in the presence of catalytic

amounts of ZnBr2, to produce bromo ester 183, protection of the free hydroxyl groups as TBS

ethers using MTBSA,118 and finally ring closure by treatment with base.103 The overall yield of

Results and Discussions

53

this sequence varied between 13% and 29%, with the most reliable step being the formation

of the cyclopropane ring. Quite remarkably, this step could be achieved in yields of up to

94%.

Bromide 184 proved to be unstable above 30°C and had the tende ncy to re-form the lactone

ring. For this reason, the best yields achieved for the lactone opening were around 50%.

Likewise, the simultaneous protection of the two hydroxyl functions gave moderate yields

only (between 33% and 58%).

Scheme 26. i. TMSBr, MeOH, ZnBr2 (cat.), rt, 25h, 53%. ii. MTBSA, CH2Cl2, rt, 4h, 58%. iii. tert-BuOK,

tert-BuOH, rt, 1h, 94%.

Different reagents were tested for the introduction of the tert-butyl dimethylsilyl protecting

groups and the best proved to be MTBSA (Table 1); MTBSA can be readily prepared from N-

methylacetamide and TBSCl.118

Table 1. Reagents and conditions of the TBS protection of 183 to 184.

Reagents/conditions yield

TBDMSCl, imidazole, DMF, 0°C, 12h 0%

TBDMSOTf, 2,6-lutidine, CH2Cl2, -78°C, 2h 33%

MTBSA, CH2Cl2, rt, 4h 33-58%

Other reagents tested included TBSOTf and TIPDSCl2, the latter leading to a TIPDS-

protection of the two hydroxyl groups in a cyclic fashion.119 TIPDSCl2 was reacted with diol

161 before the lactone opening step. Due to the low yield obtained 186 was not subjected to

the subsequent lactone opening with trimethylsilylbromide (Scheme 27).

Results and Discussions

54

Scheme 27. i. 1) AgNO3, py, THF, 1h. 2) TIPDSCl2, rt, 8h, 35%.

However, the TIPDS-protecting group would have required an exchange to a different set of

protecting groups at a later stage of the synthesis, as it does not allow selective deprotection

of the primary hydroxyl group, which was required in order to proceed with the synthesis in

the projected way, and also the yield of the TIPDS protection step was rather low (35%).

The tert-butyl dimethylsilyl group was thus our first choice for the protection of both hydroxyl

groups in 183, as it should survive the conditions required for subsequent transformations; in

addition, a primary TBS ether can be selectively cleaved over a secondary one under

relatively mild acidic conditions.120 Nevertheless, we also contemplated the use of the benzyl

ether protecting group, which can be cleaved reductively under neutral conditions121 and

would have been stable to the conditions needed for the remaining functionalizations. Most

importantly, we assumed that benzyl ethers would enhance the stability of the bromo ester

and (in contrast to the TBS ethers) would be stable under the acid conditions employed for

lactone opening. This led us to attempt protecting the two hydroxyl functionalities of 161 as

benzyl ethers before lactone opening with MeOH/TMSBr. Unfortunately, the reaction of 161

with sodium hydride, benzyl bromide and tetrabutylammonium iodide as a catalyst gave low

yields of the monobenzylated product at best. This is surprising as this transformation has

been reported in literature to proceed in 54% yield122, a result that could simply not be

reproduced here.

Scheme 28. i. TMSCl, MeOH, ZnCl2, rt, 24h, 62%. ii. MTBSA, CH2Cl2, rt, 16h, 62%. iii. tert-BuOK, tert-

BuOH, rt, 2h, 64%.

In an attempt to further improve the moderate yields in the steps from 161 to 185, we also

prepared the chloro derivative 187 by opening lactone 161 with TMSCl (Scheme 28).123 Due

to the higher stability of compound 187, it could be isolated in a somewhat higher yield of

Results and Discussions

55

62%, in comparison with 53% for bromide 183. Likewise, chloride 187 could also be TBS-

protected in higher yields (61-100%) than 183 (Scheme 35). However, the yield in the

subsequent cyclopropanation reaction was only 64%, compared to 94% in the case of the

bromide 184. These findings added up to approximately the same yields over the three

steps; as the reaction sequence from 161 to 189 via bromide 184, in spite of the lower

stability of the bromide was more reproducible, this approach was routinely used for the

preparation of 185.

The conversion of ester 185 into the key intermediate for the preparation of target structures

1, i.e. 3, proceeded uneventful. As illustrated in Scheme 29, this involved cleavage of the

primary TBS ether followed by conversion of the ensuing free hydroxyl group into a mesylate,

displacement of the mesyloxy group with azide anion, and finally ester cleavage with TMSOK

in THF at 80°C. 3 was thus obtained in 45% overall yield for the 4 steps from ester 185.

Scheme 29. i. CSA, CH2Cl2/MeOH 1:1, 0°C, 2.5h, 67%. ii. MsCl, Et 3N, Et2O, 0°C, 3h, 96%. iii. NaN 3,

DMF, 70°C, 20h, 76%. iv. TMSOK, THF, 80°C,))), 2h, 92%. [))) = microwave irradiation]

In principle, azido acid 3 could serve as the precursor for the preparation of intermediate 5

(with PG = TBS). However, for reasons that will be explained in section 3.1.3.1.1 subsequent

transformations mandated a C3-O-protecting group that would be stable under acidic

conditions, a condition which is not fulfilled for a TBS protecting group. 5 was thus prepared

with a benzyl protecting group on C3-O; to this end, ester 192 was converted into C3-O

benzyl-protected azido acid 194 by TBS-removal with TBAF, followed by treatment with

BnBr/NaH and cleavage of the methyl ester moiety with TMSOK in 75% overall yield for the

3-step sequence from ester 192 (Scheme 30). Acid 194 was then transformed into the

desired amine 5 by acyl azide formation and in situ Curtius rearrangement.124 The ensuing

isocyanate was quenched with aqueous sodium hydroxide, thus affording directly the primary

amine. This approach avoids the removal of an amine protecting group, which could have

Results and Discussions

56

been problematic in the presence of a benzyl group, but it has the disadvantage that the

amine 5 had to be used to the next step immediately after its preparation, due to its limited

stability.

Scheme 30. i.TBAF, THF, rt, 1h, 90%. ii.NaH, BnBr, DMF, -15°C-->rt, 1.5h, 92%. iii. TMSOK,

THF/DMF 4:1, ))), 90°C, 4h, 91%. iv. 1) DPPA, Et 3N, tol, 0°C-->rt-->80°C, 12h. 2) 2 N NaOH, THF,

30 min, 82%.

For the elaboration of 185 into our second key intermediate 4 the methyl ester moiety was

cleaved with KOH/EtOH to produce acid 195 in 90% yield. Acid 195 was then converted into

the corresponding acyl azide with DPPA; the azide was submitted to an in situ Curtius

rearrangement124a and the resulting isocyanate was directly reacted with benzyl alcohol to

give the Cbz-protected amine 196 in excellent yield (88% based on 195).

Scheme 31. i. KOH, EtOH, 100°C, ))), 2.5h, 90%. ii. 1) DPPA, Et3N, tol, 0°C--> 80°C, 2h. 2) BnOH, di-

n-butyltindilaurate (cat.), 80°C, 2.5h, 88%. iii. H 2 (1 bar), 10% Pd/C, tol/MeOH 1:1, rt, 4h, 88%.

The benzyloxycarbonyl (Cbz-) protecting group could be smoothly removed from 196 by

catalytic hydrogenation over Pd/C to furnish 4 in 88% yield. An attempt to convert the

isocyanate directly to primary amine 4 by quenching the reaction with water resulted in a

40% yield only. The route involving conversion of the isocyanate to the Cbz-protected amine

196 was thus preferable. It also allowed the amine to be stored in protected form; removal of

the Cbz protecting group in general was only performed immediately before use of the free

amine 4.

Results and Discussions

57

As illustrated in Fig. 22, the Curtius rearrangement125 produces isocyanates upon thermal

decomposition of carboxylic azides, which can be synthesized directly from the acid with

DPPA,126 or via acid chlorides or mixed anhydrides as intermediates.

Figure 22. Conversion of carboxylic acids into amines via Curtius rearrangement.

The nitrene species formed in the thermal decomposition of the acyl azide then undergoes

rearrangement, such that the alkyl group migrates to the nitrogen atom, most likely in a

concerted process (Fig. 23).127 The reaction can either be quenched with water, which gives

primary amines, or with alcohols, which yields carbamates; quenching with amines affords

urea derivatives.

Figure 23. Mechanism of the Curtius rearrangement.

Results and Discussions

58

3.1.2 Synthesis of Library Members with General Str ucture 1

3.1.2.1 Target Structures Incorporating Heterocycli c Amines

As illustrated in Scheme 32 the general strategy of the preparation of library members of

general structure 1 involved (i) coupling of acid 3 with the desired amine; (ii) reduction of the

azide moiety to a primary amino group; (iii) coupling of each individual amine with a set of

carboxylic acids; and (iv) cleavage of the TBS ether at position 3 of the bicyclo[3.1.0[hexane

skeleton.

Scheme 32. General strategy for the preparation of library members with general structure 1.

Scheme 33 summarizes the coupling of 3 with 5 different amines 197-201. Activation of 3

was generally carried out with HATU128 and couplings proceeded in yields of 80-100% to

provide amides 202a-e. No other coupling methods were investigated in light of the excellent

product yields obtained with HATU. Catalytic hydrogenation of 202a-e over 10% palladium

on carbon resulted in clean conversion of the azido moiety into a primary amino group to

furnish amines 203a-e in 83 to 100% yield. The reduction products were generally obtained

in purities >85% (based on NMR) and employed in the next coupling step without

chromatographic purification (in view of the good purity of the crude products and to avoid

loss of material upon flash chromatography of these very polar compounds).

Results and Discussions

59

3

COOH

TBSO

N3

TBSO

N3

O

N

202d

TBSO

N3

O

N

N

OH

HN

N

OH

N

S

NH

TBSO

N3

O

TBSO

N3

O

N

N

OMe

TBSO

N3

O

Cl

HN

OMe

HN

Cl

HN

N

S

H2N

202a

202b

202c

202e

i

197a

198b

199c

200d

201e

TBSO

H2N

O

N

203d

TBSO

H2N

O

N

N

OH

N

S

NH

TBSO

H2N

O

TBSO

H2N

O

N

N

OMe

TBSO

H2N

O

203a

203b

203c

203e

ii

i

i

i

i

ii

ii

ii

ii

Scheme 33. i. HATU, DIPEA, amine, rt, 22h, 202a: 83%, 202b: 96%, 202c: 100%, 202d: 94%, 202e:

90%. ii. H2, Pd/C, Tol/MeOH 1:1, rt, 2-6h, 203a: 100%, 203b: 100%, 203c: 91%, 203d: 100%, 203e:

87%.

In the case of azido amide 202d the reduction of the azido group, not unsurprisingly, was

accompanied by reductive removal of the chloro substituent on the aromatic ring. The

reduction of the azido functionality of 202d was therefore carried out under Staudinger

conditions, i. e. by treatment of 202d with polymer-bound Ph3P and hydrolysis of the ensuing

aza-phosphorane129, which gave the desired amino amide 203f in 67% crude yield (Scheme

34).

Results and Discussions

60

Scheme 34. i. 1) PPh3 polymer-bound, dioxane, rt, 2h. 2) NH3 (32% in H2O), rt, 12h, 67%.

As shown in Scheme 33, the different amines (including the dehalogenated reduction product

203d), where then submitted to coupling with a set of up to 20 diverse carboxylic acids,

including 5 desamino-(natural) amino acids (Scheme 35).

Scheme 35. Structures of acids used in the coupling steps.

As for the coupling of acid 3 with amines 197-201, the different acids were generally

preactivated with HATU for 30 min; the coupling products were obtained in yields varying

between 45 and 100% after extractive work-up and in most cases the crude diamides were

directly submitted to deprotection. It was aimed at coupling all acids to all prepared amines;

however, coupling with acids 204h, 204l, 204m and 204s tended to give low yields by trend,

independent of the kind of amine they were coupled to. Coupling of these acids was then

avoided in the last coupling series, the one to amine 203e, as these derivatives already

contain an amino function that could possibly act as a hydrogen bond acceptor in the

heterocycle. Acid 204c also gave low yields in all the couplings, and acid 204i gave a low

yield in the coupling with 203b.

Results and Discussions

61

The derivatization of the 7-amino group and the subsequent deprotection step are

exemplified in Scheme 36 for amine 203d and 5 different carboxylic acids, spanning a range

of coupling yields.

Scheme 36. i. R-COOH, HATU, DIPEA, CH2Cl2, rt, 12h.ii. Dowex 50W X8, MeOH, 12h, 1db : 32%

(over 2 steps), 1dg : 55% (over 2 steps), 1dc : 26% (over 2 steps), 1de: 40% (over 2 steps), 1do : 35%

(over 2 steps).

Different conditions were investigated for the final TBS deprotection steps, including HF·py,

TBAF, and Dowex-50W-8X cation exchange resin. Deprotection was found to be most

efficient with Dowex 50W-X8,130 which gave the best yields of products 1d; in addition the

use of an insoluble reagent also proved to be advantageous technically, as it could be

removed by filtration and no other work-up was required.

All final products 1 were purified by flash column chromatography and characterized by high

resolution mass spectrometry; in addition, approximately one third of the library was fully

characterized by 1H and 13C NMR spectroscopy. The purity of all compounds was assessed

by RP-HPLC on a C18 column and for the majority was found to be ≥85%, based on

integration of peaks in the HPLC chromatogram obtained at 220 nm. Figure 24 shows the

Results and Discussions

62

HPLC diagram of 1do as a representative example. The purity of this compound was

assessed to be ≥94% according to the integration of its HPLC trace.

Figure 24. HPLC chromatogram of 1do . The integration assigns 1do a purity of ≥94%.

It should be noted here that HPLC purity determined this way may not reflect the true purity

of a compound, as impurities may have higher extinction coefficients than the desired

product.

For compound 1do , which combines amine 203d and acid 204o, an x-crystal structure was

obtained, which confirmed the expected relative stereochemistry (and thus also the relative

configuration of key intermediate 3, which is the precursor for all library members 1).This

structure will be discussed in detail in section 3.4.

3.1.2.2 N-(2-((4-Methoxyphenyl)amino)ethyl)amides

N-(2-((4-Methoxyphenyl)amino)ethyl)amides of modified small peptides, such as 206 have

recently been shown to be potent reversible inhibitors of cysteine proteases.131

Results and Discussions

63

As we had planned for our library to be screened against different proteases (given the

“peptide-like” structure of diamides of type 1), we have also attempted to prepare a small

library of N-(2-((4-methoxyphenyl)amino)ethyl)amides with different carboxylic acid residues

attached to N-7. Due to a lack of intermediate 3 at this stage of the project, the synthesis of

N-(2-((4-Methoxyphenyl)amino)ethyl)amides 1g was attempted from acid 194 (Scheme 30),

which bears a benzyl protecting group on C3-O and had been prepared in the context of the

synthesis of key intermediate 5.

Activation of acid 194 with HATU allowed coupling with N1-(4-methoxyphenyl)ethane-1,2-

diamine (207) to produce amide 208 in 73%. In order to avoid competing reduction of the

benzyl moiety the subsequent reduction of the azide was conducted by catalytic

hydrogenation over Lindlar catalyst. The reduction provided amine 209 in almost quantitiative

yield, in spite of the fact that the reaction times were significantly longer than those observed

for the reduction of uracil azide 213 employing Lindlar catalyst (see also section 3.1.3.1.1),

possibly due to partial catalyst poisoning by the ethylene diamine moiety.

Scheme 37. ii. HATU, DIPEA, CH2Cl2, rt, 22h, 73%. iii. H2 (6 bar), 5% Pd/CaCO3, rt, 12h, 98%.

The coupling of all acids 204a, 204b, 204c, 204d, 204e, 204f, 204g, 204i, 204k, 204l, 204n,

204o, 204p, 204q and 204r (Scheme 35, acids 204h,j,m,s,t were not used due to low yields

obtained with these acids in earlier couplings) to amine 209 proceeded in excellent yields

again employing HATU activation (Scheme 38).

Results and Discussions

64

Scheme 38. i. R-COOH, HATU, DIPEA, CH2Cl2, rt, 12h, 92-100%.

The critical step in the synthesis of N-(2-((4-methoxyphenyl)amino)ethyl)amides 1g proved to

be the cleavage of the benzyl ether moiety at position 3 of the bicyclic core structure from

intermediates 209. Different conditions were investigated for this transformation; as

compounds 210a-210t were only available in milligram quantities, the trial reactions were

performed only on a very small scale, therefore some of the reactions were solely monitored

by mass spectrometry. Otherwise thin layer chromatography (TLC) was employed.

Treatment of compound 210c with Pd-C and formic acid as the hydrogen source in MeOH

gave some product formation according to TLC and mass spectrometry, but substantial

amounts of starting material were still left after 5 h and decomposition was visible upon TLC

analysis by the formation of several spots (Scheme 39 and Table 2). If the reaction was run

longer, the product spot could not be detected anymore.

Results and Discussions

65

Scheme 39. Debenzylation trials with selected benzyl ethers 210g.

Increasing the catalyst load (with 210a as substrate) led to decomposition only. Attempted

debenzylation of 210b with molecular hydrogen over Pd-C gave recovered starting material

only or, at high catalyst loads, a product that appeared to be overreduced (desired mass plus

6 mass units). In an attempt to avoid overreduction, 210p was treated with H2 at 8 bar over

Lindlar catalyst, but no reaction was observed. The use of Raney-Nickel instead of Pd-C led

to complete decomposition of the starting material 210b (Table 2).

Treatment of 210b with Lewis acids, such as iron trichloride or tin tetrachloride either resulted

in decomposition (FeCl3) or returned unchanged starting material. In contrast, boron

trichloride provided the desired deprotected compound 1gp (Scheme 39). In light of these

results no further efforts were made to debenzylate intermediates 210; instead these

compounds were directly used in different screening campaigns.

Results and Discussions

66

Table 2 . Debenzylation trials with benzyl ethers 210fa-p a

Cpd. Conditions Outcome

210gc HCOOH, Pd/C (1 eq.), MeOH, RT, 3h traces of prod.

210gc HCOOH, Pd/C (1 eq.), MeOH, RT, 5 h 35% (+ 32% of

start.mat.)

210ga HCOOH, Pd/C (5 eq.), MeOH, RT, 5 h decomp.

210gb H2 (5 bar), Pd/C (10%), RT, 24 h SM

210gb H2 (2 bar), Pd/C (1 eq.), EtOH, RT, 5 h SM

210gb H2 (1 bar), Pd/C (5 eq.), EtOH, RT, 15min M+6 (overred.)

210gb H2 (2 bar), Pd/C (10 eq.), EtOH, RT, 1 h M+6 (overred.)

210fb H2, Raney-Nickel, EtOH, RT, 4 h decomp.

210gb FeCl3, CH2Cl2, RT, 2 h decomp.

210gb SnCl4, CH2Cl2, RT, 2 h SM

210gp H2 (8 bar), Lindlar's cat. (3 eq), EtOH, RT, 24h SM

210gp BCl3, CH2Cl2, -78°C, 4 h desired prod.

aFor structures see Scheme 39. Reaction mixtures were analyzed by MS (ESI). Cpd.: compound, SM: starting

material, prod.: product, decomp.: decomposition, overred.: overreduction.

3.1.3 Synthesis of Library Members with General Str ucture 2

3.1.3.1 N-linked Heterocycles

3.1.3.1.1 Uracil Derivatives

Given the importance of uracil as a nucleobase, but also due to its occurrence in biologically

active synthetic nucleoside analogs132 and naturally occurring secondary metabolites, uracil

was chosen as one of the priority heterocycles in the construction of sublibraries of type 2.

The elaboration of amines into N1-substituted uracil derivatives is well established; however,

as will become apparent below (Scheme 40), the conditions required for final closure are

incompatible with the stability requirements for TBS protecting groups. As a consequence,

Results and Discussions

67

the use of key intermediate 5 was deemed most appropriate for the construction of this sub-

library, as loss of the protecting group on C3-O might have led to side reactions in the crucial

coupling step by competing acylation of the free hydroxyl group. Build-up of the uracil base

from amine 5 was accomplished in a two- step sequence that involved first reaction with acyl

isocyanate 211 and subsequent ring closure of the resulting N-acyl urea 212 under acidic

conditions in a maximum 65% overall yield. Azide 213a was then converted into amine 214a

in quantitative yield by catalytic hydrogenation; in order to avoid competing reduction of the

double bond of the uracil moiety, the reduction was carried out with Lindlar catalyst under a

hydrogen pressure of 3 bar. Amine 214a was sufficiently pure as judged by TLC analysis to

be used in the following coupling reactions without further purification.

Scheme 40. i. CH2Cl2, 0°C-->rt, 12h, 77%. ii. H 2SO4 1 M aq., dioxane, rf, 2h, 84%. iii. H2 (3 bar),

Lindlar cat. (5% Pd/CaCO3), EtOH, 4h, 100%.

Acyl-isocyanate 211 was obtained from commercially available methyl 3-methoxyacryloate

215 in three steps according to Scheme 41.133

Scheme 41. i. KOH, H2O, 50°C, 12h, 67%. ii. SOCl 2, CH2Cl2, rf, 3h, 74%. iii. AgOCN, tol, rf, 30min.

Results and Discussions

68

HATU-mediated coupling of amine 214 with carboxylic acids 204a-204r (for structures see

Scheme 35) in dichloromethane proceeded smoothly and afforded the desired amides

218a-r in yields between 47-100% (Scheme 42).

Scheme 42. i. R-COOH, HATU, DIPEA, CH2Cl2, rt, 12h, 47-100%.

Unfortunately, the clean removal of the benzyl protecting group on C3-O of compounds 218a

by catalytic hydrogenation proved to be impossible under a variety of conditions, due to

competing reduction of the double bond in the uracil base. As illustrated in Scheme 43 for

amide 218c, cleavage of the benzyl ether was feasible with boron trichloride according to

mass spectrometry and TLC analysis.

Scheme 43. i. BCl3, CH2Cl2, -78°C, 5h, yield not determined.

In light of the difficulties associated with the cleavage of the benzy ether moiety in amides

218, no further efforts were made to prepare the corresponding C3-hydroxyl derivatives.

Rather, the benzyl ethers as such were used for screening purposes.

3.1.3.1.2 Hypoxanthine Derivatives

In contrast to the uracil derivatives described above, intermediate 4 was employed as the

amine precursor for the construction of hypoxanthine derivatives of type 2. The elaboration of

the hypoxanthine base from a substituted amine does not involve any acid-promoted

transformations134 and, therefore, is fully compatible with the protection of the 3- and 7-

hydroxyl groups as TBS ethers.

Results and Discussions

69

The assembly of the hypoxanthine heterocycle started with the reaction of amine 4 with N-

(4,6-dichloropyrimidin-5-yl)formamide,135 which was followed by the in situ cyclization of the

resulting diamino-pyrimidine 220 with diethoxymethyl acetate under heating, to provide

chloropurine derivative 221 (Scheme 44). Unfortunately, the yield for this two-step

transformation did not exceed 45%; as a consequence only small quantities of 221 could be

prepared. Treatment of 221 with of 2-mercaptoethanol and sodium methoxide in MeOH gave

protected hypoxanthine derivative 222.

TBSO

TBSON

N

N N

Cl

NH2

TBSO

TBSO

HN

TBSO

TBSO

N N

NH

Cl

O

H

N

NHN

Cl

Cl

O

H

TBSO

TBSON

N

NN

OH

i

ii iii

219

220

221 222

4

Scheme 44. i. 219, Et3N, 1,4-dioxane, rf, 20h. ii. diethoxymethyl acetate (neat), 110°C, 20h, 45% (over

2 steps). iii. NaOMe, 2-mercaptoethanol, MeOH, ))), 80°C, 2h, 61%.

The mechanism for the conversion of 221 into 222 (Scheme 45) presumably involves

displacement of the 6-chloro substituent by the thiolate anion derived from 2-

mercaptoethanol to produce 223.134 The resulting 6-(2-hydroxyethyl)-purine anion 224 then

rearranges to produce 227. The latter eliminates episulfide, thus, finally leading to

hypoxanthine 222.

Results and Discussions

70

Scheme 45. Proposed mechanism for the conversion of 6-chloropurine 221 into hypoxanthine 222.

The protected hypoxanthine derivative 222 could be successfully elaborated into azide 231

by selective deprotection of the primary hydroxyl group with camphorsulphonic acid, followed

by mesylation and mesylate displacement with azide anion (Scheme 46). 231 was obtained

in 15% overall yield for the 3-step sequence from 222. Unfortunately, only small quantities of

231 could be prepared, due to a lack of material for key precursors at this stage of the

project. The synthesis of hypoxanthine-based target structures of type 2 was thus

abandoned.

Scheme 46. i. CSA, MeOH/CH2Cl2 1:1, 0°C, 5h, 81%. ii. MsCl, Et 3N, CH2Cl2, rt, 5h, 45%. iii. NaN3,

DMF, 70°C, 23h, 42%.

Results and Discussions

71

3.1.3.2 C-linked Heterocycles

Due to their known occurrence in biologically active synthetic molecules and natural

products,109 oxazole, (N-substituted) imidazole, thiazole, and tetrazole (or their partially

reduced congeners) were also of interest as heterocycle components of library structures of

type 2. While N-alkylation of tetrazole and (unsubstituted) imidazole is possible without the

formation of quaternary salts, oxazole (oxazoline) and thiazole (thiazoline) cannot be

connected to an alkyl residue by a C-N bond, except in quaternary salts (which were not of

interest here). As a consequence, our attempts to produce bicyclo[3.1.0]hexane derivatives

of oxazole (oxazoline) and thiazole (thiazoline), but also of imidazole (imidazoline) and

tetrazole all departed from acid 3 or ester 185, whose carbonyl carbon atom would become

part of the desired heterocycle. In the following, these attempts are briefly summarized.

Our first attempt at the synthesis of an oxazole derivative involved the reaction of methyl

ester 185 with methyl isocyanide in the presence of n-butyl lithium136, as the shortest

conceivable route (Scheme 47). The synthesis of oxazoles by the reaction of esters with in

situ prepared lithiomethyl isocyanide has been used by Jacobi and co-workers136 in their

synthesis of (+)-gnididione; unfortunately, we did not obtain any of the desired product 232

using this methodology. As an alternative, we have also investigated the reaction of aldehyde

234 (obtained from ester 185 by DIBAL-H reduction and subsequent PCC oxidation of the

resulting alcohol 233) with tosylmethyl isocyanide.137 While this approach did indeed deliver

the desired product 235, the yield of the reaction did not exceed 16%. As such this process

was not sufficiently effective to serve as the basis of the construction of an oxazole-based

sublibrary of type 2.

With aldehyde 234 at our disposal, we also investigated its conversion into imidazole

derivative 236 with glyoxal in aqueous ammonia solution.138 The reaction failed to produce

any of the desired product and was not investigated further.

Results and Discussions

72

Scheme 47. i. n-BuLi, CH3NC, THF, -78°C--> 0°C, 3h. ii. DIBALH, CH 2Cl2, -78°C, 4h, 100%. iii. PCC,

CH2Cl2, rt, 5h, 91%. iv. TosMIC, K2CO3, MeOH, rf, 4h, 16%. v.glyoxal, NH3 25% aq.,EtOH, rt, 12h.

As oxazoles can be prepared from N-(2-hydroxyethyl)amides via cyclization to the

corresponding oxazolines and subsequent oxidation,139 we prepared the N-(2-hydroxyethyl)-

amide of acid 3, i. e. 237, by coupling with 2-aminoethanol (Scheme 48) to give compound

237. While attempts to convert 237 into oxazoline 238 with mesyl chloride, thionyl chloride,140

or diisopropylcarbodiimide and copper triflate,141 failed in our hands, 238 could finally be

obtained by treatment of 237 with Burgess reagent at 120°C in a microwave reacto r in

excellent yield (89%).142

Scheme 48. i. DIPEA, HATU, ethanolamine, CH2Cl2, rt, 2h, 67%. ii. Burgess reagent, THF, 120°C, ))) ,

1h, 89%. iii. 1) Lawesson’s rgt, tol, 120°C, ))), 2 h. 2) Et3N, tol, rt, 1h. iv. NiO2, benzene, rf, 3d, 16%.

While the oxidation of oxazolines to oxazoles in principle can be achieved with nickel oxide,42

manganese dioxide,143 or treatment with N-bromosuccinimide and AIBN, followed by a

halogen-metal exchange with butyl lithium and quenching with water,144 these methods work

well only for oxazolines bearing electron-withdrawing substituents at the 4- or 5-position. The

most promising oxidant for the conversion of an oxazolines such as 238 into an

unsubstituted oxazole appeared to be nickel oxide;139, 145 however, even this reagent was

able to produce oxazole 239 only in an unsatisfactory yield of 16%. Thus, as in the case of

oxazole 235, this process did not provide a basis for the synthesis of a sublibrary of oxazole-

containing structures of type 2.

Results and Discussions

73

N-(2-Hydroxyethyl)amide 237 was also investigated as a possible starting material for the

synthesis of 2-substituted thiazolines and, ultimately, thiazoles. According to Nishio et al., the

conversion of 2-hydroxyethylamides to 2-mercaptoethylthioamides should be feasible with

Lawesson reagent; upon treatment with triethylamine in toluene the latter would then yield

the corresponding thiazolines.146 In our hands, this reaction did not work for N-(2-

hydroxyethyl)amide 237 and thiazoline 240 was not obtained.

While oxazoles 235 and 239 were not obtained in sufficient quantities to consider their

elaboration into the desired amides of type 2, oxazoline N-(2-hydroxyethyl)amide 238 was

reduced to amine 241, which was subsequently coupled with a limited number of carboxylic

acids (Scheme 49). The reduction of the azide moiety with hydrogen over Pd-C proceeded in

excellent yield; the subsequent coupling step gave amides 242f-o in yields between 38% and

85%.

Scheme 49. i. H2 (1 bar), 10% Pd/C, Tol/MeOH 1:1, rt, 2h, 98%. ii. R-COOH, DIPEA, HATU, CH2Cl2,

rt, 12h, 38-82%.

Unfortunately, the deprotection of 242f,g,m,n,o under our previously developed standard

conditions (Dowex 50W-X8, MeOH) gave only low to moderate yields of the desired target

structures 2 (Scheme 50), thus providing only 0.5-1 mg amounts of material. Most likely,

these yields reflect the limited stability of the oxazoline ring under acidic conditions.147

Results and Discussions

74

HO

NH O

N

TBSO

NH O

N

R

O iO

NH

F

F

O

HO

NH O

N

N

SO

O O

HO

NH O

N

242f,g,m,n,o2m

2g

2f

OO

O

O HO

NH O

N

ON

NO

HO

NH O

N

2n

2p

Scheme 50. i. Dowex 50W-X8, MeOH, rt, 48h,2f: 38%,2g: 44%, 2m: 13%, 2n:13%, 2o: 21%.

Results and Discussions

75

3.1.4 Final Library

Figure 25 shows one representative example for each of the bicyclo[3.1.0]hexane-based

sublibraries that have been prepared in the course of this project together with the number of

compounds in each sublibrary. In total, 110 compounds could be prepared in quantities that

were useful for screening purposes.

Figure 25. Structure and size of individual bicyclo[3.1.0]hexane-based sublibraries of types 1 and 2.

Single representatives are shown for each sublibrary.

Results and Discussions

76

3.2 Synthesis of Bicyclo[3.1.0]hexane-based 5-Alkyn yl-

deoxypyrimidine Nucleoside Analogue 243

In 2005 Rai and co-workers showed that 5-(1-alkynyl)-2’-deoxy uridine derivatives can have

significant in vitro antimycobacterial activity,148 with MIC90 values against Mycobacterium

bovis in the 10-100 µg/mL range. The activity was found to depend on the size of the 1-

alkynyl substituent and was postulated to be based on the interference with mycobacterial

DNA and/or RNA synthesis.

As 2’-deoxyribonucleosides preferentially adopt a 2’-endo (southern) sugar conformation,

which essentially corresponds to the conformation of the bicyclo[3.1.0]hexane moiety in

bicyclo[3.1.0]hexane-based 2’-deoxyribonucleoside analogues with the base attached to

position 1 of the bicyclic core, we have prepared analog 243 as a potential antimycobacterial

agent. The MIC90 of the corresponding 2’-deoxyribonucleoside 244 against M. bovis is

50 µg/mL.

Figure 26. 5-alkynynl-deoxypyrimidine nucleoside 244 and bicyclo[3.1.0]hexane based analogue 243.

The synthesis of 243 proceeded via 2’-deoxy uridine analog 246, which was obtained from

amine 4 and acyl isocyanate 204 in 2 steps and 71% overall yield (Scheme 51; see also

section 3.1.3.1.1). Compound 245 was cyclized to uracil derivative 246 by heating with

sulfuric acid with concomitant cleavage of both TBS protecting groups.

Results and Discussions

77

Scheme 51. i. 204, CH2Cl2, -15°C →rt, 12h, 95%. ii. H2SO4 1 M aq., dioxane, rf, 2h,75%.iii. I2, CAN,

AcOH, 80°C, 4h, 60%. iv. Pd(PPh 3)4, CuI, DIPEA, 1-dodecyne, rt, 12h, 68%.

246 was then iodinated with cerium(IV)ammonium nitrate (CAN) and iodine to produce 247;

the latter underwent smooth Sonogashira coupling with 1-dodecyne to furnish target

compound 243 in 68% yield.

3.3 Miscellaneous Derivatives

3.3.1 Tetrazole 250

As part of our work on target structures 2 with C-linked heterocycles, we have also attempted

to synthesize tetrazole derivative 250 (Scheme 52); the tetrazole ring is an important

pharmacophore, which is often used as a non-classical bioisostere for a carboxyl group.149 In

order to access 250, acid 195 was elaborated into nitrile 249 in a procedure which involved

activation of the carboxylate function as an acid chloride and subsequent amide formation

with ammonia to afford 248 which was isolated by extraction but not purified. Dehydration of

amide 248 was accomplished by treatment with oxalyl chloride/DMF in THF.150 The latter

step also led to loss of the TBS protecting groups.

Results and Discussions

78

Scheme 52. i. 1) SOCl2, THF, rf, 2h. 2) NH3 aq. ii. (COCl)2, DMF, THF, 0°C, 10min, >100%.

Nitrile 249 was difficult to purify, mainly due to its volatility. In light of these experimental

difficulties work on target structure 250 was abandoned.

3.3.2 Iodides 238 and 239

Bridgehead iodides 251 and 252 were prepared as possible precursors for transition metal-

catalyzed cross-coupling reactions. The syntheses were based on the Hundsdiecker-type

oxidative iododecarboxylation of acids 195 and 3 employing iodosobenzene diacetate and

iodine (Scheme 53).151 The reaction proceeds through a radical intermediate.

Scheme 53. i. Iodosobenzene diacetate, I2, C6H12, rf, hν, 3.5h, 39%. ii. Iodosobenzene diacetate, I2,

C6H12, rf, hν, 2.5h, 52%.

Iodides 251 and 252 were obtained in moderate yields only and no attempts were made at

cross-coupling reactions based on these intermediates.

Results and Discussions

79

3.3.3 C3-O-Alkyl Derivatives

As indicated in section 2.1, the free hydroxyl group at position 3 of our library structures was

meant to serve as a possible handle for biotinylation or the attachement of fluorescent tags in

compounds of biological interest. For this reason we conducted a series of preliminary

experiments on the alkylation of azido ester 253 as a model compound at an early stage of

the project (Scheme 54).

Scheme 54. i. TBAF, THF, rt, 1h, 90%. ii. See Table 3.

Alkylating reagents 255-262 (Table 3) were prepared in one or two steps from commercially

available starting materials and various conditions for the alkylation reaction were

investigated. As the only electrophile, idodide 261 produced any of the desired alkylation

product, albeit in low yield only (18%). In general, the starting material was recovered

unchanged. These findings indicate that the reactivity of primary alkyl halides of the types

investigated here is not sufficient for the alkylation of the C3-OH group and that other types

of linkages would have to be investigated for the preparation of conjugates of structures 1 or

2.

Results and Discussions

80

Table 3. Screening of alkylation reagents and conditions.

alkylating reagent conditions R Yield (%)

255

Et3N, Et2O, rt, 5h or NaH, DMF, 70°C, 12h

NHAloc 0 0

256

NaH, CH2Cl2, rt, 12h or DTBMP, CH2Cl2 72°C, 12h

NHAloc 0 0

257

NaH, DMF,-15°C-->rt, 2d or NaH, DMF, TBAI, rt, 2d

NHAloc 0 0

258

NaH, THF, rt, 5h NHBn 0

259

NaH, THF, rt, 5h NHBn 0

260

NaH, DMF, 70°C, 24h Br 0

261

NaH, DMF, rt, 12h or DTBMP, DMF, rt, 20h

NHCbz 18 0

262

NaH, THF, rt, 12h NH2 0

In contrast to 255-262, alkylation of 253 with methyl iodide in the presence of NaH gave the

expected C3 methyl ether in 63% yield; the benzylation of 253 gave even higher yields (see

section 3.1.1).

Results and Discussions

81

3.4 Structural Studies

To confirm the overall relative stereochemistry and the postulated boat conformation of the

bicyclo[3.1.0]hexane core of the synthesized bicyclo[3.1.0]hexane derivatives, structural

studies with three representative specific library members were performed, namely 1aa, 1ag

and 1do (Fig. 27).

Figure 27. Structure drawings of 1aa, 1do and 1ag.

3.4.1 Conformational Analysis of 1aa and 1ag by Sol ution NMR and

Simulated Annealing (SA)

The NMR experiments and computational analysis were carried out by Dr. Bernhard Pfeiffer.

Simulated Annealing as a specific computer-based simulation method refers to a randomized

algorithm executing a theoretical stepwise cooling of a structure, in order to find its global

internal energy minimum.152 Each point s of the search space is analogous to a state of the

structure, and the functions E(s) that are to be minimized correspond to the internal energy of

the structure in that state. If the cooling rate is slow enough, the probability distribution of the

current state is near thermodynamic equilibrium at all times and the system indeed relaxes

into its local energy minimum in each iteration step.153 This method was combined with a

Metropolis Monte Carlo/Stochastic Dynamics Simulation.154 The latter was designed as a

means for the examination of statistic-mechanic systems within certain parameter

boundaries.

Results and Discussions

82

In our case, the dynamic parameters represented torsion angles and distances between the

hydrogens attached to the bicyclo[3.1.0]hexane core. The boundaries of these parameters

were determined experimentally from solution NMR experiments. In the scope of this study,

the structures of compounds 1aa and 1ag (Fig. 27) were determined.

The boundaries of hydrogen-hydrogen distances of the bicyclo[3.1.0]hexane core could be

directly deduced from Rotating frame Overhauser effect spectroscopy (ROESY)

experiments. As the strength of the ROE signals is proportional to the inverse sixth power of

the distance between the hydrogens (I~1/r6), the experimentally obtained ROE peaks were

classified according to their intensity to correspond to the denoted distance ranges, (Fig. 28;

values given for 1ag, the same procedure was followed for 1aa).

Figure 28.

H’s

intensity Ia

derived distance r b ((((Å))))

3-2b s 2.15 ± 0.35

3-7a s 2.15 ± 0.35

3-7b s 2.15 ± 0.35

7-5 m 2.65 ± 0.85

4-5 m 2.65 ± 0.85

4-6b m 2.65 ± 0.85

2b-6b m 2.65 ± 0.85

3-5 w 3.40 ± 1.60

2a-3 s 2.15 ± 0.35

6b-3 m 2.65 ± 0.85 Spectra recorded in MeOH-d4.

a: s: strong,

m: medium, w: weak; b: derived from I ~ 1/r6.

The range of the torsion angles was determined by sampling the coupling constants of vicinal

hydrogen atoms, and processing them via the Karplus curve which outputs the range for

possible torsion angles (Fig. 29).

The torsion angles were calculated according to Altona and co-workers.155

Results and Discussions

83

Figure 29. Karplus curve (graph of the Karplus equation J(α) = Acos2α + Bcosα + C; A,B,C empirically

derived parameters depending upon atoms and substituents involved).

The coupling constants measured for 1aa and the corresponding calculated torsion angles

are listed next to Fig. 30 (values given for 1aa, the same procedure was followed for 1ag).

Figure 30.

vicinal H‘s 3JHH (Hz) derived ααααa

2a-3 6.7 44°/143°

2b-3 <1 (0.8) 95°/275°

3-4 <1 (0.8) 90°/273°

4-5 <1 (0.8) 274°/96°

4-7 6.6 33°/142° Spectra recorded in MeOH-d4.

a: the torsion

angles α were calculated using a program

provided at www.stenutz.eu/conf/haasnoot.php

that automatically selects the suitable Karplus

equation, based on Altonas method.155

3JHH (Hz)

α

Results and Discussions

84

For compound 1aa (Fig. 30), the simulation (Fig. 31) clearly confirms the expected relative

stereochemistry. Furthermore, it can be seen that the bicyclo[3.1.0]hexane core indeed

adopts a boat conformation and that the bicyclic core is exceptionally rigid, as all received

minimum-energy conformations superimpose each other almost completely in the range of

the bicyclo[3.1.0]hexane core of the molecule. The finding has an impact on the side chains

which, as a result, receive a defined alignment in space. The results confirm our idea that the

bicyclo[3.1.0]hexane scaffold is a suitable means to achieve a great amount of structural

preorganization of substituents attached to it.

Figure 31. NMR-derived solution structure of library member 1aa [in MeOH-d4].

Similar conclusions can be drawn from the analogous NMR-SA study of compound 1ag (Fig.

32). Again, the relative stereochemistry was confirmed; and the particular rigidity of the

bicyclo[3.1.0]hexane core which also fixes the side chains in defined directions is striking.

Compound 1ag definitely resides in the boat conformation.

Results and Discussions

85

Figure 32. NMR-derived solution structure of library member 1ag [ in MeOH-d4].

3.4.2 Conformational Analysis by X-ray Crystallogr aphy

For compound 1do a monocrystal suitable for X-ray structural analysis was obtained by slow

evaporation of the solvent (MeOH). The analysis was performed by Dr. Bernd Schweizer and

Michael Solar from LOC, ETH Zurich.

The crystallographic data indicated the existence of a twinned crystal, suggesting two

conformational minima for compound 1do in the solid state; however, only very slight

differences were observed between the two structures. These differences mostly relate to the

torsion angles around the bonds that connect the indoline ring and the phthalazinone moiety,

respectively, to the bicyclo[3.1.0]hexane core (Figure 33). The differences in the

corresponding torsion angles were small, ranging from 3-8° (Table 4, entries 5-12).

Results and Discussions

86

Figure 33. Atom numbering for twinned crystal 1do . Torsion angle deviations for the 2 structures are

listed in table 4.

The bicyclo[3.1.0]hexane core adopts the expected boat conformation with the following

torsion angles: C5-C1-C2-C3 15.2°, C3-C4-C5-C1 -12. 3°, C2-C1-C6-C5 97.3°, C4-C5-C6-C1

-95.3° (Table 4, entries 1-4).

Table 4. Torsion angles of twin structures of 1do .

bond

torsion angle structure 1

torsion angle structure 2

C5-C1-C2-C3 15.2° 15.2°

C3-C4-C5-C1 -12.3° -12.3°

C2-C1-C6-C5 97.3° 97.3°

C4-C5-C6-C1 -95.3° -95.3°

C10-C12-C13-N14 109.6° 113.3°

C10-C12-C13-C21 -67.3° -64.4°

C34-N26-C24-C5 174.1° 170.7°

C4-C5-C24-N26 -59.3° -67.1°

The approximate angle between the planes spanned by C2-C1-C5 and C1-C6-C5 is 94°, the

angle between the planes spanned by C4-C5-C1 and C1-C6-C is 91°, for the planes

between C1-C2-C5 and C2-C3-C4, the angle is 151°, a nd the one between C4-C5-C1 and

C2-C3-C4 planes is 154°. These values are in good a greement with the ones that were

received in quantum-mechanical calculations with unsubstituted bicyclo[3.1.0]hexane,26 with

the slight difference that in our case the cyclopropane moiety is almost perpendicular in

relation to the connection of carbons C2, C1, C5, and C4 (they can be viewed approximately

as spanning a plane), whereas a slightly wider angle of about 110° was usually received in

the calculations (c.f. section 1.3).

The crystal structure also confirms the relative stereochemistry of 1do (Fig. 34), and it

reveals the absence of any intramolecular H-bonds.

Results and Discussions

87

Figure 34. Ball and stick depiction of the X-ray structure of 1do .

The crystal structure of 1do is in good agreement with the solution structures obtained for

compounds 1aa and 1ag, as both analyses confirm the expected boat conformation of the

bicyclo[3.1.0]hexane core as well as the overall relative stereochemistry. The existence of

two conformational minima in the solid state is in good agreement with the solution structures

obtained, which also show the existence of several conformational minima, differing only

slightly in the torsion angles for those bonds connecting the side chains to the

bicyclo[3.1.0]hexane core.

Results and Discussions

88

3.5 Synthesis of Bicyclo[3.1.0]hexane-based Analogu es of

Pentostatin and S-Adenosylhomocysteine

3.5.1 Bicyclo[3.1.0]hexane-based Pentostatin 6

As outlined in section 2.1, bicyclo[3.1.0]hexane-based pentostatin analog 6 was to be

obtained from the (1S, 3S, 4S, 5S)-enantiomer of racemic intermediate 4. Amine (+)-4 was

obtained via bicyclic lactone (-)-161; as for the racemic case, the latter can be accessed from

tetrahydrophthalic acid anhydride (174). Entry into the desired enantiomeric series is based

on the racemic resolution of rac-175 (Scheme 55) by conversion into a pair of

diastereoisomeric salts with (-)-ephedrine, which can be separated by fractional

crystallization according to Lindner et al.117b

Scheme 55. Racemic resolution of 175 via fractional crystallization.

After 2-3 recrystallizations and liberation of the acid from the salt by an acidic extraction with

diethylether, (-)-175 was obtained in 49% yield (based on rac-175) and with an ee of 85%,

based on HPLC analysis using a DAICEL chiral column (Fig 35).

Results and Discussions

89

Figure 35. Chiral HPLC analysis of (-)-175.

The elaboration of (-)-175 first into lactone (-)-161 and then into amine (+)-4 followed the

same sequence of steps that is outlined in Schemes 25, 26, and 31 for the synthesis of

racemic 4. The construction of 6 from amine (+)-4 was to be based on an approach that had

previously been elaborated for the simple carbocyclic pentostatin analog 265 (Fig. 36) by

Rapoport and co-workers.156

Figure 36. Structure of carbocyclic pentostatin analogue 265 prepared by Rapoport and co-workers.

Following this lead, amine (+)-4 was then reacted with protected chiral formimidate 162 to

produce imidazole (+)-163 (Scheme 56).

(-)-175

Results and Discussions

90

Scheme 56. Reagents and conditions see Table 5.

Table 5 . Conditions for the reaction of (+)-4 with 162.

entry eq. of (+)-4 eq. of 162 additives solvent temp., time yield (%)

1 1 1 CF3CH2OH 1,2-dichloroethane 80°C (mw),

6.5 h (12)a

2 1 2 CF3CH2OH 1,2-dichloroethane 80°C (mw),

7 h 23b

3 1 0.5 + 0.5c mol. sieves 4Å CH3CN 80°C,

3 h 50b

4 1 0.75 + 0.25 + 0.4c mol. sieves 4Å CH3CN 80°C,

5 h (97)a

5 1 1 + 0.66c CF3CH2OH mol. sieves 4Å CH3CN

80°C, 3.5 h (78)a

a: Formal yields based on mass recovery. Material was impure in all cases. b: Yields based on 1H-NMR

analysis for product mixture. No pure product could be obtained. c: Stepwise addition of 162 with

indicated number of equivalents. eq.: equivalents, temp.: temperature.

Formimidate 162 was prepared in 9 steps starting from N-Cbz-(L)-vinylglycine methyl ester

266 as a chiral starting material according to a procedure also developed by Rapoport and

co-workers (Scheme 57).156b

The epoxide distereoisomers formed in the first step could be separated easily via column

chromatography by switching to a Boc protecting group, while this process had been

ineffective with the originally employed Cbz protecting group. Syn-epoxide 268b was then

transformed to azido-alcohol 269 by treatment with sodium azide. TBS protection and

saponification afforded 271 which was converted into amide 272. After nitrile formation and

deprotection ethoxy-imine 162 was prepared upon reaction with triethylorthoformate.

Results and Discussions

91

Scheme 57. i. m-CPBA, CH2Cl2, rt, 96h, 81%. ii. H2, Pd/C, Boc2O, MeOH, rt, 45min, 50%. iii. NaN3,

NH4Cl, MeOH, rt-->rf, 6h, 84%. iv. TBSCl, im, DMF, rt, 72h, 90%. v. LiOH, dioxane/H2O, 0°C, 3h, 85%.

vi. N-methylmorpholin, i-BuCO2Cl , NH3, THF, -20°C, 3h, 80%. vii. TsCl, py, CH 2Cl2, rt, 72h, 75%. viii.

TFA, CH2Cl2, 0°C, 45min, 53%. ix. HC(OEt 3)3 (neat), 160°C, 2h, 54%.

The yields obtained in the reaction of (+)-4 with 162 were highly variable and strongly

depended upon the exact reaction conditions (Table 5); in addition, the reaction showed poor

reproducibility. The yields reported in entries 2 and 3 of Table 5 are based on 1H-NMR

analysis of material that had been subjected to purification by flash chromatography; even in

these case, however, the purity of the product obtained did not exceed 50%. For entries 1, 4,

and 5, yields are reported for crude products that were used immediately in the subsequent

amidination step. The 1H-NMR spectra of 163 that were recorded after purification by column

chromatography showed a partially decomposed material already upon recording. The poor

reproducibility of the reaction was initially ascribed to the potentially limited thermal stability

of 162 (the reaction was carried out at reflux temperature) and attempts were made to

overcome this problem by the addition of an excess of 162 to the reaction mixture in several

portions, until amine (+)-4 was no longer detectable. This stepwise addition was employed in

entries 3 (addition of 1 eq. in 2 portions), 4 (addition of 1.4 eq. in 3 portions) and 5 (addition

of 1.67 eq. in 2 portions). While this procedure showed a trend toward improvements in the

yield of 163, the latter could never be obtained in purities >50%, presumably due to the

instability of the compound. It was thus used in the next step immediately, either after

purification by column chromatography or as a crude material (Scheme 58).

As illustrated in Scheme 62, 163 was to be reacted with dimethylformamide dimethylacetal to

form 165, which would then be reduced and cyclized to give the bis-TBS-protected target

structure 6.

Results and Discussions

92

Scheme 58. Attempted synthesis of protected pentostatin analog 6.

Unfortunately, the amidination step turned out to be highly problematic. Different reagents

and conditions were sampled for this transformation (163 was treated with 164 and

trifluoroethanol in dichloroethane, microwave irradiation was employed, the solvent was

changed to CH3CN, molecular sieves were added, reaction times were varied), but the

desired 165 was never obtained in pure form. This may be related to a pronounced instability

of the compound, although this has not been investigated in any detail. Attempts to convert

crude preparations into the target structure under a variety of conditions (by the addition of

propanedithiol as reducing agent with Et3N in MeOH at 50°C, various reaction times) proved

to be unsuccessful.

3.5.2 Bicyclo[3.1.0]hexane-based S-Adenosylhomocyst eine 7

The projected synthesis of bicyclo[3.1.0]hexane-based S-adenosylhomocysteine 7, as for

pentostatin analog 6, was to proceed through amine (+)-4 as a key intermediate.

As summarized in Scheme 60, (+)-4 was elaborated into (-)-221 in analogy to the synthesis

of rac-221 (section 3.1.3.1.2, Scheme 44) in 45% overall yield.

Results and Discussions

93

Scheme 60. i. 219, Et3N, 1,4-dioxane, rf, 20h. ii. diethoxymethyl acetate (neat), 110°C, 20h, 45% (over

2 steps).

221 was then converted into the protected 2’-deoxyadenosine analog 167 by chloride

displacement with ammonia through heating of 221 in a saturated solution of ammonia in

methanol. Subsequent selective deprotection of the primary hydroxyl group with camphor

sulfonic acid led to 281. Unfortunately, the projected conversion of 281 into bromide 282

failed.

Scheme 61. i. NH3 in MeOH, ))), 100°C, 4h, 87%. ii. CSA, CH 2Cl2/MeOH 1:1, 0°C, 4h, 89%. iii. PPh 3,

CBr4, dioxane, ))), 80°C, 3d.

Results and Discussions

94

Based on the assumption that the free primary amino group of the adenine base might be the

cause of the problems with the bromination at C7, 221 was converted to its N6-Alloc

derivative 283 prior to cleavage of the C7 TBS ether (Scheme 62).

Scheme 62.i. N-methylimidazole, allylchloroformate, CH2Cl2, 0°C, 36h, 61%. ii. CSA, CH 2Cl2/MeOH

1:1, 0°C, 4h, 67%. iii. TsCl, Et 3N, DMAP, CHCl3, rt, 4d, 36%.

Selective cleavage of the primary TBS ether followed by tosylation with Ts-Cl then gave

tosylate 285, albeit in low overall yield (15% over 3 steps). While the yield of the tosylation

step was clearly unsatisfactory, model experiments with cyclopentylmethanol indicated that

tosylation was still more efficient than mesylation. More significantly, however, compared to

mono-allylated compound 283, bis-BOC-protected 2’-deoxyadenosine analog 168, obtained

from 167 by treatment with an excess of BOC2O (4 eq.) in excellent yield, could be converted

into tosylate 170 with much higher efficiency (66 % yield for the two-step sequence from 168

compared to 24% for the conversion of 283 into 285). In addition, BOC-protection of N6

promised to offer the possibility of simultaneous cleavage with the TBS groups.

Results and Discussions

95

Scheme 63. i. BOC2O, DMAP, DMF, rt, 14h, 87%. ii. CSA, CH2Cl2/MeOH 1:1, 0°C, 10h, 88%. iii. TsCl,

Et3N, DMAP, CHCl3, rt, 3d, 75%. iv. i. 1) NaOMe (1.3 eq.), MeOH, rt, 1h. 2) rf, 8h, 60%.

Our original plan had been to displace the tosyloxy group with N-α-BOC-(L)-homocysteine

tert-butyl ester, thus providing a fully protected derivative of the target structure with mostly

tert-butyl-based protecting groups. Unfortunately, this strategy could not be implemented,

due to the inability to acess the required homocysteine derivative according to a known

literature procedure.157 As an alternative approach, the side chain was attached through the

reaction of tosylate 170 with thiolactone 286 to provide methyl ester 287 in 60% yield.

Thiolactone 286 was prepared in one step and 84% yield from commercially available (L)-

homocysteine thiolactone. Upon addition of sodium methoxide in methanol, 286 is converted

to a methyl ester with a terminal thiolate group, which then displaces the tosylate upon

heating of the reaction mixture. In principle, this method bears the risk of epimerization at the

α-carbon of the amino acid.

Saponification of the methyl ester moiety in 287 was accomplished with potassium hydroxide

and afforded acid 173 in 70% yield (Scheme 64).

Results and Discussions

96

Scheme 64. i. KOH aq. (2 eq.), THF/H2O 1:1, rt, 24h, 70%.

During two trials for the conversion of tosylate 170 to 287, a product corresponding to 287

but lacking two BOC-groups was obtained. This product (287a) was also subjected to ester

hydrolysis, yielding 173a, and the complete deprotection of this material was attempted with

HCl in ethyl acetate, it was hoped that the final product would precipitate from the solution as

its hydrochloride, which would have obviated the need for chromatographic purification of the

final product. (Given the high polarity of 7 we were skeptical about the prospects for the

purification of this material, which turned out to be justified). As the deprotection with HCl did

not go to completion in 3 days and the resulting hydrochloride salt was impure and poorly

soluble even in water, the final deprotection with trifluoroacetic acid was investigated as an

alternative. Upon stirring of 173 with a large excess of trifluoroacetic acid in dichloromethane

the TBS and the three Boc groups could be cleaved within 32 hours (Scheme 65).

Scheme 65. i. TFA, CH2Cl2, rt, 32h, yield not determined.

The resulting TFA-salt of 7 was purified by semi-preparative HPLC on a Zorbax-SB-Phenyl

reversed-phase column. As the analytical HPLC trace of crude 7 showed 2 peaks with the

same mass under certain conditions, we had to reconsider the possibility that epimerization

of the chiral centre at C-8 had taken place earlier during the nucleophilic substitution reaction

of 170 with 286 and that we had obtained a mixture of diastereoisomers (Fig. 36). The two

peaks showed an integration ratio of 2:1. They were separated in several batches using a

precisely determined injection volume and ratio of the two eluents water and acetonitrile; the

slightest change in these two parameters or in the HPLC method led to the loss of

separation.

Results and Discussions

97

Figure 36. C-8 epimers of 7.

However, the materials isolated by preparative HPLC gave identical NMR spectra (Fig. 37),

which did not support the idea that the two peaks observed in the HPLC trace corresponded

to two different diastereoisomers.

Figure 37. 1H-NMR spectra of 7 [D2O].

Reanalysis of the purified samples by analytical HPLC again gave two peaks in both cases.

The peaks had identical masses and they displayed the same intensity ratio as prior to the

purification process, although they had clearly been separated in the preparative runs (Fig.

38, depicting the HPLC chromatograms recorded after the preparative separation). These

findings led us to reject the epimerization hypothesis; rather, the compound appears to exist

as an equilibrium mixture of two different forms.

7 (6 min ret.time)

7 (8 min ret. time)

Results and Discussions

98

Figure 38a. HPLC chromatogram of 7 (first peak).

Figure 38b. HPLC chromatogram of 7 (second peak).

This hypothesis is also supported by the fact that HPLC analysis of a commercial sample of

S-adenosylhomocysteine (dissolved in H2O with 0.03% TFA, under identical conditions to

those employed for 7) also gave two peaks with identical mass (Fig. 39). No attempts were

made to clarify the nature of the species that are involved in the equilibrium reaction, as only

small amounts of material were available at this stage.

Results and Discussions

99

Figure 39. HPLC chromatogram of S-Adenosylhomocysteine (SAH).

Results and Discussions

100

3.6 Biological Evaluation of the Bicyclo[3.1.0]hexa ne-based

Chemical Library

3.6.1 Reporter Gene Assay Screening for Inhibitory Effects on Selected

Cellular Signaling Pathways

40 library members were tested against a panel of 9 reporter gene assays158 reflecting the

transcriptional output of different cellular signaling networks. (These experiments were

performed at the Novartis Institiute for Biomedical Research in Basel in collaboration with Dr.

Doriano Fabbro and Dr. Daniel D’Orazio at the Novartis Institute for Biomedical Research, in

Basel). The 40 compounds of the library were selected such as to achieve maximal structural

diversity and thus included representatives of each ‘sub family’ of target structures 1 and 2.

Reporter gene assays (RGAs) in the most general sense are cellular detection systems that

rely on the transfection of cells with genes, whose expression results in a readily recordable

physical signal, such as fluorescence. By providing integrated readouts for the activity of

signalling pathways or networks, RGAs, among other things, allow to study the interference

of small molecules with such signaling cascades or networks in live cells.159 The assays

employed in this study involved transcriptional readouts requiring the activation of TFAP1

(via EGF or TNFα stimulation), NFκB (via TNFα stimulation), the tumor suppressor protein

p53 (via TGFβ-stimulation), the cAMP pathway response element CRE, the TGFb signaling

pathway response element SBE, the ARE response element, reflecting oxidative stress, and

the control CMV promoter element to monitor for non signaling pathway-related interactions

within the assay system. In all cases, the different transcriptional response elements control

the expression of a β-lactamase reporter gene, whose expression results in the cleavage of

an exogenously added substrate. Due to fluorescence resonance energy transfer (FRET)

this substrate emits green fluorescence upon excitation at 409 nm; cleavage by the β-

lactamase enzyme eliminates the FRET and the cleavage product exhibits blue

fluorescence. The ratio of blue vs. green fluorescence is a direct measure for reporter gene

activity and thus for the activity of the pathway or network under investigation. The substrate

is added to the cells as a lipophilic pro-form, which penetrates the cell membrane by passive

diffusion and is subsequently converted to the charged substrate by cellular esterases. Due

to its charged nature the substrate remains trapped within the cells. These principles are

summarized in Scheme 66.

Results and Discussions

101

Scheme 66. Schematic representation of the reporter gene assay principle. (depiction adopted from

Invitrogen)

Across all compounds and pathways/networks investigated in this thesis, inihibitory activity

was observed only in the EGF- and/or TNFα-triggered AP1 assay (13 compounds) and in the

TGFβ-sensitive SBE assay (3 compounds). The structures of the active compounds together

with their IC50 values are depicted in Table 6. In particular, compounds 1dc , 1de and 1dm

showed sub-µM inhibition of AP1 activation after stimulation with TNFα. Quite intriguingly,

none of these compounds (or any of the other actives identified) showed any measurable

inhibition of TNFα-triggered NFκB activation at concentrations < 10 µM.

Results and Discussions

102

Table 6. Active compounds identified in reporter gene assays.

Compound Reporter gene assay (IC 50 [µµµµM])a

AP1-EGF AP1-TNFαααα SBE

2.43 0.68

1.00 0.31

3.58 0.66

1.69

6.10 3.83

6.57 4.12

Results and Discussions

103

3.61 3.19

8.78

HO

NH

O

N

1fd Cl

O

8.94 4.03

3.62

4.50

8.98

3.99 6.80

a:IC50 values were determined by measuring each compound in a dilution series of 8 concentrations

(single measurement at each concentration) and using a 4-Parameter Logistic or Sigmoidal Dose-

Response Model for data fitting. The ratio of blue/green fluorescence was measured ratiometrically. Z-

values (measure for assay reliability, Z = 1 for ideal assay) were 0.6-0.7 (0.21 for TNFα).

Results and Discussions

104

The epidermal growth factor (EGF) receptor is a tyrosine kinase and together with the three

additional members ERBB2-ERBB4 constitutes the EGF receptor family. Activating mutants

and over-expression of these family members contribute to oncogenesis by inducing cells to

proliferate and to resist apoptosis. EGFR signals through the RAS/ERK-, the PI3-kinase and

the JAK/STAT pathways (Fig. 40).160

Figure 40. EGF-mediated signaling pathways.

TNFα acts through three main pathways upon binding to the TNFα-receptor type 1 and

subsequent TRADD binding; the pathways involving NFκB, the MAPK pathways, both

associated with cell proliferation and inflammation, and the pathways signaled through

FADD, leading to apoptosis.

Activity against EGF- or TNFα-mediated AP-1 activation seems to benefit from an indoline

moiety at the carboxy terminus, as all compounds active in the sub-micromolar range share

this structural feature; only two compounds with a structurally similar (methoxylated)

tetrahydroisoquinoline moiety were found to exhibit effects below a concentration of 10 µM.

Of these two compounds, 1be bears the same N-terminal capping group as 1de, but is ca.

20-fold less potent in the AP1-TNFα assay than the latter.

Results and Discussions

105

No specific conclusions can be drawn with regard to the nature of the N-terminal moiety,

except that polar acyl residues like in compounds 1dg and 1do are not tolerated.

3.6.2 In Vitro Protease Inhibition

By cleaving proteins, proteases are involved in a large number of key physiological

processes such as cell-cycle progression, cell proliferation and cell death, DNA replication,

tissue re-modelling, haemostasis (coagulation) and the immune response. We chose to

investigate the potential inhibitory activity of the bicyclo[3.1.0]hexane based chemical library

on the proteases thrombin, cathepsin B and urokinase (uPA). As our small chemical library

consists of compounds representing general peptidic structures, some of them also including

natural desamino acids, an interaction with proteases was well conceivable.

Thrombin is a serine protease and a potential target for antithrombotic agents.161 Known

inhibitors include benzamidines, benzoxazinones and macrocyclic peptides. Urokinase (uPA)

is a serine protease whose expression is upregulated in a number of diseases, e.g. in

cancer.162 Cathepsin B is a cysteine protease whose overexpression has been associated

with certain tumors163 and Alzheimer’s disease.164 The potential inhibitory activities against

these proteases were determined in kinetic assays using fluorometric substrates: if the

protease is inhibited by a certain compound, it cannot cleave the substrate to the same

extent anymore and therefore the fluorescence, which is released upon cleavage, drops.165

3.6.2.1 Thrombin Inhibition Assay

Thrombin (100 nM) in PBS (20 mM NaH2PO4, 30 mM Na2HPO4, 100 mMNaCl, pH 7.4) was

incubated with 84 library members as potential inhibitors at a concentration of 10 µM in 384-

well microtiter plates for 30 min at room temperature. The reaction was started by the

addition of the fluorogenic substrate Z-GGR-AMC (Bachem, Bubendorf, Switzerland, Fig. 43)

dissolved in PBS and 1% DMSO to a final concentration of 0.25 mM in a total volume of

50 µl.

Results and Discussions

106

O

HN O

NH

O

O NH

O

NH

O ONH

H2N

NH

Figure 43. Structure of thrombin and uPA substrate Z-GGR-AMC.

The change in fluorescence signal (ex: 360 nm, em: 460 nm) was recorded over 30 min

using a SpectraMaxmicroplate reader (Molecular Devices). As a positive control a 0.5 µM

solution of the known inhibitor hirudin fragment 54-65 was employed.

No thrombin inhibitory activity was found for any of the 84 tested library members at a

concentrations of 10 µM (Fig. 44). Figure 33 shows the dispersion of the fluorescence

intensities between 10’000 and 20’000 RFU for individual compounds, but this finding had to

be attributed partly to the assay conditions. The measurements were repeated at least three

times under identical conditions and gave a similar dispersion each time, but no correlating

with individual compounds was observed. The library compounds corresponding to lower

fluorescence values in at least two of three or four repeat experiments were additionally

investigated in dilution series, in which the concentrations of the library compounds were

increased up to 1 mM, but no inhibitor was found even at this concentration.

The large dispersion is also caused by the fact that certain library members such as 1bb ,

1bo , 1cg , 1cn , and 218a, 218ab, 218ac boosted fluorescence. The mechanism underlying

this effect remains unclear. The enhanced fluorescence could not be attributed to auto-

fluorescence of the corresponding compounds, as measurements showed.

Results and Discussions

107

Figure 44. Thrombin inhibition kinetic assay: measured data of 84

compounds. Relative fluorescence units (RFU) were measured

against time in seconds. curve a: positive control (hirudin 54-65

0.5 µM).

3.6.2.2 Urokinase Inhibition Assay

Urokinase (62 nM) in PBS (20 mM NaH2PO4, 30 mM Na2HPO4, 100 mM NaCl, pH 7.4) was

incubated with 84 library members as potential inhibitors at a concentration of 10 µM in 384-

well microtiter plates for 30 min at room temperature. The reaction was started by the

addition of the fluorogenic substrate Z-GGR-AMC (Bachem, Bubendorf, Switzerland), which

had also been used in the thrombin assay. The substrate was dissolved in PBS and 1%

DMSO to a final concentration of 0.1 mM in a total volume of 50 µl. The change in

fluorescence signal (ex: 383 nm, em: 460 nm, cutoff: 420 nm) was recorded over 30 min

using a SpectraMaxmicroplate reader (Molecular Devices). As a positive control a 120 µM

solution of the known inhibitor benzamidine was used.

Time (secs)

Results and Discussions

108

Figure 45. Urokinase inhibition kinetic assay: measured data of 84

compounds. Relative fluorescence units (RFU) were measured against

time in seconds. curve a: positive control (benzamidine 120 µM).

No urokinase inhibitory activity was found for any of the 84 library members at a

concentration of 10 µM (Fig. 45). All the measurements were repeated at least three times

under identical conditions and gave a similar dispersion each time, not correlating with

individual compounds. The library compounds corresponding to lower fluorescence values in

at least two of the quadruplicate measurements were additionally investigated in dilution

series, in which the concentrations of the library compounds were increased up to 1mM, but

no inhibitor was found even at this concentration.

3.6.2.3 Cathepsin B Inhibition Assay

Cathepsin B (5.4 nM) in 0.1% (v/v) Brij 35 solution and a solution of (L)-Cysteine (8 mM) in

PBS (352 mM KH2PO4, 48mM Na2HPO4, 4 mM ethylenediamine tetraacetic acid, pH 6.0)

was incubated with 84 library members as potential inhibitors at a concentration of 10 µM in

384-well microtiter plates for 30 min at room temperature. The reaction was started by the

addition of the fluorogenic substrate Arg-Arg-7-AMC (Bachem, Bubendorf, Switzerland)

dissolved in 0.1% Brij 35 solution and 1% DMSO to a final concentration of 0.15mM in a total

volume of 50 µl. The change of fluorescence signal (ex: 360 nm, em: 460 nm, cutoff: 420 nm)

was recorded over 30 min using a SpectraMaxmicroplate reader (Molecular Devices). As a

positive control a 10.3 µM solution of the known inhibitor cystatin was used.

Time (secs)

Results and Discussions

109

Figure 46. Cathepsin B inhibition kinetic assay: data of the duplicate

measurement of 48 compounds. Relative fluorescence units (RFU) were

measured against time in seconds. curve a: positive control (cystatin

10.3 µM).

No cathepsin B inhibitory activity was found for any of the 84 library members at a

concentration of 10 µM (Fig. 46, for clarity, the data of 36 compounds were omitted). All the

measurements were repeated at least three times under identical conditions and gave a

similar dispersion each time, not correlating with individual compounds. The library

compounds corresponding to lower fluorescence values in at least two of the triplicate

measurements were additionally investigated in dilution series, in which the concentrations of

the library compounds were increased up to 1 mM, but no inhibitor was found even at this

concentration.

3.6.3 Antibiotic Activity against Mycobacteria

3.6.3.1 Compounds of General Structure 1 and 2

78 library members were tested for their antibiotic activity against Mycobacterium

tuberculosis, a growing strain (H37Rv) and a non-growing one (18b), in collaboration with

Prof. Stewart Cole and Dr. Ruben Hartkoorn at the EPF Lausanne. For the determination of

anti-tuberculosis activity, a resazurin reduction microplate assay (REMA) was used.166 This

assay determines the results also by fluorescence, making use of reazurin, a blue dye, being

reduced to fluorescent resorufin by NADPH/NADH in viable cells. If the cell viability

decreases due to the presence of a growth inhibitor, the reduction of reazurin can no longer

occur and as a consequence fluorescence will be reduced.167

Time (secs)

Results and Discussions

110

Compound 1ea showed some activity against the growing H37Rv strain (table 7), but its

activity is substantially lower than that of rifampicin, which was used as the reference

compound in the assay and is a standard drug in tuberculosis treatment. None of the tested

compounds showed any activity against the non-growing M. tuberculosis strain 18b.

Table 7. Results of the screening of 78 library compounds for anti-tuberculosis activity. 1ea

showed a moderate inhibiting effect on growing M.tuberculosis (H37Rv).

Conc. compound

growing

M.tuberculosis

(H37Rv)

non-growing

M.tuberculosis

(18b)

20 µM

3742 RFUa 20'000 RFUb

0.5 µM rifampicin 2754 RFU 6487 RFU

a: Average value of a duplicate measurement. b: Against non-growing M.tuberculosis, single measurements were carried out. RFU = relative fluorescence units.

3.6.3.2 Antibiotic Activity against Mycobacterium Tuberculosis of 5-Alkynyl-

deoxypyrimidine Nucleoside Analogue 243

Bicyclo[3.1.0]hexane based 5-alkynyl-deoxypyrimidine nucleoside analogue 243 was tested

for antimycobacterial activity (collaboration with Dr. Thomas Keller at the Novartis Institute for

Tropical Diseases, in Singapore).

Figure 47. Structures of 2’-deoxy-nucleoside 244 and its bicyclo[3.1.0]hexane-based analogue 243.

The corresponding 5-alkynyl-2’-deoxypyrimidine nucleoside 244 was reported in 2005 (see

section 3.2) to inhibit M.bovis at a minimum inhibiting concentration (MIC90) of 50 µg per ml,

with some activity against other mycobacteria, e.g. M.avium (Table 8).

Table 8. Antimycobacterial activity of 5-alkynyl-dideoxypyrimidine168 nucleoside 244.

Results and Discussions

111

compound

M.bovis

% inhibition

(concentration, µg/ml)a

MIC90 (µg/ml)

M.avium

% inhibition

(concentration, µg/ml)a

244 90 (100, 50), 70 (10), 25(1) 50 75 (100), 0 (50)

MIC90: Concentration of compound exhibiting 90% inhibition on mycobacterial growth a: Antimicobacterial activity was determined at concentrations 100, 50, 10, and 1 µg/ml.

However, the synthesized bicyclo[3.1.0]hexane-based analogue 243, corresponding to a

southern conformation of the nucleoside, exhibited no antimycobacterial activity at all.

This finding can mean that the molecular target(s) of 244 in the bacteria interact(s)

preferentially or even exclusively with the northern conformer of the nucleoside. Rai and co-

workers found in 2007 that the corresponding 5-alkynyl-dideoxy-pyrimidine nucleoside was

even more active than 244 by a factor of 25-50 regarding the inhibition of M.bovis.169

3.6.4 Cytotoxicity

A subset of 78 library compounds were tested for their ability to inhibit human cancer cell

proliferation (collaboration with Prof. Jürg Gertsch and Dr. Andrea Chicca at the University of

Bern). The cytotoxicity of the library compounds against MCF-7 (breast cancer) and U937

(lymphoma) cells was determined after 72 hours incubation time in a WST-1 (4-[3-(4-

iodophenyl)-2-(4-nitrophenyl)-2H-5-tetrazolio]-1,3-benzen disulphate) based cell viability

assay.170 The assay is based on the enzymatic cleavage of the tetrazolium salt WST-1 to

formazan by cellular mitochondrial dehydrogenases present in viable cells, and the extent of

the cleavage is then measured colorimetrically. All compounds were tested in a

concentration of 5 µM. Every test was performed in triplicate.

There was no significant effect detectable on the proliferation of U937 cells for any of the

library compounds (Fig. 48). However, proliferation of MCF-7 cells was inhibited by library

members 218r, 1ai, 1fd , 1bc , 1cg , 1be, 1bm and 210i in the range of 35-42%. Noticeably,

compound 1fd increased cell proliferation by 42%. In MCF-7 cells, cell-growth is known to be

regulated by estrogen receptors.171 The experimental finding regarding compound 1fd may

suggest that this compound is exhibiting its activity through these receptors. The compounds

diminishing cell viability may also act on these receptors.

Results and Discussions

112

Figure 48. Effect of selected library members on cell

proliferation of MCF-7 and U937 cells. Compound conc.:

5 µM.

The structures of active compounds are shown in Fig. 49. The all carry a hydrogen bond

acceptor functionality on the right-hand side of the bicyclo[3.1.0]hexane core structure, with

the only exception being compound 1ai. Regarding the left-hand side modifications, different

structural entities are represented and allow no definite conclusion what features are

conducing to cytotoxic effects. Compounds 218r, 210i, 1fd , 1be, 1bm also exhibited effects

on signaling pathways associated with cell proliferation.

218

r

1ai

1fd

1bc

1cg

1be

1bm

210

i

Results and Discussions

113

Figure 49. Structures of compounds showing weak effects on cell viability.

Results and Discussions

114

3.7 Biological Evaluation of the Bicyclo[3.1.0]hexa ne-based S-

Adenosylhomocysteine Analogue

3.7.1 Inhibition of AdoMet-dependant Methyltransfer ases by S-

Adenosylhomocysteine analogue 7

The group of AdoMet-dependant methyltransferases includes 7 DNA methyltransferases, 5

RNA methyltransferases, 4 protein methyltransferases and 4 small molecule methyl-

transferases acting on the carbon, oxygen or nitrogen atoms of their substrates. For the

biological evaluation of bicyclo[3.1.0]hexane based S-adenosylhomocysteine analogue 7, the

conformationally restricted analogue of (S)-Adenosylhomocysteine, we chose to investigate

the inhibitory effect on dengue N-7 and O-2’ methyltransferases, as this enzyme represents a

valid target for antiviral agents, due to a virus-conserved cavity located next to its co-

substrate, S-adenosylmethionine (AdoMet). The cavity has been shown to be occupied by

product inhibitor S-adenosylhomocysteine and related compounds, whereas these small

molecules had no inhibitory effect on related human enzymes.172

Dengue methyltransferase sequentially methylates viral RNA at the N7 and O-2’ positions,

using adenosylmethionine as a co-substrate. Using different RNA substrates and buffer

conditions, it is possible to examine the N7 and the O-2’ methylation by Dengue

methyltransferase separately. After incubation of the enzyme with various concentrations of

7, radioactively labeled AdoMet [S-adenosyl-L-(methyl-3H)-methionine] was added and the

levels of methylation were monitored (Fig. 50) in a scintillation proximity assay.173 The assay

employs biotinylated RNA substrates bound to streptavidin beads which emit light upon

methylation (binding of the radioactive methyl group). This event enables photometric

detection of methylation.

Results and Discussions

115

Figure 50. Dose-response curve of 7 for different concentrations on dengue MT methylation activity.

As a control, AdoMet and the enzyme were monitored accordingly. No inhibitory effect on

either of the two methylation sites was found for 7.

S-adenosylhomocysteine (SAH), however, inhibits dengue methyltransferase N7-

methylation with an IC50 value of 1.8 µM, and the O-2’-methylation at 0.5 µM.174 The lack of

any inhibitory effect of 7 on dengue methyltransferases activity may suggest that the enzyme

interacts preferentially with SAH in its 3’-endo conformation, as in contrast our

conformationally restricted analogue 7 corresponds to a 2’-endo or southern conformation.

dose response for 7

Conclusions

116

4 Conclusions and Outlook

The synthesis of a small, structurally unique chemical library of 110 new bicyclo[3.1.0]hexane

derivatives has been accomplished. Due to the unique conformational properties of the

bicyclo[3.1.0]hexane system, these derivatives were considered to provide an attractive

basis for the identification of new lead structures for drug discovery. The synthesis has

exploited the efficient conversion of the bicyclic lactone 161 into acid 3 and amine 4, which

were then elaborated into individual library members. Structural studies on 3 representative

library members confirmed the expected boat conformation of the bicyclo[3.1.0]hexane core

in these derivatives, and it is very resonable to assume that this conformational preference is

also prevalent in the majority, if not all other library members.

The chemisty developed in the course of the library work was also exploited for the synthesis

of S-adenosyl homocysteine (SAH) analog 7, based on chiral amine (+)-4. SAH analog 7

appears to exist as an equlibrium mixture of two species, at least under certain conditions. In

contrast to 7, the projected synthesis of pentostatin analog 6 from amine (+)-4 could not be

implemented. Based on the experiments performed in this thesis, it would appear that the

envisioned route from (+)-4, which parallels an appraoch that had been successfully applied

to the synthesis of a cyclopentane-based pentostatin analog, is not viable.

Screening of the library against 3 proteases (thrombin, urokinase, and cathepsin B) did not

reveal any inhibitory activity. However, not definitive conclusions are possible on the basis of

the limited selection of enzymes with respect to the general potential of compounds of type 1

to act as protease inhibitors.

Investigation of the antiproliferative activity of a selection of compounds 1 against MCF-7

human breast carcinoma cells provided 8 library members with weak inhibitory activity

(inihibition of proliferation by 35-42% at 5 µM compound concentration). Interestingly, one

compound increased cell proliferation by 42% (at 5 µM); it remains to be investigated

whether these findings my reflect interaction of (at least some) of the compounds with

estrogen receptors.

The most promising biological effects were observed in cellular reporter gene assays. Thus,

4 compounds, 1dc , 1dm , 1de, and 1df , were found to selectively inhibit the EGF- or

TNFα−mediated activation of the transcription factor AP-1. The exact molecular

mechanism(s) underlying these effects are unknown at this point.

Conclusions

117

Future work should include the screening of our unique library against a variety of additional

biological targets, the elucidation of the mechanism(s) underlying the inhibition of AP-1

activation, and the establishment of a more comprehensive SAR for the inhibition of these

pathways. The chemistry established in this thesis provides a sound basis for the efficient

synthesis of additional derivatives of type 1, as would be required as part of the SAR work.

Experimental Section

118

5 Experimental Section

5.1 General Methods

All non-aqueous reactions were carried out using oven-dried or flame-dried glassware under

a positive pressure of dry argon or nitrogen unless otherwise stated. Tetrahydrofuran,

acetonitrile, toluene, diethyl ether, dichloroethane, N, N-dimethylform-amide, dimethyl

sulfoxide and methylene chloride were purchased as anhydrous from Fluka. All chemicals

were purchased from Acros, Aldrich, Fluka, Merck, Lancaster, ABCR or TCI and used as

such unless otherwise stated. Deuterated solvents were obtained from Aldrich and

Cambridge Isotope Labs.

Reactions were magnetically stirred if not indicated otherwise and monitored by thin layer

chromatography using Merck silica gel 60 F254 TLC aluminium backed plates and visualized

by fluorescence quenching under UV light. In addition, TLC plates were stained with cerium

molybdate or potassium permanganate stain. Chromatographic purification was performed

as flash chromatography on Fluka Silica Gel 60 (230-400 mesh) using a forced flow of eluant

at 0.3 bar. Technical grade solvents were employed, which were distilled prior to use.

Concentration under reduced pressure was performed by rotary evaporation at 40 °C at the

appropriate pressure. Purified compounds were further dried for 12 – 48 h under high

vacuum (0.01 – 0.05 Torr). Yields refer to chromatographically purified and spectroscopically

pure compounds, unless stated otherwise.

Melting points: Melting points were measured on a Büchi B-540 melting point apparatus

using open glass capillaries and are uncorrected.

Optical rotations: Optical rotations were measured on a JASCO P-1020 digital polarimeter at

the sodium D line with a 100 mm or 10 mm path length cell, and are reported as follows:

[α]DT, concentration (g/100 ml), and solvent.

NMR spectra: 1H- and 13C-NMR spectra were recorded on a Bruker AV-400 400 MHz and a

Bruker DRX-500 500MHz spectrometer at room temperature. Chemical shifts (δ) are

reported in ppm with the solvent resonance as the internal standard relative to chloroform (δ

7.26 ppm for 1H and 77.0 ppm for 13C). All 13C spectra were measured with complete proton

decoupling. Data are reported as follows: s = singlet, d = doublet, t = triplet, q = quartet, m =

multiplet, br = broad, coupling constants in Hz, integration.

IR spectra: IR spectra were recorded on a Jasco FT/IR-6200 instrument as thin film.

Absorptions are given in wavenumbers (cm–1).

Experimental Section

119

Mass spectra: Mass spectra were recorded by the MS service at ETH Zürich. EI-MS (m/z):

EI-HIRES Micromass Autospel-ULTIMA spectrometer at 70 eV. ESI-MS (m/z): IONSPEC

Ultima ESI-FT-ICR spectrometer at 4.7 T. MALDI-MS (m/z): Ion Spec Ultima HR. Low

resolution mass spectra were acquired as ESI-MS (m/z) with a Waters ZQ mass

spectrometer coupled to a Waters LC unit.

HPLC analyses: HPLC analyses were carried out on a Merck-Hitachi device using a Waters

Symmetry C18 column (3.5 µm, 4.6 x 100 mm) or an Agilent Zorbax SB-Phenyl column (3.5

µm, 4.6 x 150 mm). Semi-preparative HPLC was carried out using an Agilent Zorbax SB-

Phenyl column (5 µm, 9.4 x 150 mm). Preparative HPLC was carried out using Waters

Symmetry C18 columns (5 µm, 7.8 x 100 mm and C18, 5 µm, 19 x 100 mm).

Experimental Section

120

5.2 Experimental Procedures and Analytical Data

5.2.1 Synthesis of Bicyclo[3.1.0]hexane-based Chemi cal Library

5.2.1.1 Synthesis of Lactone 161

Lactone 161 in racemic form was prepared as described in section 6.2.4.1, except for the

fractionate crystallization step that was not carried out. Instead, 175 was directly converted to

176 and 177, respectively. In general, for the preparation of 161, procedures were followed

as described in 117b and in 117a. The analytical data were the same except for chiroptical data

that were not measured.

5.2.1.2 Synthesis of Advanced Intermediates 3, 4, a nd 5

183, 184, and 185 were prepared from 161 in the same way as described for the

enantiomeric series in section 6.2.4.2. Their analytical data were identical except for

chiroptical data that were not measured.

187

(1R,2S,3R,4S)-methyl 2-(chloromethyl)-4-hydroxy-3-

(hydroxymethyl)cyclopentanecarboxylate

To 161 dissolved in MeOH (3 ml), TMSCl (0.63 g, 5.810 mmol) was added under stirring,

then ZnCl2 (8 mg, cat.) was added and the reaction mixture was stirred at RT for 24 h. MeOH

was evaporated and the crude product, a green oil, was purified by flash column

chromatography (EtOAc/MeOH 11:1) to afford 80 mg (0.359 mmol, 62%) of the product as a

colorless oil.

Rf = 0.4 (EtOAc/MeOH 11:1).

1H-NMR (400 MHz, CDCl3): δ = 4.10-4.04 (m, 1H), 3.79 (dd, J = 10.7, 5.0 Hz, 1H), 3.70 (s,

3H), 3.71-3.66 (m, 1H), 3.66-3.61 (m, 1H), 3.58 (dd, J = 11.0, 8.0 Hz, 1H), 3.06-3.01 (m, 1H),

2.38-2.29 (m, 1H), 2.20-2.12 (m , 1H), 2.05-1.98 (m, 1H), 1.97-1.89 (m, 1H).

13C-NMR (100 MHz, CDCl3): δ = 175.9, 75.3, 63.3, 53.2, 52.2, 45.5, 45.0, 44.2, 37.5.

Experimental Section

121

HR-MS (ESI): calcd for C9H16ClO4 [M+H]+: 223.0737, found: 223.0732.

188

(1R,2S,3R,4S)-methyl 4-((tert-butyldimethylsilyl)ox y)-3-(((tert-

butyldimethylsilyl)oxy)methyl)-2-(chloromethyl)cycl opentanecarboxylate

To a solution of 187 (0.49 g, 2.201 mmol) in 6 ml of CH2Cl2 MTBSA (1.72 g, 9.903 mmol) was

added drop by drop (over 10 min) under cooling to 0°C. The reaction mixture was stirred at

RT for 16 h and diluted with 20 ml of CH2Cl2. The organic phase was washed with water (3 x

40 ml), dried over MgSO4 and evaporated. The residue was purified by flash column

chromatography (CH2Cl2) to afford 0.61 g (1.338 mmol, 62%) of the product as a colorless

oil.

Rf = 0.5 (CH2Cl2).

1H-NMR (400 MHz, CDCl3): δ = 4.03-3.96 (m, 1H), 3.72-3.67 (m, 2H), 3.69 (s, 3H), 3.66-

3.63 (m, 2H), 2.95-2.86 (m, 1H), 2.54-2.41 (m, 1H), 2.16-2.08 (m, 1H), 1.96-1.87 (m, 2H),

0.89 (s, 9H), 0.87 (s, 9H) 0.05 (s, 3H), 0.05 (s, 3H), 0.04 (s, 3H), 0.04 (s, 3H).

13C-NMR (100 MHz, CDCl3): 174.1, 72.6, 61.7, 53.2, 51.8, 45.7, 43.2, 42.5, 37.6, 25.9, 25.8,

18.2, 17.9, -4.5, -4.9, -5.5, -5.6.

MS (ESI): calcd for C21H44ClO4Si2 [M+H]+: 451.25, found: 450.79.

185

(1S,3S,4R,5S)-methyl 3-((tert-butyldimethylsilyl)ox y)-4-(((tert-

butyldimethylsilyl)oxy)methyl) bicyclo[3.1.0]hexane -1-carboxylate

188 (8.6 g, 0.019 mol) was dissolved in 60 ml of tert-BuOH, then tert-BuOK (2.87 g, 0.025

mol) was added slowly under stirring. The resulting suspension was stirred for 20h at RT,

then additional 660 mg of tert-BuOK were added. After additional 2h of stirring the reaction

mixture was poured on 600 ml of Et2O and extracted with ice water (3x 200 ml). The organic

phase was dried over MgSO4 and evaporated. Purification by flash column chromatography

(CH2Cl2/Et3N 0.5%) yielded 5.06 g (0.012 mmol, 64%) of the product as a white solid.

Experimental Section

122

The analytical data were identical to the ones stated for 185 in enantiomeric form (section

6.2.4.2), except for chiroptical data that were not measured.

190

(1S,3S,4R,5S)-methyl 3-((tert-butyldimethylsilyl)ox y)-4-

(hydroxymethyl)bicyclo[3.1.0]hexane-1-carboxylate

To a solution of 185 (4.5 g, 0.011 mol) in CH2Cl2/MeOH 1:1 (100 ml) CSA (3.1 g, 0.013 mol)

was added at 0°C. The reaction mixture was stirred for 2.5 h at 0°C. The reaction was

quenched with saturated aqueous NaHCO3 –solution and extracted with CH2Cl2 (4 x 120 ml).

The combined organic phases were dried over MgSO4 and evaporated. The residue was

purified by flash column chromatography (EtOAc/hex 1:2) to give 2.21 g (0.007 mol, 67%) of

the product as a colorless oil.

Rf = 0.34 (EtOAc/Hex 1:1)

1H-NMR (400 MHz, CDCl3): δ = 4.16 (d, J = 6.6 Hz, 1H), 3.63 (s, 3H), 3.53 (dd, J = 10.7, 6.5

Hz, 1H), 3.42 (dd, J = 10.7, 7.7 Hz, 1H), 2.51 (ddd, J = 14.2, 6.8, 1.1 Hz, 1H), 2.07 (t, J = 7.2

Hz, 1H), 1.80 (d, J = 14.2 Hz, 1H), 1.75 (dd, J = 8.9, 5.6 Hz, 1H), 1.58 (s, br, 1H), 1.47-1.41

(m, 2H), 1.24 (t, 7.3 Hz, 1H), 0.84 (s, 9H), 0.01 (s, 6H).

13C-NMR (100 MHz, CDCl3): δ = 175.1, 75.5, 64.6, 53.1, 51.6, 37.0, 32.2, 30.6, 25.8, 20.7,

17.6, -4.8.

MS (ESI): calcd for C15H29O4Si [M+H]+: 301.18, found: 301.13 (I = 100%).

191

(1S,3S,4R,5S)-methyl 3-((tert-butyldimethylsilyl)ox y)-4-(((methylsulfonyl)oxy)methyl)

bicyclo[3.1.0]hexane-1-carboxylate

Mesylchloride (0.5 g, 4.413 mmol) was added dropwise at 0°C to a stirred solution of 190

(0.884 g, 2.942 mmol) and Et3N (0.62 ml, 4.413 mmol) in Et2O (20 ml). The resulting

suspension was stirred for 3 h, then it was diluted with water and Et2O and the phases were

Experimental Section

123

separated. The organic phase was washed with water, dried over MgSO4 and evaporated to

afford 1.1 g (2.816 mmol, 96%) of the product as a colorless oil.

Rf = 0.31 (EtOAc/Hex 1:1)

1H-NMR (400 MHz, CDCl3): δ = 4.19 (d, J = 6.5 Hz, 1H), 4.12 (dd, J = 9.9, 5.5 Hz, 1H), 4.02

(dd, J = 10.2, 7.9 Hz, 1H), 3.66 (s, 3H), 3.02 (s, 3H), 2.57 (dd, J = 14.2, 6.5 Hz, 1H), 2.31

(7.6, 5.8 Hz, 1H), 1.86 (d, J = 14.4 Hz, 1H), 1.75 (t, J = 6.8 Hz, 1H), 1.50 (d, J = 6.8 Hz, 2H),

0.86 (s, 9H), 0.03 (s, 6H).

13C-NMR (100 MHz, CDCl3): δ = 174.3, 75.2, 69.6, 51.9, 49.9, 37.7, 36.7, 31.0, 30.6, 25.5,

20.6, 18.1, -4.8, -4.9.

MS (ESI): calcd for C16H31NaO6SSi [M+H]+: 401.14, found: 401.19 (I = 90%).

192

(1S,3S,4R,5S)-methyl 4-(azidomethyl)-3-((tert-

butyldimethylsilyl)oxy)bicyclo[3.1.0]hexane-1-carbo xylate

Sodium azide (1.195 g, 0.018 mol) was added to a solution of 191 (2.91 g, 0.007 mol) in dry

DMF (40 ml) and stirred for 20 h at 70°C. The react ion mixture was poured on water and

extracted with Et2O (4x 70 ml). The phases were separated, the combined organic phases

were dried over MgSO4 and evaporated. Purification of the residue by flash column

chromatography (Hex/EtOAc 10:1) yielded 1.82 g (0.006 mol, 76%) of the product as a

colorless oil.

Rf = 0.6 (EtOAc/Hex 1:1)

1H-NMR (400 MHz, CDCl3): δ = 4.09 (d, J = 6.5 Hz, 1H), 3.64 (s, 3H), 3.26 (dd, J = 12.1, 6.0

Hz, 1H), 3.09 (dd, J = 12.1, 8.3 Hz, 1H), 2.53 (ddd, J = 14.4, 6.9, 1.1 Hz, 1H), 2.11 (t, J = 7.3

Hz, 1H), 1.82 (d, J = 14.1 Hz, 1H), 1.72 (dd, J = 8.8, 5.6 Hz, 1H), 1.49-1.43 (m, 2H), 0.84 (s,

9H), 0.02 (s, 3H), 0.02 (s, 3H).

13C-NMR (400 MHz, CDCl3): δ = 174.6, 76.1, 53.9, 50.4, 36.9, 31.9, 30.9, 25.7, 20.4, 17.9, -

4.8, -4.9.

HR-MS (ESI): calcd for C15H28N3O3Si [M+H]+: 326.1894, found: 326.1903.

Experimental Section

124

3

(1S,3S,4R,5S)-4-(azidomethyl)-3-((tert-butyldimethy lsilyl)oxy)bicyclo[3.1.0]hexane-1-

carboxylic acid

192 (240 mg) was dissolved in THF (5 ml) and 3 drops of DMF and potassium

trimethylsilanolate (350 mg) was added immediately. The reaction mixture was heated in the

microwave to 80°C for 2 h. The reaction mixture was quenched by the addition of ice and 2 N

HCl that was added dropwise. The water phase was extracted with EtOAc (3x 30 ml), the

combined organic phases were dried over MgSO4 and evaporated to give 212 mg (92%) of a

colorless oil that crystallized to a white solid upon storage in the fridge.

Rf = 0.18 (CH2Cl2)

1H-NMR (400 MHz, CDCl3): δ = 4.11 (d, J = 7.0 Hz, 1H), 3.28 (dd, J = 12.3, 6.0 Hz, 1H), 3.12

(dd, J = 12.3, 8.2 Hz, 1H), 2.53 (ddd, J = 14.5, 7.0, 1.2 Hz, 1H), 2.13 (dd, J = 8.4, 7.0 Hz,

1H), 1.87-1.79 (m, 2H), 1.59-1.53 (m, 2H), 0.85 (s, 9H), 0.03 (s, 3H), 0.02 (s, 3H).

13C-NMR (300 MHz, CDCl3): δ = 180.8, 75.9, 54.1, 50.5, 36.4, 33.4, 31.1, 25.8, 21.6, 18.2, -

4.8.

IR (film): ν 2950, 2929, 2858, 2099, 1683, 1449, 1253, 1165, 1084, 1047, 907, 837, 776,

730, 539.

MS (ESI): calcd for C14H24N3O3Si [M-H]-: 310.16, found: 310.16 (I = 100%).

253

(1S,3S,4R,5S)-methyl 4-(azidomethyl)-3-hydroxybicyc lo[3.1.0]hexane-1-carboxylate

192 (0.85 g) was dissolved in THF (20 ml), then TBAF (3.3 ml, 1 M sol. in THF) was added

slowly under stirring. The reaction mixture was stirred for 1 h at RT. 20 ml of a saturated

NH4Cl-sol. was added and the mixture was extracted with EtOAc (3x 50 ml). The organic

phases were combined, driedover MgSO4 and evaporated. Purification by flash column

chromatography (EtOAc/hex 1:1) yielded 0.5 g (90 %) of the title compound as a colorless

oil.

Experimental Section

125

Rf = 0.31 (EtOAc/hex 1:1)

1H-NMR (400 MHz, CDCl3): δ = 4.17 (dd, J = 6.9, 1.2 Hz, 1H), 3.63 (s, 3H), 3.27 (dd, J =

12.3, 6.6 Hz, 1H), 3.19 (dd, J = 12.3, 7.5 Hz, 1H), 2.61 (ddd, J = 14.6, 6.9, 1.7 Hz, 1H), 2.14

(t, J = 7.0 Hz, 1H), 2.04 (s, br, 1H), 1.84 (dd, J = 14.5, 1.2 Hz, 1H), 1.76 (ddd, J = 9.0, 5.2,

1.0 Hz, 1H), 1.52 (ddd, J = 9.0, 4.3, 1.7 Hz, 1H), 1.39 (t, J = 4.9 Hz, 1H).

13C-NMR (100 MHz, CDCl3): δ = 174.3, 76.0, 53.9, 52.0, 49.5, 36.6, 32.5, 31.0, 21.4.

HR-MS (ESI): calcd for C9H13N3NaO3 [M-Na]+: 234.0849, found: 234.0844.

193

(1S,3S,4R,5S)-methyl 4-(azidomethyl)-3-(benzyloxy)b icyclo[3.1.0]hexane-1-carboxylate

253 (1.68 g) was dissolved in anhydrous DMF (90 ml) and NaH (412 mg, 60% dispersion in

oil) was added under cooling to -15°C; after 5 min benzylbromide (1.79 g) was added, then

the mixture was warmed to RT and stirred for 1.5 h. The reaction was quenched with H2O

and extracted with EtOAc. The combined organic phases were dried over MgSO4 and

evaporated to afford the crude product which was purified by flash column chromatography

(EtOAc/Hex 1:12-->1:1) to yield 2.2 g (92%) of 193 as a slightly yellow oil.

Rf = 0.3 (EtOAc/Hex 1:10).

1H-NMR (400 MHz, CDCl3): δ = 7.34-7.21 (m, 5H), 4.37 (s, 2H), 3.82 (dd, J = 6.9, 0.8 Hz,

1H), 3.62 (s, 3H), 3.25 (dd, J = 12.2, 6.2 Hz, 1H), 3.12 (dd, J = 12.2, 8.0 Hz, 1H), 2.50 (ddd, J

= 14.6, 6.9, 1.7 Hz, 1H), 2.33 (t, J = 7.0 Hz, 1H), 2.05 (dd, J = 14.5, 0.6 Hz, 1H), 1.73 (ddd, J

= 9.0, 5.3, 1.0 Hz, 1H), 1.46 (ddd, J = 9.0, 4.3, 1.7 Hz, 1H), 1.38 (t, J = 4.9 Hz, 1H).

13C-NMR (100 MHz, CDCl3): δ = 174.6, 138.3, 128.4, 127.7, 127.5, 83.3, 71.2, 53.9, 52.0,

47.4, 33.4, 31.9, 30.9, 20.6.

IR (film): ν 3031, 2955, 2866, 2096, 1717, 1494, 1442, 1355, 1266, 1208, 1143, 1072, 1029,

904, 732, 693.

MS (ESI): calcd for C16H19N3NaO3 [M-Na]+: 324.13 , found: 323.95 (I = 60%).

Experimental Section

126

194

(1S,3S,4R,5S)-4-(azidomethyl)-3-(benzyloxy)bicyclo[ 3.1.0]hexane-1-carboxylic acid

193 (300 mg) was dissolved in THF (8 ml) and DMF (2 ml) and TMSOK (425 mg) was added

immediately. The reaction mixture was heated in the microwave at 90°C for 4 h. The reaction

mixture was quenched by adding ice, along with dropwise addition of 2 N aqueous HCl. The

mixture was extracted with EtOAc (3 x 30 ml) and the combined organic phases were dried

and evaporated. Purification by flash chromatography (CH2Cl2/MeOH 50:1) yielded 262 mg

(91%) of the product as a slightly yellow oil.

Rf = 0.11 (CH2Cl2/MeOH 50:1)

1H-NMR (400 MHz, CDCl3): δ = 7.37 – 7.26 (m, 5H), 4.42 (s, 2H), 3.88 (d, J = 6.6 Hz, 1H),

3.32 (dd, J = 12.3, 6.4 Hz, 1H), 3.18 (dd, J = 12.3, 8.0 Hz, 1H), 2.54 (ddd, J = 14.6, 7.0, 1.5

Hz, 1H), 2.44 – 2.37 (m, 1H), 2.09 (d, J = 14.7 Hz, 1H), 1.90 – 1.82 (m, 1H), 1.59 (ddd, J =

9.0, 4.4, 1.7 Hz, 1H), 1.54 – 1.48 (m, 1H).

13C-NMR (100 MHz, CDCl3): δ = 179.8, 138.4, 129.2, 128.5, 128.2, 127.8, 127.5, 125.2,

82.9, 71.3, 54.3, 47.2, 32.9, 30.8, 21.3.

IR (film): ν 2928, 2901, 2619, 2358, 2099, 1718, 1683, 1452, 1430, 1353, 1274, 1227, 1213,

1166, 1077, 1028, 912, 737, 697, 545.

HR-MS (ESI): calcd for C15H17N3NaO3 [M-Na]+: 310.1162, found: 310.1153.

5

(1S,3S,4R,5S)-4-(azidomethyl)-3-(benzyloxy)bicyclo[ 3.1.0]hexan-1-amine

194 (80 mg) was dissolved in toluene (3 ml) and cooled to 0°C, then DPPA (86 mg) and

Et3N (46 ul) were added. The reaction mixture was stirred for 1 h at 0°C, then 1.5 h at RT

and for 12 h at 80°C. During the reaction, the colo rless solution turned slightly yellow. The

crude isocyanate was diluted with THF, treated with 2 N NaOH and stirred for 30min. The

solvent was evaporated and the water phase was extracted with CH2Cl2 (3 x 50 ml). The

combined organic phases were dried over MgSO4 and evaporated. Purification by flash

Experimental Section

127

chromatography (CH2Cl2/MeOH 30:1-->CH2Cl2/MeOH 20:1) afforded 59 mg (82%) of the

product as a yellow oil.

Rf = 0.3 (EtOAc/Hex 1:1)

1H-NMR (400 MHz, MeOD): δ = 7.39 – 7.21 (m, 5H), 4.40 (s, 2H), 3.84 (d, J = 7.0 Hz, 1H),

3.54 (dd, J = 12.3, 6.4 Hz, 1H), 3.42 (ddd, J = 12.2, 8.3, 3.8 Hz, 1H), 2.31 – 2.18 (m, 2H),

2.15 (d, J = 13.8 Hz, 1H), 1.33 – 1.30 (m, 1H), 1.18 (td, J = 4.8, 1.5 Hz, 1H), 0.85 (ddd, J =

9.2, 4.7, 2.3 Hz, 1H).

13C-NMR (100 MHz, MeOD): δ = 139.1, 129.2, 128.8, 128.5, 83.6, 71.4, 55.1, 55.0, 41.7,

38.8, 28.6, 19.8.

IR (film): ν 3337, 2925, 2856, 2356, 2096, 1623, 1454, 1351, 1268, 1210, 1092, 1061, 963,

736, 697.

MS (ESI): calcd for C14H19N4O [M+H]+: 259.16, found: 259.17 (I = 100%).

4

(1S,3S,4R,5S)-3-((tert-butyldimethylsilyl)oxy)-4-(( (tert-

butyldimethylsilyl)oxy)methyl)bicyclo[3.1.0] hexan- 1-amine

4 was prepared analogeously to the procedure described for (+)-4 (section 6.2.4.2) in 3 steps

from 185. The analytical data were the same, except for chiroptical data that were not

measured.

Experimental Section

128

5.2.1.3 Synthesis of library members of general str ucture 1

202a

((1S,3S,4R,5S)-4-(azidomethyl)-3-((tert-butyldimeth ylsilyl)oxy)bicyclo[3.1.0]hexan-1-

yl)(3,4-dihydro isoquinolin-2(1H)-yl)methanone

3 (200 mg) was dissolved in CH2Cl2 (6 ml) and DIPA (272 ul) was added under stirring, after

5 min HATU (366 mg) was added. 1,2,3,4-tetrahydroisoquinoline (197a, 166 ul) was added

and the reaction mixture was stirred at RT for 24 h. The mixture was diluted with CH2Cl2, the

phases were separated and the organic phase was washed with NaHCO3 sol. and with brine,

then the organic phase was dried over MgSO4 and evaporated to dryness. The residue was

purified by flash column chromatography (Hex/EtOAc 2:1) to give 226 mg (83%) of the

product as a slightly yellow oil.

Rf = 0.33 (Hex/EtOAc 2:1)

1H-NMR (400 MHz, CDCl3): δ = 7.24-7.10 (m, 4H), 4.80-4.65 (m, 2H), 4.18 (dd, J = 6.7, 0.8

Hz, 1H), 3.94-3.76 (m, 2H), 3.44 (dd, J = 12, 6 Hz, 1H), 3.10 (dd, J = 12.5, 9.5 Hz, 1H), 2.87

(t, J = 5.8 Hz, 2H), 2.70 (ddd, J = 14.5, 6.5, 1.0 Hz, 1H), 2.12 (dd, J = 9.4, 6 Hz, 1H), 1.90

(dd, J = 13.7, 0.6 Hz, 1 H), 1.63 (ddd, J = 8.9, 4.8, 1.0 Hz, 1H), 1.39 (t, J = 4.5 Hz, 1H), 0.96

(ddd, J = 8.6, 4.2, 1.0 Hz, 1H), 0.87 (s, 9H).

13C-NMR (100 MHz, CDCl3): δ = 171.7, 133.2, 128.8, 126.7, 126.5, 76.2, 53.9, 50.4, 39.4,

32.4, 27.2, 25.8, 18.8, 17.9, -4.7, -4.8.

IR (film): ν 2927, 2855, 2096, 1637, 1432, 1254, 1167, 1086, 1036, 931, 832, 776, 747, 674.

HR-MS (ESI): calcd for C23H35N4O2Si [M-H]+: 427.2524, found: 427.2522.

Experimental Section

129

203a

((1S,3S,4R,5S)-4-(aminomethyl)-3-((tert-butyldimeth ylsilyl)oxy)bicyclo[3.1.0]hexan-1-

yl)(1,2,3,4-tetrahydronaphthalen-2-yl)methanone

202a (57 mg) was dissolved in Tol/MeOH 1:1 (1.5 ml), Pd/C (28 mg) was added and the

reaction mixture was stirred under H2 for 2 h. The reaction mixture was filtered through celite,

the residue was washed with EtOAc and the solvents were evaporated to afford 54 mg

(100%) of the crude product as a yellow oil that was immediately used to prepare

compounds 1ax.

Rf = 0.13 (EtOAc/MeOH 60:1)

1H-NMR (400 MHz, CDCl3): δ = 7.21-7.09 (m, 4H), 4.84-4.58 (m, 2H), 4.18 (d, J = 6.5 Hz,

1H), 3.88 (s, br, 2H), 3.45-3.34 (m, 1H), 2.92-2.85 (m, 2H), 2.78 (dd, J =11.5, 5.5 Hz, 1H),

2.31 (dd, J = 13.5, 6.5 Hz, 1H), 2.20-1.97 (m, 1H), 1.92-1.86 (m, 1H), 1.65 (dd, J = 8.9, 4.7

Hz, 1H), 1.39-1.34 (m, 1H), 0.99-0.94 (m, 1H), 0.87 (s, 9H), 0.03 (s, 6H).

13C-NMR (100 MHz, CDCl3): δ = 133.5, 128.8, 126.3, 77.8, 69.5, 53.2, 45.4, 40.9, 33.2, 27.7,

25.7, 18.2, 17.7, -4.4, -4.7.

HR-MS (ESI): calcd for C23H37N2O2Si [M-H]+: 401.2619, found: 401.2625.

1ax

The acids (in the amounts stated below, 0.038 mmol each) were dissolved in separate

reaction tubes in CH2Cl2 (0.3 ml each). 20-30 µl of DMF were added to each reaction tube.

204a 204b 204d 204e 204f 204g 204i 204m 204n 204p 204t

2.5ul 2.8ul 2.8 ul 3.3 mg 2.5 mg 9.7 mg 5 mg 7 mg 9 mg 8.4 mg 3 mg

DIPEA (32 ul, 0.188 mmol each) and HATU (15 mg, 0.040 mmol each) were added to each

reaction tube. The reaction mixtures were stirred for 30 min, then 203a (70 mg) was added

by syringe in equimolar amounts (10 mg, 0.025 mmol) to each reaction tube. The reaction

Experimental Section

130

mixtures were stirred for 12 h. The solutions were evaporated to dryness. The compounds

were subjected to TBS deprotection by being shaken with Dowex-50W-X8 (40 mg each) in

MeOH (0.3 ml each) for 12 h. The resin-bound acid was filtered off, the filtrates were

evaporated to dryness and the crude products were purified by flash column chromatography

to yield library members 1ax in the amounts stated below.

Amount : 4 mg. Purity : ≥ 98%. TLC: Rf 0.35 (EtOAc/MeOH 10:1, KMnO4). MS (ESI): calcd for

C22H31N2NaO3 [M-Na]+: 393.22, found: 392.97 (I = 100%). 1H-NMR (400 MHz, acetone): δ = 7.52 (s,

br, 1H), 7.23-7.16 (m, 4H), 4.76 (s, br, 2H), 4.15 (d, J = 6.6 Hz, 1H), 3.97-3.86 (m, 2H), 3.54 (ddd, J =

12.7, 7.6, 4.9 Hz, 1H), 3.10 (ddd, J = 13.4, 5.6, 3.8 Hz, 1H), 2.93-2.87 (m, 2H), 2.21 (ddd, J = 13.4,

6.7, 1.4 Hz, 1H), 2.19 (t, J = 5.2 Hz, 1H), 2.11-2.07 (m, 1H), 2.04-1.97 (m, 3H), 1.54 (ddd, J = 9.0, 4.8,

1.0 Hz, 1H), 1.40 (t, J = 4.6 Hz, 1H), 1.04 (ddd, J = 9.0, 4.3, 1.4 Hz, 1H), 0.91 (d, J = 6.6 H, 3H), 0.89

(d, J = 6.6 Hz, 3H). IR (film): ν 3355, 2926, 2867, 2359, 2341, 2100, 1635, 1518, 1455, 1236, 1066,

1034, 822, 738, 698.

Amount : 6 mg. Purity : ≥ 97%. TLC: Rf 0.22 (EtOAc/MeOH 20:1, KMnO4). MS (ESI): calcd for

C25H26F2N2NaO3 [M-H]-: 439.18, found: 438.96 (I = 100%). 1H-NMR (400 MHz, MeOD): δ = 7.20-7.13

(m, 4H), 6.92 (d, J = 2.0 Hz, 1H), 6.90 (d, J = 2.0 Hz, 1H), 6.76 (td, J = 9.3, 2.0 Hz, 1H), 4.89-4.60 (m,

2H), 4.06 (d, J = 6.3 Hz, 1H), 4.01-3.79 (s, br, 2H), 3.53 (s, 2H), 3.33 (dd, J = 13.7, 5.9 Hz, 1H), 3.21

(dd, J = 13.7, 6.6 Hz, 1H), 2.89 (t, J = 5.9 Hz, 2H), 2.15 (t, J = 6.2 Hz, 1H), 2.11 (dd, J = 13.8, 6.4 Hz,

1H), 1.96 (dd, J = 13.9, 1.0 Hz, 1H), 1.62 (dd, J = 8.8, 4.4 Hz, 1H), 1.34 (t, J = 4.7 Hz, 1H), 1.04-0.98

(m, 1H).

Experimental Section

131

Amount : 3 mg. Purity : ≥ 92%. TLC: Rf 0.33 (EtOAc/MeOH 10:1, KMnO4). MS (ESI): calcd for

C23H33N2O3 [M-H]+: 385.25, found: 385.10 (I = 100%).

Amount : 2 mg. Purity : ≥ 96%. MS (ESI): calcd for C21H27N2NaO3 [M-Na]+: 377.18, found: 377.10 (I =

100%).

HO

NH

O

N

O

1ae

Amount : 4 mg. Purity : ≥ 96%. TLC: Rf 0.33 (EtOAc/MeOH 10:1, KMnO4). MS (ESI): calcd for

C23H33N2O3 [M-H]+: 419.23, found: 419.10 (I = 100%). 1H-NMR (500 MHz, DMSO): δ = 7.97 (t, J = 5.2

Hz, 1H), 7.28-7.13 (m, 9H), 4.73 (d, J = 2.8 Hz, 1H), 4.78-4.57 (m, 2H), 3.93-3.89 (m, 1H), 3.89-3.72

(m, 2H), 3.12-3.04 (m, 1H), 3.04-2.97 (m, 1H), 2.88-2.80 (m, 2H), 2.81 (t, J = 7.9 Hz, 2H), 2.39 (t, J =

7.9 Hz, 2H), 2.02 (dd, J = 13.6, 6.7 Hz, 1H), 1.97 (t, J = 6.9 Hz, 1H), 1.85 (d, J = 13.6 Hz, 1H), 1.48

(dd, J = 8.5, 4.3 Hz, 1H), 1.25 (t, J = 4.3 Hz, 1H), 0.93-0.88 (m, 1H). 13C-NMR (500 MHz, DMSO): δ =

171.5, 171.2, 141.5, 134.6, 133.4, 128.7, 128.3, 128.2, 126.4, 126.3, 126.2, 125.9, 75.2, 49.6, 41.7,

37.2, 32.4, 31.1, 27.0, 17.8. IR (film): ν 3674, 3299, 2953, 2927, 2857, 2359, 1617, 1544, 1496, 1471,

1453, 1394, 1251, 1223, 1170, 1079, 1038, 1005, 938, 864, 836, 757, 750, 699.

Amount : 1 mg. Purity : ≥ 79%. MS (ESI): calcd for C23H33N2O3 [M-H]+: 385.25, found: 385.10 (I =

100%).

Experimental Section

132

Amount : 3 mg. Purity : ≥ 94%. TLC: Rf 0.35 (EtOAc/MeOH 10:1, KMnO4). MS (ESI): calcd for

C23H33N2O3 [M-H]+: 397.25, found: 397.10 (I = 100%).

Amount : 14 mg. Purity : ≥ 82%. TLC: Rf 0.30 (EtOAc/MeOH 20:1, KMnO4). HR-MS (ESI): calcd for

C28H34N3O5S [M-H]+: 524.2214, found: 524.2215. 1H-NMR (400 MHz, MeOD): δ = 7.48 (d, J = 1.5 Hz,

1H), 7.41 – 7.31 (m, 2H), 7.23 – 7.14 (m, 5H), 4.21 (d, J = 6.3 Hz, 1H), 3.96 – 3.89 (m, 4H), 3.90 –

3.85 (m, 2H), 3.51 (dd, J = 13.5, 6.4 Hz, 1H), 3.42 (dd, J = 13.5, 6.7 Hz, 1H), 3.15 (d, J = 5.3 Hz, 4H),

3.23 – 3.03 (m, 2H), 2.94 – 2.87 (m, 2H), 2.33 (t, J = 6.5 Hz, 1H), 2.26 (dd, J = 14.0, 6.7 Hz, 1H), 2.06

(d, J = 13.9 Hz, 1H), 1.74 (dd, J = 9.0, 4.6 Hz, 1H), 1.41 (t, J = 4.7 Hz, 1H), 1.10 – 0.99 (m, 1H). 13C-

NMR (100 MHz, MeOD): δ = 170.3, 165.4, 149.7, 136.9, 131.2, 130.0, 129.7, 127.2, 122.5, 121.2,

120.2, 119.7, 118.2, 115.9, 77.4, 51.5, 48.4, 43.9, 40.4, 38.9, 36.9, 34.0, 31.6, 28.6, 19.0.

Amount : 4 mg. Purity : ≥ 81%. TLC: Rf 0.18 (EtOAc/MeOH 20:1, KMnO4). HR-MS (ESI): calcd for

C29H31N2O6 [M-H]+: 503.2177, found: 503.2170. 1H-NMR (400 MHz, MeOD): δ = 7.70 (d, J = 8.9 Hz,

1H), 7.24 – 7.12 (m, 4H), 6.91 (dd, J = 8.9, 2.6 Hz, 2H), 6.83 (s, 1H), 6.29 (s, 1H), 4.05 (d, J = 6.6 Hz,

1H), 3.89 (d, J = 3.7 Hz, 2H), 3.84 (s, 3H), 3.34 (dd, J = 10.0, 3.8 Hz, 1H), 3.25 (dd, J = 13.6, 6.3 Hz,

1H), 2.90 (dd, J = 9.8, 5.2 Hz, 2H), 2.81 (s, 4H), 2.17 (t, J = 6.1 Hz, 1H), 2.06 (dd, J = 14.1, 7.1 Hz,

1H), 1.93 (d, J = 14.0 Hz, 1H), 1.59 (dd, J = 9.1, 4.6 Hz, 1H), 1.34 (t, J = 4.7 Hz, 1H), 1.01 (dd, J =

12.5, 7.8 Hz, 1H).

Experimental Section

133

Amount : 8.5 mg. Purity : ≥ 96%. TLC: Rf 0.14 (EtOAc/MeOH 10:1, KMnO4). HR-MS (ESI): calcd for

C28H33N4O4 [M-H]+: 489.2496, found: 489.2496.

Amount : 11.5 mg. Purity : ≥ 95%. TLC: Rf 0.23 (EtOAc/MeOH 20:1, KMnO4). HR-MS (ESI): calcd for

C28H32N3O3 [M-H]+: 458.2438, found: 458.2422. 1H-NMR (400 MHz, MeOD): δ = 7.53 (d, J = 7.9 Hz,

1H), 7.27 (d, J = 8.2 Hz, 1H), 7.23-7.1 (m, 4H), 7.00 (s, 1H), 6.97 (t, J = 7.5 Hz, 1H), 6.86 (t, J = 7.1

Hz, 1H), 4.65 (s, br, 2H), 3.86 (d, J = 6.2 Hz, 1H), 3.82 (s, br, 2H), 3.36-3.32 (m, 1H), 3.12 (dd, J =

13.5, 6.0 Hz, 1H), 3.09-3.03 (m, 2H), 2.89-2.86 (m, 2H), 2.58 (o, J = 7.0 Hz, 2H), 2.08 (t, J = 5.8 Hz,

1H), 1.70 (t, J = 12.0 Hz, 1H), 1.65 (dd, J = 14.2, 6.2 Hz, 1H), 1.45 (dd, J = 8.9, 4.5 Hz, 1H), 1.26 (t, J

= 4.6 Hz, 1H), 0.98-0.90 (m, 1H). 13C-NMR (100 MHz, MeOD): δ = 176.1, 174.2, 138.4, 129.2, 128.5,

127.0, 123.1, 122.3, 119.5, 118.6, 110.6, 78.4, 51.3, 43.6, 39.9, 38.5, 34.3, 28.6, 22.6.

Amount : 13 mg. Purity : ≥ 94%. TLC: Rf 0.27 (EtOAc/MeOH 20:1, KMnO4). HR-MS (ESI): calcd for

C23H27N2O4 [M-H]+: 395.1965, found: 395.1977. 1H-NMR (400 MHz, MeOD): δ = 7.34 (d, J = 1.0 Hz,

1H), 7.23-7.15 (m, 4H), 6.28 (s, br, 1H), 6.20 (d, J = 2.9 Hz, 1H), 4.72 (s, br, 2H), 4.08 (dd, J = 6.8, 0.9

Hz, 1H), 3.90 (s, br, 2H), 3.57 (s, 2H), 3.35-3.31 (m, 1H), 3.23 (dd, J = 13.8, 6.8 Hz, 1H), 2.91 (t, J =

5.5 Hz, 2H), 2.21-2.14 (m, 2H), 1.99 (dd, J = 14.1, 1.0 Hz, 1H), 1.64 (dd, J = 9.1, 4.8 Hz, 1H), 1.37 (t, J

= 4.6 Hz, 1H), 1.02 (dd, J = 7.9, 3.7 Hz, 1H). 13C-NMR (100 MHz, MeOD): δ = 174.1, 171.7, 150.8,

143.2, 135.4, 130.1, 128.0, 127.4, 111.4, 108.4, 77.7, 51.1, 43.4, 40.3, 39.4, 36.6, 34.1, 28.4, 19.0.

Experimental Section

134

202d

((1S,3S,4R,5S)-4-(azidomethyl)-3-((tert-butyldimeth ylsilyl)oxy)bicyclo[3.1.0]hexan-1-

yl)(5-chloro- indolin-1-yl)methanone

3 (70 mg) was dissolved in 2 ml of CH2Cl2 and DIPA (95 ul, 0.674 mmol) was added under

stirring and after 5 min HATU (128 mg, 0.338 mmol) was added. 5-Chloroindoline (200d, 44

mg) was added and the reaction mixture was stirred at RT for 24 h. It was diluted with CH2Cl2

and poured on 5% aqueous citric acid solution. The water phase was extracted with CH2Cl2

(3x 60 ml). The combined organic phases were washed with saturated aqueous NaHCO3-

solution and with brine, dried over MgSO4 and evaporated. Purification of the residue by flash

column chromatography (Hex/EtOAc 5:1) yielded 95 mg (94%) of the product as a slightly

green oil that solidified upon storage at -20°C.

Rf = 0.28 (Hex/EtOAc 5:1)

1H-NMR (400 MHz, CDCl3): δ = 7.94 (d, J = 8.2 Hz, 1H), 7.17-7.11 (m, 2H), 4.35-4.26 (m,

1H), 4.23-4.09 (m, 2H), 3.48 (dd, J = 12.3, 6.0 Hz, 1H), 3.19-3.11 (m, 3H), 2.33 (dd, J =13.3,

6.0 Hz, 1H), 2.15 (dd, J = 9.7, 6.0 Hz, 1H), 2.01 (d, J = 13.6 Hz, 1H), 1.79 (dd, J = 9.4, 5.2

Hz, 1H), 1.47 (t, J = 4.8 Hz, 1H), 1.09 (ddd, J = 9.0, 4.5, 1.4 Hz, 1H), 0.88 (s, 9H), 0.06 (s,

3H), 0.05 (s, 3H).

13C-NMR (100 MHz, CDCl3): δ = 170.4, 141.7, 133.3, 128.7, 127.3, 125.0, 117.5, 76.2, 53.7,

50.0, 48.3, 38.2, 34.5, 28.2, 27.5, 25.9, 19.3, 17.8, -4.8, -4.9.

HR-MS (ESI): calcd for C22H32ClN4O2Si [M+H, M+Na]+: 447.1978, found: 447.1977.

203d

202d (113 mg, 0.253 mmol) was dissolved in Tol/MeOH 1:1 (2.5 ml), then 10% Pd/C (54 mg,

0.051 mmol) was added and the reaction mixture was stirred vigorously under a hydrogen

atmosphere (1 bar) for 2 h. The reaction mixture was filtered through celite, the residue was

Experimental Section

135

washed with EtOAc, and the filtrate was concentrated under reduced pressure to afford 110

mg (0.261 mmol) of the crude product that was immediately used to prepare compounds

1dx .

Rf = 0.1 (Hex/EtOAc 3:1)

1H-NMR (400 MHz, CDCl3): δ = 7.97 (d, J = 7.4 Hz, 1H), 7.29 – 7.04 (m, 4H), 4.42 – 4.15 (m,

2H), 4.24 (dd, J = 6.4, 1.2 Hz, 1H), 3.21 (t, J = 8.3 Hz, 2H), 3.09 (dd, J = 12.9, 5.5 Hz, 1H),

2.92 (dd, J = 12.9, 9.8 Hz, 1H), 2.38 (dd, J = 14.5, 5.9 Hz, 1H), 2.27 (dd, J = 9.8, 5.2 Hz, 1H),

2.16 (dd, J = 13.9, 1.4 Hz, 1H), 1.88 (dd, J = 8.9, 4.6 Hz, 1H), 1.60 (t, J = 4.6 Hz, 1H), 1.26

(ddd, J = 9.2, 4.6, 1.5 Hz, 1H), 0.93 (s, 9H), 0.12 (s, 6H).

MS (ESI): calcd for C22H34N2NaO2Si [M-Na]+: 409.23, found:409.31 (I = 100%).

1dx

The compounds 1dx were prepared in the same way as exemplified on compounds 1ax.

Amount : 3 mg. Purity : ≥ 76%. TLC: Rf 0.28 (EtOAc/MeOH 10:1, KMnO4). MS (ESI): calcd for

C21H29N2O3 [M-H]+: 357.22, found: 357.30 (I = 100%). 1H-NMR (400 MHz, MeOD): δ = 7.96 (d, J = 8.0

Hz, 1H), 7.24 (d, J = 7.8 Hz, 1H), 7.16 (t, J = 7.7 Hz, 1H), 7.03 (t, J = 7.4 Hz, 1H), 4.35 (dd, J = 18.1,

8.1 Hz, 1H), 4.24 (dd, J = 17.8, 9.0 Hz, 1H), 4.13 (d, J = 6.0 Hz, 1H), 3.25 (t, J = 7.4 Hz, 2H), 3.22-

3.15 (m, 2H), 2.35 (ddd, J = 13.9, 6.6, 1.2 Hz, 1H), 2.18 (t, J = 7.2 Hz, 1H), 2.15-2.05 (m, 4H), 1.79

(dd, J = 9.0, 4.7 Hz, 1H), 1.42 (t, J = 4.7 Hz, 1H), 1.14 (ddd, J = 9.0, 4.7, 1.5 Hz, 1H), 0.97 (d, J = 1.7

Hz, 3H), 0.95 (d, J = 1.7 Hz, 3H).

Experimental Section

136

Amount : 3 mg. Purity : ≥ 92%. TLC: Rf 0.34 (EtOAc/MeOH 10:1, KMnO4). HR-MS (ESI): calcd for

C22H31N2O3 [M-H]+: 371.2335, found: 371.2348. 1H-NMR (400 MHz, MeOD): δ = 7.95 (d, J = 8.0 Hz,

1H), 7.23 (d, J = 7.8 Hz, 1H), 7.14 (t, J = 7.4 Hz, 1H), 7.02 (t, J = 7.4 Hz, 1H), 4.38-4.29 (m, 1H), 4.27-

4.18 (m, 1H), 4.12 (d, J = 6.0 Hz, 1H), 3.24 (d, J = 6.7 Hz, 2H), 3.21-3.14 (m, 2H), 2.32 (ddd, J = 13.9,

6.7, 1.3 Hz, 1H), 2.22 (dd, J = 8.5, 7.2 Hz, 2H), 2.16 (t, J = 6.8 Hz, 1H), 2.11 (dd, J = 13.8, 0.9 Hz, 1H),

1.78 (dd, J = 9.0, 4.7 Hz, 1H), 1.59-1.47 (m, 3H), 1.40 (t, J = 4.7 Hz, 1H), 1.22 (t, J = 7.2 Hz, 1H), 1.14

(ddd, J = 9.0, 4.6, 1.4 Hz, 1H), 0.91 (s, 3H), 0.90 (s, 3H).

Amount : 3.5 mg. Purity : ≥ 77%. TLC: Rf 0.42 (EtOAc/MeOH 30:1, KMnO4). MS (ESI): calcd for

C22H29N2O3 [M-H]+: 369.22, found: 369.00 (I = 100%). 1H-NMR (400 MHz, MeOD): δ = 7.94 (d, J = 8.2

Hz, 1H), 7.24 (d, J = 7.3 Hz, 1H), 7.14 (td, J = 7.8, 1.0 Hz, 1H), 7.02 (td, J = 7.4, 1.0 Hz, 1H), 4.38-

4.30 (m, 1H), 4.27-4.18 (m, 1H), 4.12 (d, J = 6.2 Hz, 1H), 3.25 (dd, J = 7.9, 6.9 Hz, 2H), 3.21-3.14 (m,

2H), 2.32 (ddd, J = 13.9, 6.7, 1.3 Hz, 1H), 2.72-2.63 (m, 1H), 2.33 (ddd, J = 13.9, 6.6, 1.3 Hz, 1H),

2.17 (t, J = 6.9 Hz, 1H), 2.13 (dd, J = 13.9, 1.1 Hz, 1H), 1.93-1.82 (m, 2H), 1.78 (dd, J = 9.1, 4.8 Hz,

1H), 1.76-1.67 (m, 4H), 1.65-1.55 (m, 2H), 1.41 (t, J = 4.7 Hz, 1H), 1.14 (ddd, J = 9.1, 4.5, 1.5 Hz, 1H).

HO

NH

O

O

N

1de

Amount : 4 mg. Purity : ≥ 87%. TLC: Rf 0.33 (EtOAc/MeOH 10:1, KMnO4). MS (ESI): calcd for

C25H29N2O3 [M-H]+: 405.22, found: 405.00 (I = 100%). 1H-NMR (400 MHz, MeOD): δ = 8.72 (dd, J =

4.5, 1.2 Hz, 1H), 8.42 (dd, J = 8.4, 1.3 Hz, 1H), 7.51 (dd, J = 8.6, 4.5 Hz, 1H), 7.23 (d, J = 7.3 Hz, 1H),

7.18-7.14 (m, 3H), 7.13-7.09 (m, 1H), 7.03 (td, J = 7.5, 1.0 Hz, 1H), 4.34-4.26 (m, 1H), 4.21-4.13 (m,

1H), 4.00 (dd, J = 6.4, 1.3 Hz, 1H), 3.27-3.14 (m, 4H), 2.93-2.88 (m, 2H), 2.54 (t, J = 7.6 Hz, 2H), 2.10

(t, J = 6.4 Hz, 1H), 2.03 (ddd, J = 14.0, 6.7, 1.3 Hz, 1H), 1.96 (dd, J = 14.1, 1.7 Hz, 1H), 1.63 (ddd, J =

9.2, 4.7, 1.0 Hz, 1H), 1.33 (t, J = 4.7 Hz, 1H), 1.09 (ddd, J = 9.2, 4.6, 1.5 Hz, 1H).

Experimental Section

137

Amount : 6 mg. Purity : ≥ 88%. TLC: Rf 0.18 (EtOAc/MeOH 40:1, KMnO4). MS (ESI): calcd for

C27H30N3O3 [M-H]+: 444.23, found: 443.98 (I = 100%). 1H-NMR (400 MHz, MeOD): δ = 8.00 (d, J = 8.0

Hz, 1H), 7.49 (d, J = 8.0 Hz, 1H), 7.28 (t, J = 8.0 Hz, 2H), 7.20 (t, J = 8.0 Hz, 1H), 7.07 (t, J = 8.0 Hz,

1H), 7.04-6.96 (m, 2H), 6.73 (t, J = 7.4 Hz, 1H), 4.25 (q, J = 8.4 Hz, 1H), 4.12 (q, J = 7.6 Hz, 1H), 3.90

(d, J = 6.2 Hz, 1H), 3.39 (dd, J = 13.5, 5.0 Hz, 1H), 3.21-3.02 (m, 4H), 2.83 (s, 2H), 2.71-2.56 (m, 2H),

2.10 (t, J = 6.2 Hz, 1H), 1.81-1.64 (m, 2H), 1.58 (dd, J = 8.0, 3.5 Hz, 1H), 1.31-1.23 (m, 1H), 1.09-1.04

(m, 1H). 13C-NMR (100 MHz, MeOD): δ =175.8, 173.1, 144.3, 137.9, 134.5, 128.5, 127.9, 126.0,

125.4, 123.6, 121.8, 119.9, 119.3, 118.1, 115.4, 111.2, 78.4, 51.0, 43.4, 39.8, 38.9, 38.3, 36.1, 29.2,

28.9, 22.5, 19.4.

Amount : 2.5 mg. Purity : ≥ 88%. TLC: Rf 0.28 (EtOAc/MeOH 10:1, KMnO4). HR-MS (ESI): calcd for

C23H31N2O3 [M-H]+: 383.2329, found: 383.2335. 1H-NMR (400 MHz, MeOD): δ = 7.98 (d, J = 7.9 Hz,

1H), 7.26 (d, J = 7.3 Hz, 1H), 7.18 (t, J = 7.7 Hz, 1H), 7.05 (t, J = 7.7 Hz, 1H), 4.41-4.33 (m, 1H), 4.30-

4.22 (m, 1H), 4.16 (d, J = 6.7 Hz, 1H), 3.27 (dd, J = 7.2, 3.10 Hz, 2H), 3.23-3.17 (m, 2H), 2.37 (ddd, J

= 13.6, 6.7, 1.0 Hz, 1H), 2.27-2.22 (m, 3H), 2.22-2.17 (m, 1), 2.14 (dd, J = 14.0, 1.1 Hz, 1H), 1.87-1.78

(m, 3H), 1.70-1.63 (m, 2H), 1.62-1.54 (m, 2H), 1.44 (t, J = 4.7 Hz, 1H), 1.30-1.20 (m, 2H), 1.17 (ddd, J

= 9.2, 4.6, 1.4 Hz, 1H).

Amount : 2 mg. Purity : ≥ 76%. TLC: Rf 0.49 (EtOAc/MeOH 20:1, KMnO4). HR-MS (ESI): calcd for

C24H24F2N2NaO3 [M-Na]+: 449.1647, found: 449.1648.

Experimental Section

138

HO

NH

O

O

N

1dg

N

SO

O

Amount : 7 mg. Purity : ≥ 96%. TLC: Rf 0.28 (EtOAc/MeOH 10:1, KMnO4). MS (ESI): calcd for

C27H31N3NaO5S [M-Na]+: 532.19, found: 531.91 (I = 100%). 1H-NMR (400 MHz, MeOD): δ = 7.96 (d, J

= 8.1 Hz, 1H), 7.52-7.49 (m, 1H), 7.39-7.36 (m, 2H), 7.28-7.16 (m, 3H), 7.06 (t, J = 7.4 Hz, 1H), 4.40-

4.32 (m, 1H), 4.29-4.23 (m, 1H), 3.95-3.89 (m, 5H), 3.51 (dd, J = 14.2, 6.7 Hz, 1H), 3.23-3.16 (m, 2H),

3.16-3.12 (m, 4H), 2.83 (s, 1H), 2.41-2.32 (m, 2H), 2.18 (d, J = 14.2 Hz, 1H), 1.89 (dd, J = 9.0, 4.7 Hz,

1H), 1.46 (t, J = 4.7 Hz, 1H), 1.18 (ddd, J = 9.1, 4.5, 1.3 Hz, 1H). 13C-NMR (100 MHz, MeOD): δ =

173.3, 169.7, 150.1, 136.9, 134.1, 130.8, 128.4, 125.7, 125.3, 120.6, 119.9, 118.1, 77.8, 61.2, 51.5,

51.2, 44.3, 39.5, 38.7, 36.8, 29.6, 28.9, 21.4, 19.9, 14.2.

Amount : 4 mg. Purity : ≥ 94%. TLC: Rf 0.16 (EtOAc/MeOH 10:1, KMnO4). MS (ESI): calcd for

C23H31N2O3 [M-H]+: 459.20, found: 459.16 (I = 100%). 1H-NMR (400 MHz, MeOD): δ = 8.26 (d, J = 7.9

Hz, 1H), 7.90-7.79 (m, 4H), 7.76-7.72 (m, 1H), 7.13 (d, J = 7.4 Hz, 1H), 7.04 (t, J = 7.7 Hz, 1H), 6.92

(t, J = 7.3 Hz, 1H), 4.27-4.19 (m, 1H), 4.16-4.08 (m, 1H), 4.03 (d, J = 6.4 Hz, 1H), 3.89 (d, J = 1.7 Hz,

2H), 3.27-3.13 (m, 2H), 3.10-3.04 (m, 2H), 2.24 (ddd, J = 13.8, 6.8, 1.1 Hz, 1H); 2.09 (t, J = 6.6 Hz,

1H), 2.00 (dd, J = 13.8, 1.1 Hz, 1H), 1.66 (dd, J = 9.0, 4.6 Hz, 1H), 1.30 (t, J = 4.8 Hz, 1H), 1.03 (ddd,

J = 9.0, 4.8, 1.4 Hz, 1H). 13C-NMR (100 MHz, MeOD): δ = 173.5, 171.7, 1144.9, 135.5, 133.8, 132.7,

129.0, 128.1, 127.4, 126.6, 126.0, 125.2, 117.9, 77.7, 51.2, 43.5, 40.9, 39.5, 36.0, 29.2, 29.0, 19.8,

14.4.

203f

202d was dissolved in dioxane, polymer-bound PPh3 added and the suspension slightly

shaken for 2 h (11:30 -13:30) at RT. Then ammonia was added and RM shaken overnight.

Experimental Section

139

Resin filtered off and washed with dichloromethane (3x 1 ml) and solvents evaporated to

dryness to give 7 mg (67%) of the crude amine as a yellow oil that was used for the

preparation of compounds 1fx .

Rf = 0.1 (EtOAc, KMnO4).

MS (ESI): calcd for C22H34ClN2O2Si [M-H]+: 421.2078, found: 420.99 (I = 100%).

Amount : 2.5 mg. Purity : ≥ 87%. TLC: Rf 0.31 (EtOAc/MeOH 20:1, KMnO4). MS (ESI): calcd for

C23H30ClN2O3 [M-H]+: 417.19, found: 416.97 (I = 100%). 1H-NMR (400 MHz, DMSO): δ = 7.43 – 7.36

(m, 2H), 6.78 (s, 1H), 6.66 (d, J = 8.6 Hz, 1H), 3.76 (dd, J = 17.1, 9.5 Hz, 1H), 3.69 (dd, J = 18.3, 9.6

Hz, 1H), 3.45 (d, J = 6.2 Hz, 1H), 2.63 (t, J = 8.3 Hz, 2H), 1.99 (s, 2H), 1.70 (dd, J = 13.5, 6.5 Hz, 1H),

1.61 (dd, J = 14.6, 7.3 Hz, 1H), 1.56 (d, J = 7.3 Hz, 2H), 1.45 (dd, J = 13.7, 5.4 Hz, 2H), 1.21 – 1.12

(m, 2H), 1.07 – 1.02 (m, 2H), 0.99 – 0.91 (m, 2H), 0.79 – 0.75 (m, 2H), 0.63 – 0.55 (m, 2H), 0.51 (dd, J

= 8.7, 3.4 Hz, 1H), 0.39 – 0.32 (m, 1H). 13C-NMR (100 MHz, DMSO): δ =172.0, 170.3, 142.1, 134.4,

126.9, 126.3, 125.0, 117.4, 74.8, 49.4, 48.3, 41.8, 41.5, 37.9, 36.7, 33.9, 31.9, 31.8, 278, 27.5, 24.5,

18.4.

Amount : 0.5 mg. Purity : ≥ 66%. TLC: Rf 0.16 (EtOAc/MeOH 20:1, KMnO4). HR-MS (ESI): calcd for

C22H24ClN3NaO4 [M-Na]+: 452.1348, found: 452.1341.

Experimental Section

140

Amount : 1 mg. Purity : ≥ 79%. TLC: Rf 0.26 (EtOAc/MeOH 20:1, KMnO4). MS (ESI): calcd for

C28H28ClN2NaO6 [M-Na]+: 545.15, found: 544.90 (I = 100%).

Amount : 1 mg. Purity : ≥ 82%. TLC: Rf 0.21 (EtOAc/MeOH 20:1, KMnO4). HR-MS (ESI): calcd for

C27H29ClN4NaO4 [M-Na]+: 531.1770, found: 531.1779.

Amount : 1.5 mg. Purity : ≥ 66%. TLC: Rf 0.30 (EtOAc/MeOH 20:1, KMnO4). MS (ESI): calcd for

C22H24ClN3NaO3S [M-Na]+: 468.11, found: 467.89 (I = 100%).

202b

((1S,3S,4R,5S)-4-(azidomethyl)-3-((tert-butyldimeth ylsilyl)oxy)bicyclo[3.1.0]hexan-1-

yl)(6-methoxy-3,4-dihydroisoquinolin-2(1H)-yl)metha none

3 (135 mg) was dissolved in 2 ml of CH2Cl2 and DIPEA (226 ul) was added under stirring and

after 5 min HATU (200 mg, 0.520 mmol) was added. In another flask the 6-methoxy-1,2,3,4-

Experimental Section

141

tetrahydroisoquinoline hydrochloride (198a) was dissolved in CH2Cl2 (2 ml) and DMF (1 ml),

then DIPEA (166 ul, 0.953 mmol) was added, after 10 min of stirring this mixture was added

to the solution of AS-115. The reaction mixture was stirred at RT for 22 h. It was diluted with

CH2Cl2 and poured on saturated aqueous NaHCO3 –solution. The water phase was

extracted with CH2Cl2 (3x 60 ml), the combined organic phases were dried over MgSO4 and

evaporated. Purification of the residue by flash column chromatography (Hex/EtOAc 2:1)

yielded 190 mg (96%) of the product as a colorless oil.

Rf = 0.32 (Hex/EtOAc 2:1, CPS).

1H-NMR (400 MHz, CDCl3): δ = 7.02 (d, J = 8.1 Hz, 1H), 6.76 (dd, J = 8.5, 2.6 Hz, 1H), 6.67

(d, J = 2.8 Hz, 1H), 4.76-4.55 (m, 2H), 4.17 (dd, J = 6.5, 1.1 Hz, 1H), 3.91-3.79 (m, 2H), 3.78

(s, 3H), 3.45 (dd, J = 12.3, 5.9 Hz, 1H), 3.11 (dd, J = 12.7, 9.5 Hz, 1H), 2.85 (t, J = 6.3 Hz,

1H), 2.26 (ddd, J = 14.1, 6.9, 1.1 Hz, 1H), 2.12 (dd, J = 9.7, 6.2 Hz, 1H), 1.90 (dd, J = 13.6,

1.1 Hz, 1H), 1.63 (dd, J = 9.1, 4.5 Hz, 1H), 1.40 (t, J = 4.6 Hz, 1H), 0.97 (J = 9.03, 4.40 Hz,

1H), 0.88 (s, 9H), 0.05 (s, 3H), 0.04 (s, 3H).

13C-NMR (100 MHz, CDCl3): δ = 171.9, 158.5, 127.2, 125.6, 113.7, 112.4, 76.4, 55.4, 54.4,

49.9, 39.9, 32.9, 26.9, 25.9, 21.2, 18.1, 17.7, -4.7, -4.9.

IR (film): υ 2951, 2929, 2856, 2096, 1635, 1505, 1462, 1431, 1389, 1257, 1225, 1168, 1084,

1037, 936, 862, 836, 808, 776, 670.

HR-MS (ESI): calcd for C24H37N4O3Si [M-H]+: 457.2629, found: 457.2638.

203b

((1S,3S,4R,5S)-4-(aminomethyl)-3-((tert-butyldimeth ylsilyl)oxy)bicyclo[3.1.0]hexan-1-

yl)(6-methoxy-3,4-dihydroisoquinolin-2(1H)-yl)metha none

AS-142-1 (115 mg) was dissolved in Tol/MeOH 1:1 (0.5 ml), Pd/C (54 mg, 10% Pd on

charcoal) was added and the mixture was stirred under a hydrogen atmosphere (1 bar) for 3

h. The reaction mixture was filtered through celite and the residue was washed with EtOAc.

The filtrate was concentrated under reduced pressure to afford 130 mg (0.302 mmol, >100%)

of the crude amine as a yellow oil that was immediately used for the preparation of

compounds 1bx .

Rf = 0.24 (EtOAc/MeOH 10:1)

Experimental Section

142

1H-NMR (400 MHz, Acetone): δ = 7.05 (d, J = 8.4 Hz, 1H), 6.78 – 6.71 (m, 2H), 4.71 – 4.52

(m, 2H), 4.45 (d, J = 5.2 Hz, 1H), 3.90 – 3.78 (m, 2H), 3.77 – 3.76 (m, 1H), 3.75 (s, 3H), 3.42

– 3.30 (m, 2H), 2.87 – 2.79 (m, 3H), 2.37 – 2.31 (m, 1H), 2.29 – 2.23 (m, 1H), 1.97 (d, J =

13.3 Hz, 1H), 1.58 (ddd, J = 5.6, 4.2, 0.4 Hz, 1H), 1.37 (t, J = 4.4 Hz, 1H), 0.89 (s, 9H), 0.08

(s, 3H), 0.07 (s, 3H).

13C-NMR (100 MHz, Acetone): δ = 129.7, 128.9, 126.2, 114.2, 113.1, 78.0, 55.7, 40.7, 28.2,

27.3, 26.3, 20.9, 20.7, 18.5, 18.2, -4.0, -4.6.

MS (ESI): calcd for C24H39N2O3Si [M-H]+: 431.27, found: 430.99 (I = 100%).

1bx

The compounds 1bx were prepared in the same way as exemplified on compounds 1ax.

Amount : 4 mg. Purity : ≥ 95%. MS (ESI): calcd for C24H33N2O4 [M-H]-: 413.24, found: 413.02 (I =

100%). 1H-NMR (400 MHz, DMSO): δ = 8.10-8.03 (m, 1H), 7.08 (d, J = 8.1 Hz, 1H), 6.76-6.71 (m, 2H),

4.88-4.78 (m, 1H), 4.64-4.46 (m, 1H), 3.95-3.89 (m, 1H), 3.83-3.73 (m, 1H), 3.69 (s, 3H), 3.06-2.99 (m,

1H), 2.97-2.87 (m, 1H), 2.82-2.70 (m, 2H), 2.09-2.02 (m, 1H), 1.98-1.93 (m, 1H), 1.86 (d, J = 13.5 Hz,

1H), 1.58-1.49 (m, 2H), 1.46-1.41 (m, 2H), 1.37-1.33 (m, 1H), 1.27 (q, J = 7.4 Hz, 2H), 1.22-1.18 (m,

1H), 0.91 (d, 6.6 Hz, 3H), 0.80 (d, J = 6.6 Hz, 3H). 13C-NMR (100 MHz, CDCl3): δ = 173.1, 171.1,

157.9, 127.3, 114.0, 112.2, 74.8, 57.6, 55.4, 49.4, 41.7, 34.7, 33.2, 32.1, 27.1, 23.2, 21.9, 19.2, 13.5.

Experimental Section

143

Amount : 3 mg. Purity : ≥ 67%. TLC: Rf 0.15 (EtOAc/MeOH 20:1, KMnO4). MS (ESI): calcd for

C24H31N2O4 [M-H]-: 411.23, found: 411.05 (I = 100%).

Amount : 3 mg. Purity : ≥ 94%. TLC: Rf 0.23 (EtOAc/MeOH 10:1, KMnO4). MS (ESI): calcd for

C27H31N2O4 [M-H]-: 447.23, found: 447.02 (I = 100%).

Amount : 3 mg. Purity : ≥ 97%. TLC: Rf 0.12 (EtOAc/MeOH 10:1, KMnO4). MS (ESI): calcd for

C29H32N3O4 [M-H]-: 486.24, found: 486.03 (I = 100%).

Amount : 2.5 mg. Purity : ≥ 85%. TLC: Rf 0.13 (EtOAc/MeOH 20:1, KMnO4). HR-MS (ESI): calcd for

C25H34N2NaO4 [M-Na]+: 449.2416, found: 449.2422.

Experimental Section

144

Amount : 3 mg. Purity : ≥ 88%. TLC: Rf 0.18 (EtOAc/MeOH 10:1, KMnO4). HR-MS (ESI): calcd for

C28H30N4NaO5 [M-Na]+: 525.2108, found: 525.2117.

HO

NH

O

N

O

OMe1ba

Amount : 7 mg. Purity : ≥ 78%. TLC: Rf 0.3 (EtOAc/MeOH 10:1, KMnO4). HR-MS (ESI): calcd for

C23H33N2O4 [M-H]+: 401.2435, found: 401.2442.

HO

NH

O

N

O

OMe

OH

1bt

Amount : 11 mg. Purity : ≥ 82%. TLC: Rf 0.2 (EtOAc/MeOH 1:1, KMnO4). HR-MS (ESI): calcd for

C22H31N2O5 [M-H]+: 403.2227, found: 403.2235.

Amount : 1 mg. Purity : ≥ 98%. TLC: Rf 0. (EtOAc/MeOH 10:1, KMnO4). HR-MS (MALDI): no signal

found. MS (ESI): calcd for C23H29N4O4 [M-H]+: 425.22, found: 425.01. (I = 65%).

Experimental Section

145

Amount : 3.5 mg. Purity : ≥ 85%. TLC: Rf 0.37 (EtOAc/MeOH 40:1, KMnO4). HR-MS (ESI): calcd for

C26H29F2N2O4 [M-H]+: 471.2090, found: 471.2098. 1H-NMR (400 MHz, DMSO): δ = 8.29-8.24 (m, 1H),

7.13-7.03 (m, 2H), 6.99-6.93 (m, 2H), 6.78-6.72 (m, 2H), 4.78 (s, br, 1H), 4.67-4.46 (m, 2H), 3.92 (d, J

= 6.5 Hz, 1H), 3.74-3.68 (m, 1H), 3.71 (s, 3H), 3.68-3.62 (m, 1H), 3.06-3.02 (m, 2H), 2.81-2.76 (m,

2H), 2.68 (s, 2H), 2.07 (dd, J = 13.4, 7.0 Hz, 1H), 1.97 (t, J = 6.7 Hz, 1H), 1.86 (d, J = 13.4 Hz, 1H),

1.50 (dd, J = 8.5, 4.4 Hz, 1H), 1.17-1.13 (m, 1H), 0.91-0.87 (m, 1H).

Amount : 4 mg. Purity : ≥ 70%. TLC: Rf 0.12 (EtOAc/MeOH 10:1, KMnO4). HR-MS (ESI): no signal

found. MS (ESI): calcd for C24H30N3O5 [M-H]+: 440.22, found: 440.00.

Amount : 11 mg. Purity : ≥ 79%. TLC: Rf 0.34 (EtOAc/MeOH 40:1, KMnO4). HR-MS (ESI): calcd for

C29H36N3O6S [M-H]+: 554.2319, found: 554.2306.

Experimental Section

146

Amount : 2 mg. Purity : ≥ 76%. TLC: Rf 0.31 (EtOAc/MeOH 40:1, KMnO4). HR-MS (ESI): calcd for

C24H29N2O5 [M-H]+: 425.2071, found: 425.2081. 1H-NMR (400 MHz, MeOD): δ = 7.97 (s, br, 1H), 7.40-

7.32 (m, 1H), 7.07 (d, J = 8.4 Hz, 1H), 6.79-6.71 (m, 2H), 6.34-6.25 (m, 1H), 6.22-6.18 (m, 1H), 5.05-

4.60 (s, br, 2H), 4.07 (dd, J = 6.6, 0.9 Hz, 1H), 3.80-3.71 (m, 2H), 3.76 (s, 3H), 3.70-3.62 (m, 1H), 3.56

(s, 2H), 3.25-3.18 (m, 1H), 2.90-2.84 (m, 2H), 2.20-2.12 (m, 2H), 1.98 (dd, J = 14.1, 0.9 Hz, 1H), 1.63

(dd, J = 9.0, 4.6 Hz, 1H), 1.35 (t, J = 4.6 Hz, 1H), 1.04-0.97 (m, 1H). 13C-NMR (100 MHz, MeOD): δ =

153.4, 146.2, 116.8, 116.3, 114.8, 111.5, 80.6, 58.2, 45.9, 43.0, 41.9, 39.3, 36.5, 34.3, 22.0, 21.4.

Note: Not all carbon peaks were found.

Amount : 2.5 mg. Purity : ≥ 60%. TLC: Rf 0.29 (EtOAc/MeOH 40:1, KMnO4). HR-MS (ESI): calcd for

C30H33N2O7 [M-H]+: 533.2282, found: 533.2280. 1H-NMR (400 MHz, MeOD): δ = 7.68 (d, J = 8.9 Hz,

1H), 7.08 (d, J = 8.4 Hz, 1H), 6.89 (dd, J = 8.9, 2.4 Hz, 1H), 6.85-6.80 (m, 1H), 6.77 (dd, J = 8.4, 2.7

Hz, 1H), 6.74-6.72 (m, 1H), 6.28 (s, 1H), 4.04 (d, J = 6.5 Hz, 1H), 3.89-3.84 (m, 4H), 3.84 (s, 2H), 3.77

(s, 6H), 3.39-3.37 (m, 1H), 3.36-3.33 (m, 1H), 2.16 (t, J = 6.0 Hz, 1H), 2.05 (dd, J = 13.5, 6.3 Hz, 1H),

1.92 (dd, J = 14.0, 1.2 Hz, 1H), 1.58 (dd, J = 8.7, 4.2 Hz, 1H), 1.34-1.29 (m, 1H), 1.02-1.97 (m, 1H).

Amount : 7 mg. Purity : ≥ 92%. TLC: Rf 0.1 (EtOAc/MeOH 10:1, KMnO4). MS (ESI): calcd for

C29H35N4O5 [M-H]+: 519.26, found: 519.02 (I = 100%).

Experimental Section

147

Amount : 1 mg. Purity : ≥ 30%. MS (ESI): calcd for C24H30N3NaO4S [M-Na]+: 478.18, found: 477.96 (I =

100%).

202c

(1S,3S,4R,5S)-4-(azidomethyl)-3-((tert-butyldimethyl silyl)oxy)-N-(thiazol-2-

yl)bicyclo[3.1.0]hexane-1-carboxamide

3 (70 mg, 0.225 mmol) was dissolved in CH2Cl2 (2 ml) and a few drops of DMF, then DIPEA

(192 ul, 1.124 mmol) and after 5 min HATU (96 mg, 0.248 mmol) were added. After 5 min 2-

amino-thiazole (199c, 30 mg) was added and the reaction mixture was stirred at RT

overnight. The solvents were evaporated and purification by flash chromatography

(CH2Cl2/MeOH 100:1) afforded 89 mg (0.226 mmol, >100%) of the product as a yellow oil.

Rf = 0.29 (CH2Cl2); 0.48 (CH2Cl2/MeOH 50:1)

1H-NMR (400 MHz, CDCl3): δ = 7.44 (d, J = 3.4 Hz, 1H), 6.96 (d, J = 3.1 Hz, 1H), 4.20 (d, J =

6.5 Hz, 1H), 3.36 (dd, J = 12.4, 5.7 Hz, 1H), 3.13 (dd, J = 12.4, 8.3 Hz, 1H), 2.48 (ddd, J =

13.1, 6.5, 1.6 Hz, 1H), 2.19 (dd, J = 8.3, 5.7 Hz, 1H), 1.98 (d, J = 13.2 Hz, 1H), 1.92 (ddd, J =

9.1, 5.2, 1.1 Hz, 1H), 1.69 – 1.62 (m, 1H), 1.54 (ddd, J = 9.1, 4.4, 1.7 Hz, 1H), 0.87 (s, 9H),

0.05 (s, 3H), 0.05 (s, 3H).

13C-NMR (100 MHz, CDCl3): δ = 170.9, 162.0, 137.5, 113.9, 99.3, 75.8, 53.8, 49.3, 39.5,

36.3, 32.2, 25.7, 21.3, 17.9, -5.0.

IR (film): ν 3667, 2953, 2929, 2858, 2358, 2338, 2099, 1672, 1537, 1470, 1362, 1320, 1254,

1162, 1084, 936, 839, 778.

HR-MS (ESI): calcd for C17H28N5O2SSi [M-H]+: 394.1733, found: 394.1728.

Experimental Section

148

203c

202c (88 mg) was dissolved in MeOH (3 ml), then 10% Pd/C (48 mg) and Et3N (6 ul) were

added. The reaction mixture was stirred vigorously under a hydrogen atmosphere (1 bar) for

6 h. The catalyst was filtered off and the filtrate was evaporated under reduced pressure. The

crude amine (75 mg, 91%) was used immediately to prepare compounds 1cx .

Rf = 0.1 (CH2Cl2/MeOH 50:1)

IR (film): ν 3302, 2952, 2927, 2860, 2100, 1666, 1538, 1320, 1252, 1163, 1081, 1027, 938,

837, 776, 708, 620.

MS (ESI): calcd for C17H30N3O2SSi [M-H]+: 368.1828, found: 368.05 (I = 100%).

1cx

The compounds 1cx were prepared in the same way as exemplified on compounds 1ax.

Amount : 3 mg. Purity : ≥ 46%. TLC: Rf 0.53 (EtOAc/MeOH 10:1, KMnO4). HR-MS (ESI): calcd for

C16H24N3O3S [M-H]+: 338.1533, found: 338.1533.

Amount : 3 mg. Purity : ≥ 78%. TLC: Rf 0.41 (EtOAc/MeOH 10:1, KMnO4). HR-MS (ESI): calcd for

C17H26N3O3S [M-H]+: 352.1689, found: 352.1695.

Experimental Section

149

Amount : 2 mg. Purity : ≥ 57%. TLC: Rf 0.19 (EtOAc/MeOH 10:1, KMnO4). HR-MS (ESI): calcd for

C17H24N3O3S [M-H]+: 350.1533, found: 350.1531.

Amount : 3 mg. Purity : ≥ 75%. TLC: Rf 0.15 (EtOAc/MeOH 10:1, KMnO4). HR-MS (ESI): calcd for

C20H24N3O3S [M-H]+: 386.1533, found: 386.1537.

Amount : 3 mg. Purity : ≥ 91%. TLC: Rf 0.5 (EtOAc/MeOH 5:1 with 0.5% Et3N, KMnO4). HR-MS (ESI):

calcd for C22H25N4O3S [M-H]+: 425.1642, found: 425.1635.

Amount : 2 mg. Purity : ≥ 90%. TLC: Rf 0.16 (EtOAc/MeOH 10:1, KMnO4). HR-MS (ESI): calcd for

C18H26N3O3S [M-H]+: 364.1689, found: 364.1692.

Amount : 12 mg. Purity : ≥ 96%. TLC: Rf 0.1 (EtOAc/MeOH 10:1, KMnO4). MS (ESI): calcd for

C21H22N5O4S [M-H]-: 440.14, found: 439.99 (I = 100%).

Experimental Section

150

Amount : 3 mg. Purity : ≥ 43%. TLC: Rf 0.17 (EtOAc/MeOH 5:1 with 0.5% Et3N, KMnO4). HR-MS

(ESI): calcd for C17H22N5O3S [M-H]+: 376.1438, found: 376.1435.

Amount : 3 mg. Purity : ≥ 57%. TLC: Rf 0.1 (EtOAc/MeOH 5:1 with 0.5% Et3N, KMnO4). HR-MS (ESI):

calcd for C16H20N5O3S [M-H]+: 362.1281, found: 362.1282.

Amount : 2 mg. Purity : ≥ 73%. TLC: Rf 0.5 (EtOAc/MeOH 10:1, KMnO4). HR-MS (ESI): calcd for

C19H20F2N3O3S [M-H]+: 408.1188, found: 408.1195.

Amount : 11 mg. Purity : ≥ 80%. TLC: Rf 0.5 (EtOAc/MeOH 10:1, KMnO4). HR-MS (ESI): calcd for

C22H27N4O5S2 [M-H]+: 491.1417, found: 491.1409. 1H-NMR (400 MHz, MeOD): δ = 7.43 (d, J = 3.6 Hz,

1H), 7.43-7.41 (m, 1H), 7.30.7.27 (m, 2H), 7.18-7.14 (m, 1H), 7.09 (d, J = 3.6 Hz, 1H), 5.17 (s, br, 1H),

4.60 (s, 1H), 4.31 (d, J = 6.6 Hz, 1H), 3.91-3.87 (m, 4H), 3.41 (dd, J = 10.8, 7.2 Hz, 1H), 3.21 (q, J =

7.3 Hz, 1H), 3.17-3.13 (m, 4H), 2.65 (ddd, J = 14.0, 6.8, 1.5 Hz, 1H), 2.36 (t, J = 6.5 Hz, 1H), 2.10-2.05

(m, 1H), 1.94 (d, J = 13.4 Hz, 1H), 1.59 (t, J = 4.9 Hz, 1H), 1.52 (ddd, J = 9.1, 4.4, 1.6 Hz, 1H). 13C-

NMR (100 MHz, MeOD): δ = 170.8, 149.5, 136.7, 130.7, 120.4, 119.7, 115.8, 114.5, 77.4, 51.4, 47.9,

43.6, 37.0, 34.1, 33.7, 21.7.

Experimental Section

151

Amount : 14 mg. Purity : ≥ 84%. TLC: Rf 0.39 (EtOAc/MeOH 10:1, KMnO4). HR-MS (ESI): calcd for

C17H20N3O4S [M-H]+: 362.1169, found: 362.1168. 1H-NMR (400 MHz, MeOD): δ = 7.43 (d, J = 3.6 Hz,

1H), 7.37 (dd, J = 1.9, 0.8 Hz, 1H), 7.08 (d, J = 3.6 Hz, 1H), 6.30 (dd, J = 3.1, 1.9 Hz, 1H), 6.18 (dd, J

= 3.3, 0.8 Hz, 1H), 5.15 (s, br, 1H), 4.58 (s, br, 1H), 4.16 (d, J = 6.7 Hz, 1H), 3.22-3.14 (m, 4H), 2.59

(ddd, J = 14.0, 6.8, 1.7 Hz, 1H), 2.19 (t, J = 7.3 Hz, 1H), 2.02 (dd, J = 14.0, 0.5 Hz, 1H), 1.95 (ddd, J =

9.0, 5.2, 0.9 Hz, 1H), 1.55 (t, J = 4.8 Hz, 1H), 1.49 (ddd, J = 9.0, 4.4, 1.7 Hz, 1H). 13C-NMR (100 MHz,

MeOD): δ = 173.7, 171.4, 160.4, 150.7, 143.9, 139.1, 115.1, 111.6, 109.3, 76.6, 51.4, 47.9, 43.0, 36.6,

33.9, 33.7, 21.7.

Amount : 6 mg. Purity : ≥ 86%. TLC: Rf 0.28 (EtOAc/MeOH 10:1, KMnO4). HR-MS (ESI): calcd for

C23H23N3NaO6S [M-H]+: 492.12, found: 491.95.

Amount : 8 mg. Purity : ≥ 90%. TLC: Rf 0.17 (EtOAc/MeOH 6:1 with 0.5% Et3N, KMnO4). MS (ESI):

calcd for C22H26N5O4S [M-H]+: 456.17, found: 456.00 (I = 100%).

208

(1S,3S,4R,5S)-4-(azidomethyl)-3-(benzyloxy)-N-(2-(( 4-

methoxyphenyl)amino)ethyl)bicyclo[3.1.0]hexane-1-ca rboxamide

194 (216 mg) was dissolved in CH2Cl2 (8 ml) and DMF (1 ml), and DIPEA (0.9 ml), after 5min

HATU (310 mg) was added, and the N1-(4-methoxyphenyl)ethane-1,2-diamine (207g, 150

Experimental Section

152

mg) was added and the reaction mixture was stirred for 22 h at RT. The solvents were

evaporated and the crude product was purified by flash chromatography (EE/Hex 4:1) to

afford 239 mg (73%) of the product as a very slightly brown oil.

Rf = 0.28 (CH2Cl2/MeOH 50:1)

1H-NMR (400 MHz, MeOD): δ = 7.36 – 7.24 (m, 5H), 6.78 – 6.70 (m, 2H), 6.69 – 6.61 (m,

2H), 4.42 (s, 2H), 3.89 (d, J = 7.0 Hz, 1H), 3.69 (s, 3H), 3.44 – 3.29 (m, 3H), 3.23 – 3.15 (m,

3H), 2.43 – 2.29 (m, 2H), 2.06 (d, J = 14.0 Hz, 1H), 1.72 (ddd, J = 8.8, 5.2, 1.0 Hz, 1H), 1.31

– 1.27 (m, 2H).

13C-NMR (400 MHz, MeOD): δ = 176.5, 153.8, 143.9, 139.9, 129.7, 128.8, 128.5, 115.9,

115.4, 84.7, 71.8, 56.2, 55.0, 47.9, 45.6, 40.7, 34.4, 33.7, 31.6, 19.7.

MS (ESI): calcd for C24H30N5O3 [M-H]+: 436.2349, found: 436.09 (I = 100%).

209

(1S,3S,4R,5S)-4-(aminomethyl)-3-(benzyloxy)-N-(2-(( 4-methoxyphenyl)amino)ethyl)

bicyclo[3.1.0]hexane-1-carboxamide

208 (90 mg) was dissolved in EtOH (2.5 ml), Lindlar’s catalyst (88 mg, ~5% Pd on CaCO3)

was added and the reaction mixture was stirred under an atmosphere of hydrogen (6 bar) for

12 h. The solution was filtrated over celite and the filter cake was washed with MeOH/EtOAc.

The solvents were evaporated to yield 83 mg of the crude product (98%) as a slightly brown

oil that was immediately used for the preparation of compounds 210x.

Rf = 0.10 (CH2Cl2/MeOH 50:1)

MS (ESI): calcd for C24H32N3O3 [M-H]+: 410.2444, found: 410.09 (I = 100%).

210x

The compounds 210x were prepared in the same way as described for compounds 1ax,

without the deprotection step employing DOWEX-50W.

Experimental Section

153

Amount : 3 mg. Purity : ≥ 65%. TLC: Rf 0.17 (EtOAc/MeOH 20:1, CPS). MS (ESI): calcd for

C32H36F2N3O4 [M-H]+: 564.27, found: 563.92 (I = 100%).

Amount : 2.5 mg. Purity : ≥ 60%. TLC: Rf 0.26 (EtOAc/MeOH 20:1, CPS). MS (ESI): calcd for

C30H37N4O5 [M-H]+: 533.28, found: 532.93 (I = 100%).

Amount : 15 mg. Purity : ≥ 90%. TLC: Rf 0.23 (EtOAc/MeOH 20:1, CPS). HR-MS (ESI): calcd for

C35H43N4O6S [M-H]+: 647.2898, found: 647.2906. 1H-NMR (400 MHz, MeOD): δ =

Amount : 11 mg. Purity : ≥ 22%. TLC: Rf 0.24 (EtOAc/MeOH 20:1, CPS). HR-MS (ESI): calcd for

C30H36N3O5 [M-H]+: 518.2649, found: 518.2654.

Amount : 12 mg. Purity : ≥ 40%. TLC: Rf 0.17 (EtOAc/MeOH 20:1, CPS). HR-MS (ESI): calcd for

C36H40N3O7 [M-H]+: 626.2861, found: 626.2873.

Experimental Section

154

Amount : 9 mg. Purity : ≥ 90%. TLC: Rf 0.13 (EtOAc/MeOH 20:1, CPS). HR-MS (ESI): calcd for

C34H38N5O5 [M-H]+: 596.2867, found: 596.2883. 1H-NMR (500 MHz, DMSO): δ = 12.60 (s, 1H), 8.41 (t,

J = 5.6 Hz, 1H), 8.25 (d, J = 7.6 Hz, 1H), 7.87 (d, J = 3.6 Hz, 2H), 7.85-7.78 (m, 1H), 7.60 (t, J = 5.6

Hz, 1H), 7.37-7.32 (m, 2H), 7.30-7.23 (m, 3H), 6.69 (d, J = 8.6 Hz, 2H), 6.52 (d, J = 8.6 Hz, 2H), 5.19

(t, J = 5.1 Hz, 1H), 4.28 (s, 2H), 3.82 (s, 2H), 3.76 (d, J = 6.4 Hz, 1H), 3.61 (s, 3H), 3.26-3.18 (m, 2H),

3.04-3.95 (m, 4H), 2.37 (dd, J = 14.0, 6.6 Hz, 1H), 2.23 (t, J = 7.6 Hz, 1H), 1.96 (d, J = 14.1 Hz, 1H),

1.61 (dd, J = 8.6, 4.9 Hz, 1H), 1.19 (dd, J = 9.0, 2.8 Hz, 1H), 1.11 (t, J = 4.2 Hz, 1H). 13C-NMR (500

MHz, DMSO): δ = 172.6, 168.5, 159.4, 150.6, 143.0, 141.7, 138.2, 133.1, 131.7, 129.5, 128.2, 127.6,

127.3, 127.2, 126.0, 125.6, 114.7, 112.9, 82.7, 69.7, 55.4, 46.6, 43.3, 41.6, 38.6, 33.0, 31.9, 29.6,

18.2.

Amount : 6 mg. Purity : ≥ 68%. TLC: Rf 0.16 (EtOAc/MeOH 10:1 with 0.5% Et3N, CPS). HR-MS (ESI):

calcd for C35H42N5O5 [M-H]+: 612.3180, found: 612.3166.

1gp

210gp (5 mg) was dissolved in CH2Cl2 and cooled to -78°C. BCl 3 (131 ul, 1M sol. in CH2Cl2) was

added dropwise and the reaction mixture was stirred for 5 h at -78°C. The reaction mixture was slowly

warmed to -25°C, quenched with MeOH (250 ul) and st irred at RT overnight. The solvents were

evaporated and the crude product was purified by flash chromatography (EtOAc/MeOH 5:1 0.5%

Et3N) to give 1 mg (24%) of 2gp .

TLC: Rf 0.10 (EtOAc/MeOH 6:1 with 0.5% Et3N, CPS). MS (ESI): calcd for C28H36N5O5 [M-H]+:

522.27, found: 521.86 (I = 100%).

Experimental Section

155

Amount : 12 mg. Purity : ≥ 93%. TLC: Rf 0.42 (EtOAc/MeOH 10:1, CPS). MS (ESI): calcd for

C34H48N3O5 [M-H]+: 578.36, found: 578.10 (I = 100%).

Amount : 15 mg. Purity : ≥ 96%. TLC: Rf 0.46 (EtOAc/MeOH 10:1, CPS). MS (ESI): calcd for

C36H52N3O5 [M-H]+: 606.39, found: 606.09 (I = 100%).

Amount : 12 mg. Purity : ≥ 97%. TLC: Rf 0.40 (EtOAc/MeOH 10:1, CPS). MS (ESI): calcd for

C36H47N3NaO5 [M-H]+: 624.34, found: 624.07 (I = 100%).

NH

O

210e

BnO

O

NH

HN

O

Amount : 20 mg. Purity : ≥ 80%. TLC: Rf 0.46 (EtOAc/MeOH 10:1, CPS). MS (ESI): calcd for

C33H40N3O4 [M-H]+: 542.30, found: 542.20 (I = 60%). 1H-NMR (500 MHz, DMSO): δ = 7.83 (t, J = 5.5

Hz, 1H), 7.39 (t, J = 5.5 Hz, 1H), 7.13-7.07 (m, 2H), 7.05-7.00 (m, 6H), 6.98-6.94 (m, 2H), 6.49 (d, J =

8.9 Hz, 2H), 6.33 (d, J = 8.9 Hz, 2H), 4.96 (s, br, 1H), 4.07 (d, J = 6.0 Hz, 1H), 3.54-3.48 (m, 2H), 3.40

(s, 3H), 3.08-2.96 (m, 2H), 2.81 (t, J = 6.3 Hz, 2H), 2.51 (s, 1H), 2.46 (s, 1H), 2.19-2.11 (m, 3H), 1.98

(t, J = 7.5 Hz, 1H), 1.75 (d, J = 14.1 Hz, 1H), 1.37 (dd, J = 8.3, 4.9 Hz, 1H), 0.99-0.95 (m, 1H), 0.89 (t,

J = 4.1 Hz, 1H). 13C-NMR (500 MHz, DMSO): δ = 173.0, 171.8, 150.9, 143.3, 141.4, 138.7, 128.4,

127.5, 125.7, 114.9, 112.9, 82.5, 69.9, 55.3, 46.6, 43.3, 41.3, 38.6, 38.2, 36.9, 35.7, 32.9, 31.8, 30.9,

29.7.

Experimental Section

156

Amount : 16 mg. Purity : ≥ 79%. TLC: Rf 0.12 (EtOAc/MeOH 10:1, CPS). MS (ESI): calcd for

C30H41N4O5 [M-H]+: 537.31, found: 537.30 (I = 60%).

NH

O

BnO

O

NH

N

O

210d

O

Amount : 12 mg. Purity : ≥ 97%. TLC: Rf 0.41 (EtOAc/MeOH 10:1, CPS). MS (ESI): calcd for

C38H51N3NaO5 [M-H]+: 652.37, found: 652.30 (I = 100%).

Amount : 10 mg. Purity : ≥ 34%. TLC: Rf 0.10 (EtOAc/MeOH 10:1, CPS). MS (ESI): calcd for

C29H36N5O4 [M-H]+: 518.28, found: 518.20 (I = 100%).

Amount : 25 mg. Purity : ≥ 60%. TLC: Rf 0.16 (EtOAc/MeOH 20:1, CPS). MS (ESI): calcd for

C35H43N4O5 [M-H]+: 599.32, found: 599.30 (I = 100%).

Amount : 14 mg. Purity : ≥ 94%. TLC: Rf 0.16 (EtOAc/MeOH 10:1, CPS). MS (ESI): calcd for

C30H35N4O4S [M-H]-: 547.24, found: 547.20 (I = 75%).

Experimental Section

157

202e

((1S,3S,4R,5S)-4-(azidomethyl)-3-((tert-butyldimeth ylsilyl)oxy) icycle[3.1.0]hexan-1-

yl)(3-hydroxy-7,8-dihydro-1,6-naphthyridin-6(5H)-yl )methanone

3 (25 mg, 0.080 mmol) was dissolved in CH2Cl2 (1 ml) and DIPEA (42 ul, 0.241 mmol) was

added under stirring and after 5 min HATU (37 mg, 0.096 mmol) was added. In another flask

the 5,6,7,8-tetrahydro-1,6-naphthyridin-3-ol hydrochloride was dissolved in CH2Cl2 (0.5 ml)

and DMF (0.5 ml), then DIPEA (31 ul, 0.180 mmol) was added, after 10 min of stirring this

mixture was added to the solution of AS-115. The reaction mixture was stirred at RT for 22 h.

It was diluted with CH2Cl2 and poured on saturated aqueous NaHCO3 -solution. The water

phase was extracted with CH2Cl2 (3x 20 ml), the combined organic phases were dried over

MgSO4 and evaporated. Purification of the residue by flash column chromatography

(Hex/EtOAc 1:1--> EtOAc/MeOH 95:5) yielded 32 mg (0.072 mmol, 90%) of the product as a

colorless oil.

Rf = 0.29 (EtOAc/MeOH 20:1)

1H-NMR (400 MHz, CDCl3): δ = 8.13 (d, J = 2.6 Hz, 1H), 7.03 (d, J = 2.5 Hz, 1H), 4.69 (q, J =

17.3 Hz, 2H), 4.17 (d, J = 6.0 Hz, 1H), 4.01 (t, J = 5.9 Hz, 1H), 3.95 (s, 1H), 3.45 (dd, J =

12.5, 6.1 Hz, 1H), 3.12 (dd, J = 12.4, 9.2 Hz, 1H), 2.99 (t, J = 5.6 Hz, 2H), 2.29 (dd, J = 13.4,

6.4 Hz, 1H), 2.13 (dd, J = 9.0, 6.3 Hz, 1H), 1.92 (d, J = 13.4 Hz, 1H), 1.66 (dd, J = 8.9, 4.6

Hz, 1H), 1.44 (t, J = 4.6 Hz, 1H), 1.00 (dd, J = 8.6, 4.1 Hz, 1H), 0.88 (s, 9H), 0.06 (s, 3H),

0.05 (s, 3H).

13C-NMR (100 MHz, CDCl3): δ = 172.4, 153.1, 136.3, 129.8, 121.7, 76.1, 54.0, 50.4, 40.0,

38.6, 32.5, 30.8, 27.1, 26.0, 18.2, 17.9, -4.5, -4.9.

IR (film): ν 2952, 2928, 2856, 2359, 2341, 2098, 1617, 1455, 1388, 1302, 1258, 1168, 1087,

1038, 928, 837, 766, 669.

MS (ESI): cacld for C22H34N5O3Si [M-H]+: 444.24, found: 444.12 (I = 100%).

Experimental Section

158

203e

((1S,3S,4R,5S)-4-(aminomethyl)-3-((tert-butyldimeth ylsilyl)oxy)bicyclo[3.1.0]hexan-1-

yl)(3-hydroxy-7,8-dihydro-1,6-naphthyridin-6(5H)-yl )methanone

202e (88 mg) was dissolved in Tol/MeOH 1:1 (2 ml), Pd/C (42 mg) was added and the

reaction mixture was stirred under H2 (2 bar) for 3 h. The mixture was filtered through celite,

the residue was washed with EtOAc, and the solvents were evaporated to give 72 mg (87%)

of product (white foam) that was used to prepare compounds 1ex.

Rf = 0.1 (EtOAc/MeOH 20:1)

MS (ESI): cacld for C22H36N3O3Si [M-H]+: 418.2526, found: 418.20 (I = 100%).

1ex

The compounds 1ex were prepared as described for compounds 1ax.

Amount : 3 mg. Purity : ≥ 85%. TLC: Rf 0.16 (EtOAc/MeOH 10:1, CPS). MS (ESI): calcd for

C27H33N4O5 [M-H]+: 493.25, found: 493.30 (I = 80%).

Experimental Section

159

Amount : 4 mg. Purity : ≥ 85%. TLC: Rf 0.11 (EtOAc/MeOH 10:1, CPS). MS (ESI): calcd for

C27H31N4O6S [M-H]-: 539.20, found: 539.30 (I = 100%).

Amount : 9 mg. Purity : ≥ 79%. TLC: Rf 0.10 (EtOAc/MeOH 10:1, CPS). HR-MS (ESI): calcd for

C22H26N3O5 [M-H]+: 412.1867, found: 412.1877.

NH

O

1ea

N

N

O

HO

O

O

Amount : 4 mg. Purity : ≥ 92%. TLC: Rf 0.41 (EtOAc/MeOH 10:1, KMnO4). MS (ESI): calcd for

C26H38N3O5 [M-H]+: 472.28, found:472.10 (I = 100%).

Amount : 3.5 mg. Purity : ≥ 95%. TLC: Rf 0.15 (EtOAc/MeOH 6:1 with 0.5% Et3N, CPS). MS (ESI):

calcd for C22H32N3O4 [M-H]+: 402.24, found:402.10 (I = 100%).

Experimental Section

160

Amount : 2.5 mg. Purity : ≥ 65%. TLC: Rf 0.15 (EtOAc/MeOH 6:1 with 0.5% Et3N, CPS). MS (ESI):

calcd for C22H30N3O4 [M-H]+: 400.22, found:400.10 (I = 100%).

Amount : 2 mg. Purity : ≥ 68%. TLC: Rf 0.14 (EtOAc/MeOH 10:1, KMnO4). HR-MS (ESI): calcd for

C25H29N3NaO4 [M-H]+: 458.2050, found:458.2056.

Amount : 4.5 mg. Purity : ≥ 60%. TLC: Rf 0.10 (EtOAc/MeOH 6:1 with 0.5% Et3N, CPS). HR-MS

(ESI): calcd for C27H31N4O4 [M-H]+: 475.2340, found:475.2348.

Amount : 7 mg. Purity : ≥ 95%. TLC: Rf 0.07 (EtOAc/MeOH 10:1, KMnO4). HR-MS (ESI): calcd for

C27H32N5O5 [M-H]+: 506.2398, found: 506.2401.

Experimental Section

161

Amount : 4 mg. Purity : ≥ 77%. TLC: Rf 0.43 (EtOAc/MeOH 10:1, CPS). MS (ESI): calcd for

C24H26F2N3NaO4 [M-Na]+: 480.17, found: 480.00 (I = 100%).

5.2.1.4 Synthesis of library members of general str ucture 2

233

((1S,3S,4R,5S)-3-((tert-butyldimethylsilyl)oxy)-4-( ((tert-butyldimethylsilyl)oxy)methyl)

bicyclo[3.1.0]hexan-1-yl)methanol

185 (60 mg, 0.145 mmol) was dissolved in CH2Cl2 (1 ml) and cooled to -78°C and DIBALH

(435 ul, 0.435 mmol) was added dropwise. The reaction mixture was stirred at -78°C for 4 h,

then it was quenched at this temperature with saturated aqueous NaHCO3 –solution and

extracted with CH2Cl2. The organic phases were dried and evaporated to afford 56 mg (0.145

mmol, 100%) of the product as a colorless oil.

Rf = 0.61 (Hex/EtOAc 1:1)

1H-NMR (400 MHz, CDCl3): δ = 4.14 (d, J = 6.5 Hz, 1H), 3.74-3.70 (m, 1H), 3.63 (d, J = 11.1

Hz, 1H), 3.54 (dd, J = 10.1, 5.1 Hz, 1H), 3.43-3.38 (m, 2H), 2.15 (ddd, J = 13.5, 6.6, 1.6 Hz,

1H), 2.00 (dd, J = 6.6, 5.4 Hz, 1H), 1.69 (d, J = 13.5 Hz, 1H), 1.07 (t, J = 4.3 Hz, 1H), 0.96

(ddd, J = 9.1, 3.9, 1.2 Hz, 1H), 0.88 (s, 9H), 0.83 (s, 9H), 0.53 (ddd, J = 8.7, 4.4, 1.7 Hz, 1H),

0.04 (s, 3H), 0.03 (s, 3H), -0.01 (s, 3H), -0.01 (s, 3H).

13C-NMR (100 MHz, CDCl3): δ = 76.4, 68.1, 67.1, 65.7, 38.6, 31.8, 26.0, 25.8, 24.5, 18.6,

17.9, 15.3, -4.6, -4.8, -5.4, -5.7.

IR (film): ν 3358, 2928, 2856, 2359, 1471, 1462, 1255, 1082, 834, 773, 665.

MS (ESI): calcd for C20H42NaO3Si2 [M-Na]+: 409.26, found: 409.14 (I = 100%).

Experimental Section

162

234

(1S,3S,4R,5S)-3-((tert-butyldimethylsilyl)oxy)-4-(( (tert-butyldimethylsilyl)oxy)methyl)

bicyclo[3.1.0]hexane-1-carbaldehyde

233 (56 mg, 0.145 mmol) was dissolved in CH2Cl2, pyridinium chlorochromate (94 mg, 0.435

mmol) was added and the reaction mixture was stirred at RT for 5 h, then it was diluted with

EtOAc/Hex 1:5 and filtered through a short column (EtOAc/Hex 1:5) to afford 51 mg of the

product (0.133 mmol, 91%) as a slightly yellow oil.

Rf = 0.40 (Hex/EtOAc 10:1)

1H-NMR (400 MHz, CDCl3): δ = 8.84 (s, 1H), 4.20 (d, J = 6.5 Hz, 1H), 3.74 (dd, J = 10.3, 6.3

Hz, 1H), 3.33 (dd, J = 10.3, 7.7 Hz, 1H), 2.52 (ddd, J = 14.2, 6.4, 1.5 Hz, 1H), 2.14 (t, J = 6.9

Hz, 1H), 1.85-1.75 (m, 2H), 1.69 (d, J = 14.3 Hz, 1H), 1.46 (ddd, J = 8.6, 4.4, 1.6 Hz, 1H),

0.88 (s, 9H), 0.85 (s, 9H), 0.04 (s, 3H), 0.03 (s, 3H), 0.03 (s, 3H), 0.02 (s, 3H).

13C-NMR (100 MHz, CDCl3): δ = 199.9, 76.1, 63.8, 52.5, 42.4, 34.4, 30.4, 25.9, 25.7, 19.2,

18.4, 17.8, -4.8, -4.9, -4.4, -5.5.

HR-MS (ESI): no signal found. MS (ESI): calcd for C20H41O3Si2 [M-H]+: 385.26 found: 385.12

(I = 100%).

235

5-((1S,3S,4R,5S)-3-((tert-butyldimethylsilyl)oxy)-4 -(((tert-butyldimethylsilyl)oxy)methyl)

bicyclo[3.1.0]hexan-1-yl)oxazole

234 (51 mg) was dissolved in MeOH (2 ml), K2CO3 (55 mg) and TosMIC (27 mg) were added

and the reaction mixture was refluxed for 4 h, then it was diluted with EtOAc/hex 1:5 and

purified by flash column chromatography (EtOAc/Hex 1:5) to give 9 mg of the product as a

colorless oil.

Rf = 0.31 (Hex/EtOAc 5:1)

1H-NMR (400 MHz, CDCl3): δ = 7.67 (s, 1H), 6.69 (s, 1H), 4.25 (d, J = 6.4 Hz, 1H), 3.55 (dd,

J = 10.1, 5.7 Hz, 1H), 3.36 (dd, J = 10.1, 8.1 Hz, 1H), 2.37 (ddd, J = 13.5, 6.4, 1.5 Hz, 1H),

2.16 (dd, J = 8.1, 5.7 Hz, 1H), 1.95 (d, J = 13.5 Hz, 1H), 1.54 (t, J = 4.5 Hz, 1H), 1.49 (ddd, J

Experimental Section

163

= 8.5, 4.6, 0.9 Hz, 1H), 1.14 (ddd, J = 8.6, 4.3, 1.7 Hz, 1H), 0.88 (s, 9H), 0.87 (s, 9H), 0.04 (s,

3H), 0.03 (s, 3H), 0.03 (s, 3H), 0.02 (s, 3H).

13C-NMR (100 MHz, CDCl3): δ = 149.0, 120.1, 74.8, 64.7, 54.3, 39.7, 33.0, 25.9, 25.7, 24.5,

17.8, 17.6, -4.8, -4.9, -5.5.

MS (ESI): calcd for C22H42NO3Si2 [M-H]+: 424.27, found: 424.00 (I = 100%).

237

(1S,3S,4R,5S)-4-(azidomethyl)-3-((tert-butyldimethy lsilyl)oxy)-N-(2-

hydroxyethyl)bicyclo[3.1.0] hexane-1-carboxamide

3 (60 mg, 0.193 mmol) was dissolved in CH2Cl2 (2 ml) and a few drops of DMF. DIPEA (165

ul, 0.963 mmol) was added, then after 5 min HATU (82 mg, 0.212 mmol) and ethanolamine

(14 ul, 0.232 mmol) were added. The reaction mixture was stirred for 2 h at RT. The reaction

mixture was then diluted with CH2Cl2 and quenched with water. The water phase was

extracted with CH2Cl2, the combined organic phases were dried over MgSO4 and

evaporated. The residue was purified by flash column chromatography (EtOAc/MeOH 20:1)

to give 46 mg (0.139 mmol, 67%) of the product as a yellow oil.

Rf = 0.24 (EtOAc/MeOH 20:1)

1H-NMR (400 MHz, CDCl3): 6.09-6.02 (m, 1H), 4.18-4.14 (d, J = 6.5 Hz, 1H), 3.73-3.69 (t, 4.8

Hz, 1H), 3.47-3.37 (m, 2H), 3.35-3.30 (dd, J = 12.4, 5.4 Hz, 1H), 3.10-3.04 (dd, J = 12.4, 8.3

Hz, 1H), 2.37-2.30 (m, 1H), 2.16-2.11 (dd, J = 8.5, 5.5 Hz, 1H), 1.88-1.85 (d, J = 13.1 Hz,

1H), 1.72-1.67 (ddd, J = 9.0, 4.8, 1 Hz, 1H), 1.46-1.42 (t, J = 4.5 Hz, 1H), 1.36-1.32 (ddd, J =

9.2, 4.1, 1.5 Hz, 1H), 0.85 (s, 9H), 0.04 (s, 3H), 0.03 (s, 3H).

13C-NMR (100 MHz, : 175.0, 76.2, 62.2, 54.4, 50.2, 42.4, 37.5, 32.9, 30.8, 25.4, 20.5, 18.4, -

4.8, -4.9.

MS (ESI): calcd for C16H31N4O3Si [M-H]+: 355.22, found: 355.05 (I = 100%).

238

237 (100 mg, 0.282 mmol) was dissolved in THF (3 ml) and Burgess reagent (83 mg, 0.338

mmol, ((Methoxycarbonyl)sulfamoyl)triethylammonium hydroxide) was added. The reaction

Experimental Section

164

mixture was heated in the microwave at 120°C for 1 h. The solvent was evaporated and the

residue was purified by flash column chromatography (EtOAc/MeOH 20:1) to give 84 mg

(0.250 mmol, 89%) of the product as a colorless oil.

Rf = 0.4 (EtOAc/MeOH 20:1)

1H-NMR (400 MHz, CDCl3): δ = 4.22-4.15 (m, 2H), 4.12-4.08 (m, 1H), 3.84-3.78 (m, 2H),

3.28-3.22 (dd, J = 12.4, 6.5 Hz, 1H), 3.14-3.08 (dd, J = 12.4, 8.3 Hz, 1H), 2.57-2.50 (ddd, J =

14.0, 6.5, 1.6 Hz, 1H), 2.17-2.12 (dd, J = 8.2, 6.5 Hz, 1H), 1.95-1.89 (d, J = 14.0 Hz, 1H),

1.68-1.63 (ddd, J = 9.0, 4.9, 1.0 Hz, 1H), 1.46-1.42 (t, J = 4.6 Hz, 1H), 1.36-1.31 (ddd, J =

9.0, 4.3, 1.7 Hz, 1H), 0.84 (s, 9H), 0.02 (s, 3H), 0.01 (s, 3H).

13C-NMR (100 MHz, CDCl3): δ = 169.9, 75.6, 67.7, 54.4, 53.6, 50.6, 38.4, 30.8, 27.0, 25.8,

19.4, 17.5, -4.6, -4.9.

MS (ESI): calcd for C16H29N4O2Si [M-H]+: 337.21, found: 337.03 (I = 100%).

241

241 (58 mg, 98%) was prepared from 64 mg of 238 according to the procedure followed for

compound 203e.

Rf = 0.1 (EtOAc/MeOH 20:1)

MS (ESI): calcd for C16H31N2O2Si [M-H]+: 311.22, found: 311.05 (I = 100%).

2x

Compounds 2x were prepared as described for compounds 1ax.

Experimental Section

165

Amount : 2 mg. Purity : ≥ 30%. MS (ESI): calcd for C18H21F2N2O3 [M-H]+: 351.15, found: 351.00. (I =

80%).

Amount : 0.5 mg. Purity : ≥ 50%. MS (ESI): calcd for C24H30N3O4S [M-H]+: 434.17, found: 434.00 (I =

50%).

HO

NH

O

2n

O

NO

O

O

Amount : 1 mg. Purity : ≥ 33%. MS (ESI): calcd for C22H25N2O6 [M-H]+: 413.17, found: 413.10. (I =

100%).

Amount : 1 mg. Purity : ≥ 50%. MS (ESI): calcd for C21H27N4O4 [M-H]+: 399.20, found: 399.00 (I =

50%). 1H-NMR (500 MHz, DMSO): δ = 8.15 (t, J = 5.0 Hz, 1H), 7.09-6.99 (m, 2H), 6.98-6.93 (m, 1H),

3.92 (d, J = 6.3 Hz, 1H), 3.60 (s, 2H), 3.51-3.43 (m, 2H), 3.43-3.33 (m, 2H), 3.30 (s, 6H), 3.21-3.05 (m,

2H), 3.03-2.92 (m, 1H), 2.89-2.77 (m, 1H), 2.04-1.91 (m, 2H), 1.75-1.68 (m, 1H), 1.55 (dd, J = 8.5, 5.0

Hz, 1H), 1.37 (t, J = 4.7 Hz, 1H), 1.14-1.10 (m, 1H). 13C-NMR (500 MHz, DMSO): δ = 172.0, 154.1,

128.2, 127.2, 123.0, 121.9, 74.5, 59.9, 51.3, 49.4, 43.8, 42.4, 41.7, 30.2, 27.1, 26.8.

Experimental Section

166

Amount : 1 mg. MS (ESI): calcd for C21H26N3O3 [M-H]+: 368.20, found: 368.05 (I = 50%).

219

N-(4,6-dichloropyrimidin-5-yl)formamide

Acetic anhydride (1.12 ml) was added dropwise over 5min to a solution of 4,6-dichloro-5-

aminopyrimidine (240 mg) in formic acid (3 ml) at 0°C, then it was stirred at RT for 4 h. The

solvent was removed and the residue was co-evaporated with toluene (3x) to yield 302 mg of

219 (1.573 mmol, >100%).

Rf = 0.13 (Hex/EtOAc 2:1)

1H NMR (400 MHz, DMSO): δ = 10.49 (s, br, 1H), 8.85 (s, 1H), 8.34 (s, 1H).

222

9-((1S,3S,4R,5S)-3-((tert-butyldimethylsilyl)oxy)-4 -(((tert-butyldimethylsilyl)oxy)

methyl)bicyclo [3.1.0]hexan-1-yl)-1H-purin-6(9H)-on e

221 (52 mg) was dissolved in MeOH (15 ml), 2-mercaptoethanol (145 ul) and NaOMe (110

mg) were added. The reaction mixture was heated to 80°C in the microwave for 1.5 h. Acetic

acid (117 ul) was added and the mixture was stirred for 10 min. The solvents were

evaporated and the residue was purified by flash column chromatography (EE/MeOH 20:1)

to yield 32 mg (61%) of the product as a white solid.

Rf = 0.16 (EtOAc/MeOH 20:1)

1H NMR (400 MHz, MeOD): δ = 8.03 (s, 1H), 7.99 (s, 1H), 4.39 (d, J = 6.4 Hz, 1H), 3.92 (dt, J

= 10.4, 7.5 Hz, 2H), 2.58 (ddd, J = 13.3, 6.5, 2.1 Hz, 1H), 2.25 (d, J = 13.4 Hz, 1H), 2.17 (dd,

Experimental Section

167

J = 8.5, 5.7 Hz, 1H), 1.84 – 1.76 (m, 1H), 1.71 (t, J = 5.1 Hz, 1H), 1.39 (ddd, J = 9.7, 5.3, 2.1

Hz, 1H), 0.95 (s, 6H), 0.91 (s, 6H), 0.13 (s, 3H), 0.13 (s, 3H), 0.08 (s, 3H), 0.07 (s, 3H).

13C-NMR (100 MHz, MeOD): δ = 159.2, 150.5, 144.3, 141.2, 125.2, 74.2, 60.5, 53.0, 43.4,

42.0, 41.4, 26.1, 25.8, 18.5, 17.9, 16.9, -4.7, -4.8, -5.3, -5.3.

IR (film): ν 2929, 2857, 1686, 1401, 1254, 1083, 1041, 907, 837, 778, 731, 649.

HR-MS (ESI): calcd for C24H42N4NaO3Si2 [M-Na]+: 513.2688, found: 513.2690.

229

9-((1S,3S,4R,5S)-3-((tert-butyldimethylsilyl)oxy)-4 -(hydroxymethyl)bicyclo[3.1.0]hexan-

1-yl)-1H-purin-6(9H)-one

To a solution of 222 (120 mg, 0.245 mmol) in CH2Cl2/MeOH 1:1 (6 ml) CSA (74 mg, 0.319

mmol) was added at 0°C. The reaction mixture was st irred for 3 h at 0°C, additional 25 mg of

CSA were added and the reaction mixture was stirred for additional 2 h at 0°C. The reaction

mixture was quenched with Et3N (55 ul, 0.392 mmol) and evaporated. The residue was

purified by flash chromatography (EE/MeOH 10:1) to afford 80 mg of the product as a white

solid. 1H-NMR revealed that the product contained CSA. It was therefore re-dissolved in

aqueous NaHCO3 solution and extracted with a mixture of EtOAc/MeOH 5% to give 75 mg

(0.199 mmol, 81%) of the pure product as a white solid.

Rf = 0.4 (EtOAc/MeOH 10:1)

1H-NMR (400 MHz, CDCl3): δ = 8.22 (s, 1H), 7.95 (s, 1H), 5.42 (d, J = 11.7 Hz, 1H), 4.43 (d,

J = 5.5 Hz, 1H), 4.02 (d, J = 11.4 Hz, 1H), 3.83 (t, J = 10.0 Hz, 1H), 2.60 (ddd, J = 8.7, 6.3,

1.7 Hz, 1H), 2.16 (d, J = 13.1 Hz, 2H), 1.77 (dd, J = 8.5, 4.9 Hz, 1H), 1.72 (t, J = 5.0 Hz, 1H),

1.30 (ddd, J = 10.8, 5.7, 2.8 Hz, 1H), 0.89 (s, 9H), 0.05 (s, 3H), 0.05 (s, 3H).

13C-NMR (100 MHz, CDCl3): δ = 158.6, 148.9, 144.5, 142.4, 75.9, 64.9, 53.6, 44.5, 44.2,

26.9, 25.2, 17.9, 16.9, -4.5.

IR (film): ν 2952, 2927, 2856, 2365, 1714, 1591, 1546, 1467, 1404, 1344, 1253, 1210, 1171,

1084, 1048, 1011, 939, 963, 913, 863, 833, 775, 742, 647, 611.

HR-MS (ESI): calcd for C18H28N4NaO3Si [M-Na]+: 399.1823, found: 399.1829.

Experimental Section

168

216

(E)-3-methoxyacrylic acid

KOH (7.4 g, 0.132 mol) and Methyl 3-methoxyacrylate (215, 11 ml, 0.102 mol) were

dispensed in 30 ml of H2O, and the reaction mixture was stirred overnight at 50°C. The

reaction mixture was cooled to 0°C and acidified sl owly to pH ~1 with 1 N aqueous HCl. The

water phase was extracted with Et2O (3 x 200 ml), the combined organic phases were dried

and evaporated to yield the crude product as a light yellow solid. Purification by

recrystallization from hexane/CHCl3 gave 4.44 g (0.034 mol, 33%) of the pure product as

light yellow crystals.

Rf = 0.3 (EtOAc/Hex 1:1)

1H NMR (400 MHz, CDCl3): δ = 11.82 (s, br, 1H), 7.69 (d, J = 12.6 Hz, 1H), 5.17 (d, J = 12.6

Hz, 1H), 3.71 (s, 3H).

13C-NMR (100 MHz, CDCl3): δ = 173.6, 165.3, 95.6, 57.5.

O

NCOO

211

(E)-3-methoxyacryloyl isocyanate

Thionyl chloride (3.6 ml) was added to a solution of 216 (2.93 g) in dry CH2Cl2 (300 ml). The

mixture was refluxed for 4 h. The solvent was evaporated to give a brown liquid that was

purified by kugelrohr distillation to afford 2.6 g (74%) of 3-methoxyacryloyl chloride (217) as a

colorless liquid. The liquid was dissolved in toluene (20 ml), silver cyanate (5.9 g, 0.039 mol))

was added and this mixture was refluxed at 130°C fo r 45 min in the dark. The suspension

was cooled with ice and the supernatant was transferred by syringe to the solution of 5 in

CH2Cl2 at -15°C.

212

(E)-N-(((1S,3S,4R,5S)-4-(azidomethyl)-3-(benzyloxy) bicyclo[3.1.0]hexan-1-

yl)carbamoyl)-3-methoxyacrylamide

Experimental Section

169

3-methoxyacryloyl chloride (0.6 g, 5 mmol) was dissolved in toluene and 1.4 g of silver

cyanate (dried over P2O5 at 100°C for 2.5 h in the dark before use) was add ed and the

mixture was refluxed at 120°C in the dark for 30 mi n. The suspension was cooled with ice

and the supernatant was transferred by syringe to the sol. of 5 (130 mg, 0.503 mmol) in

CH2Cl2 at 0°C. The reaction mixture was slowly warmed to RT, then stirred overnight at RT.

The mixture had turned into a dispersion. The solvents were evaporated and the crude

product was purified by flash chromatography (Hex/EtOAc 2:1--> Hex/EtOAc 1:1) to yield

150 mg (0.389, 77%) of the product as a white solid.

Rf = 0.3 (EtOAc/Hex 1:1)

1H-NMR (400 MHz, CDCl3): δ = 8.84 (s, 1H), 8.63 (s, 1H), 7.64 (d, J = 12.1 Hz, 1H), 7.37 –

7.27 (m, 5H), 5.24 (d, J = 12.1 Hz, 1H), 4.42 (s, 2H), 3.84 (d, J = 7.2 Hz, 1H), 3.71 (s, 3H),

3.54 (dd, J = 12.3, 6.5 Hz, 1H), 3.39 (dd, J = 12.3, 9.3 Hz, 1H), 2.45 (ddd, J = 13.8, 7.1, 2.0

Hz, 1H), 2.31 (dd, J = 9.2, 6.6 Hz, 1H), 2.18 (d, J = 13.9 Hz, 1H), 1.40 (dd, J = 8.4, 4.8 Hz,

1H), 1.28 (dd, J = 11.2, 6.2 Hz, 2H), 0.97 (ddd, J = 9.2, 5.2, 2.1 Hz, 1H).

13C-NMR (100 MHz, CDCl3): δ = 164.1, 138.8, 128.4, 127.6, 127.4, 97.9, 82.6, 70.9, 58.6,

53.9, 47.9, 39.9, 36.3, 26.9.

MS (ESI): calcd for C19H22N5O4 [M-H]- : 384.1672, found: 384.10 (I = 100%).

213

1-((1S,3S,4R,5S)-4-(azidomethyl)-3-(benzyloxy)bicyc lo[3.1.0]hexan-1-yl)pyrimidine-

2,4(1H,3H)-dione

A 1 M aq. H2SO4 sol.(3.4 ml) and dioxane (3 ml) were added to 212 and the reaction mixture

was refluxed for 2 h. After cooling, the mixture was neutralized with 2 M aq. NaOH sol. (3.4

ml) to pH 7 and the solvents were evaporated to give the crude product, ~0.8 g, that was

purified by flash chromatography (EtOAc/Hex 2:1) to afford 100 mg (0.283 mmol, 84%) of the

product.

Rf = 0.25 (EtOAc/Hex 2:1)

1H-NMR (400 MHz, CDCl3): δ = 9.10 (d, J = 10.0 Hz, 1H), 7.35 – 7.28 (m, 5H), 5.67 (d, J =

10.0 Hz, 1H), 4.43 (q, J = 11.8 Hz, 2H), 3.90 (d, J = 7.4 Hz, 1H), 3.66 (dd, J = 12.4, 6.8 Hz,

1H), 3.56 (dd, J = 12.4, 8.9 Hz, 1H), 2.47 (ddd, J = 13.8, 7.1, 2.4 Hz, 1H), 2.38 (dd, J = 8.8,

Experimental Section

170

6.8 Hz, 1H), 2.22 (d, J = 13.8 Hz, 1H), 1.70 (ddd, J = 9.8, 4.9, 1.2 Hz, 1H), 1.56 – 1.49 (m,

1H), 1.09 (ddd, J = 9.7, 5.7, 2.4 Hz, 1H).

13C-NMR (100 MHz, CDCl3): δ = 163.7, 150.4, 145.1, 138.2, 129.5, 127.8, 127.3, 102.3, 82.1,

71.2, 52.9, 48.4, 47.3, 36.7, 27.8, 18.1.

MS (ESI): calcd for C18H20N5O3 [M-H]+: 354.1566, found: 354.04 (I = 100%).

214

1-((1S,3S,4R,5S)-4-(aminomethyl)-3-(benzyloxy)bicyc lo[3.1.0]hexan-1-yl)pyrimidine-

2,4(1H,3H)-dione

213 (14 mg, 0.040 mmol) was dissolved in EtOH, Lindlar’s catalyst (5 mg, 0.004 mmol, ~5%

Pd/CaCO3) was added and the reaction mixture was stirred under an atmosphere of

hydrogen (3 bar) for 4 h. The solution was filtrated over celite and the filter cake was washed

with MeOH/EtOAc. The solvents were evaporated and the crude product (16 mg, 0.048

mmol, >100%), a slightly yellow solid, was used to prepare compounds 218x.

Rf = 0.1 (CHCl3/MeOH 10:1)

HR-MS (ESI): cacld for C18H22N3O3 [M-H]+: 328.1661, found: 327.84.

218x

Compounds 218x were prepared as described for compounds 1ax, without the deprotection

step.

Amount : 9 mg. Purity : ≥ 79%. TLC: Rf 0.33 (EtOAc/MeOH 30:1, CPS). HR-MS (ESI): calcd for

C26H26F2N3O4 [M-H]+: 482.1886, found: 482.1903.

Experimental Section

171

Amount : 13 mg. Purity : ≥ 98%. TLC: Rf 0.19 (EtOAc/MeOH 30:1, CPS). HR-MS (ESI): calcd for

C29H33N4O6S [M-H]+: 565.2115, found: 565.2122. 1H-NMR (500 MHz, DMSO): δ = 8.67 (t, J = 5.7 Hz,

1H), 7.68 (d, J = 8.0 Hz, 1H), 7.39 (s, 1H), 7.36-7.29 (m, 2H), 7.25-7.17 (m, 7H), 5.52 (dd, J = 7.8, 1.4

Hz, 1H), 4.31 (q, J = 12.2 Hz, 2H), 3.87 (d, J =6.7 Hz, 1H), 3.83-3.77 (m, 4H), 3.64-3.56 (m, 4H), 3.11-

3.05 (m, 2H), 2.39 (t, J = 7.5 Hz, 1H), 2.31 (dd, J = 13.3, 7.0 Hz, 1H), 2.22 (d, J = 13.7 Hz, 1H), 1.69

(dd, J = 9.5, 4.8 Hz, 1H), 1.29 (t, J = 5.0 Hz, 1H), 1.23-1.08 (m, 1H). 13C-NMR (500 MHz, DMSO): δ =

162.2, 151.0, 128.0, 127.1, 118.5, 118.1, 114.3, 101.1, 81.3, 69.4, 53.5, 49.9, 48.0, 46.8, 46.3, 38.1,

35.6, 17.9, 16.6. Note: Quaternary C’s and 1 C=O were not detected.

Amount : 11 mg. Purity : ≥ 98%. TLC: Rf 0.23 (EtOAc/MeOH 30:1, CPS). HR-MS (ESI): calcd for

C24H26N3O5 [M-H]+: 436.1867, found: 436.1876.

Amount : 4 mg. Purity : ≥ 52%. TLC: Rf 0.20 (EtOAc/MeOH 30:1, CPS). HR-MS (ESI): calcd for

C30H30N3O7 [M-H]+: 544.2078, found: 544.2075.

Amount : 11 mg. Purity : ≥ 80%. TLC: Rf 0.10 (EtOAc/MeOH 20:1 with 0.5% Et3N, CPS). HR-MS

(ESI): calcd for C28H28N5O5 [M-H]+: 514.2085, found: 514.2083.

Experimental Section

172

Amount : 6 mg. Purity : ≥ 98%. TLC: Rf 0.12 (EtOAc/MeOH 10:1 with 0.5% Et3N, CPS). HR-MS (ESI):

calcd for C29H32N5O5 [M-H]+: 530.2398, found: 530.2401.

Amount : 7.5 mg. Purity : ≥ 85%. TLC: Rf 0.28 (EtOAc/MeOH 20:1, CPS). HR-MS (ESI): calcd for

C23H30N3O4 [M-H]+: 412.2231, found: 412.2225.

NH

O

218e

N

BnO

NHO

O

Amount : 3 mg. Purity : ≥ 75%. TLC: Rf 0.32 (EtOAc/MeOH 20:1, CPS). HR-MS (ESI): calcd for

C27H30N3O4 [M-H]+: 460.2231, found: 460.2236.

Amount : 8 mg. Purity : ≥ 76%. TLC: Rf 0.16 (EtOAc/MeOH 10:1 with 0.5% Et3N, CPS). MS (ESI):

calcd for C24H31N4O5 [M-H]+: 455.23, found: 455.10 (I = 100%).

Amount : 4 mg. Purity : ≥ 95%. TLC: Rf 0.30 (EtOAc/MeOH 20:1, CPS). HR-MS (ESI): calcd for

C25H32N3O4 [M-H]+: 438.2387, found: 438.2393.

Experimental Section

173

NH

O

N

218l

NN

BnO

NHO

O

Amount : 7 mg. Purity : ≥ 84%. TLC: Rf 0.10 (EtOAc/MeOH 6:1 with 0.5% Et3N, CPS). HR-MS (ESI):

calcd for C23H26N5O4 [M-H]+: 436.1979, found: 436.1973.

Amount : 10 mg. Purity : ≥ 73%. TLC: Rf 0.26 (EtOAc/MeOH 20:1, CPS). MS (ESI): calcd for

C29H33N4O5 [M-H]+: 517.25, found: 517.20 (I = 100%).

Amount : 12 mg. Purity : ≥ 72%. TLC: Rf 0.26 (EtOAc/MeOH 10:1 with 0.5% Et3N, CPS). HR-MS

(ESI): calcd for C24H27N4O4S [M-H]+: 467.1748, found: 467.1736.

Experimental Section

174

5.2.1.5 Crystallographic Data

Crystallographic data for compound 1do

Experimental Crystal data C26H26N4O4 C26H26N4O4 Mr = 458.518 Triclinic P 1̄ a = 10.5120 (5)Å b = 11.8209 (6)Å c = 18.0423 (8)Å α = 89.020 (2)° β = 85.086 (2)° γ = 82.639 (2)° V = 2215.3 (2)Å3 Z = 4

F(000) = 968 Dx = 1.375 Mg m-3 Density measured by: not measured fine-focus sealed tube Mo Kα radiation λ = 0.71073 Cell parameters from 31246 refl. θ = 2.425—26.022 ° µ = 0.095 mm-1 T = 173 K Cube 0.225 x 0.12 x 0.075 mm Colourless Crystal source: Pharm ETH Zurich

Data Collection KappaCCD CCD diffractometer Absorption correction: none 14716 measured reflections 8684 independent reflections 6398 observed reflections Criterion: >2sigma(I)

Rint = 0.091 θmax = 26.09 ° h = -12 →12 k = -14 →14 l = -21 →22

Refinement Refinement on F2 fullmatrix least squares refinement R(all) = 0.1548 R(gt) = 0.1269 wR(ref) = 0.3024 wR(gt)= 0.2871

S(ref) = 1.781 8684 reflections 618 parameters 0 restraints H positions constr

Experimental Section

175

Calculated weights 1/[σ2(Io)+(Io+Ic)2/900]

∆/σmax = 0.001 ∆ρmax = 1.067eÅ3 ∆ρmin = -0.705eÅ3 Extinction correction: none Atomic scattering factors from International

Tables Vol C Tables 4.2.6.8 and 6.1.1.4

Experimental Section

176

Data collection: KappaCCD Cell refinement: HKL Scalepack (Otwinowski & Minor 1997) Data reduction: Denzo and Scalepak (Otwinowski & Minor, 1997) Program(s) used to solve structure: SIR97(Altomare et al., J. Appl. Cryst.,1999) Program(s) used to refine structure: SHELXL-97 (Sheldrick, 1997

Table 1. Fractional atomic coordinates and equivalent isotropic thermal parameters (Å2) Ueq = 1/3ΣiΣj Uij ai*a j* ai.aj .

x y z Ueq Occ O7 0.3073 (3) 0.8428 (3) -1.05909 (18) 0.0392 (9) 1 O11 0.7530 (3) 0.8765 (3) -1.1849 (2) 0.0384 (9) 1 O23 1.1496 (3) 0.5122 (3) -0.92699 (19) 0.0335 (8) 1 O25 0.6758 (3) 0.6085 (3) -0.88858 (19) 0.0343 (8) 1 O40 1.1917 (3) 0.9388 (3) -0.7266 (2) 0.0389 (9) 1 O44 0.7498 (3) 1.1098 (3) -0.79465 (16) 0.0315 (8) 1 O56 0.3546 (3) 0.8773 (3) -0.48087 (18) 0.0362 (9) 1 O58 0.8265 (4) 0.8133 (4) -0.5139 (2) 0.0499 (12) 1 N9 0.7662 (4) 0.7077 (4) -1.1240 (2) 0.0281 (9) 1 N14 1.0902 (4) 0.6319 (4) -1.1053 (2) 0.0300 (9) 1 N15 1.1347 (4) 0.5649 (4) -1.0478 (2) 0.0301 (9) 1 N26 0.4787 (4) 0.6061 (4) -0.8292 (2) 0.0250 (9) 1 N42 0.7368 (4) 1.0983 (3) -0.6700 (2) 0.0228 (8) 1 N47 0.4110 (4) 1.0897 (4) -0.6173 (2) 0.0285 (9) 1 N48 0.3682 (4) 1.0309 (4) -0.5564 (2) 0.0278 (9) 1 N59 1.0144 (4) 0.7080 (4) -0.4987 (2) 0.0278 (9) 1 C1 0.5865 (5) 0.7813 (5) -0.9947 (3) 0.0308 (11) 1 C2 0.5421 (4) 0.7818 (4) -1.0725 (2) 0.0241 (10) 1 C3 0.4047 (5) 0.7462 (4) -1.0620 (3) 0.0296 (11) 1 C4 0.3967 (5) 0.6814 (5) -0.9875 (3) 0.0303 (11) 1 C5 0.4992 (4) 0.7174 (4) -0.9423 (2) 0.0250 (10) 1 C6 0.4989 (6) 0.8448 (5) -0.9348 (3) 0.0359 (12) 1 C8 0.6305 (4) 0.6961 (4) -1.1211 (2) 0.0256 (10) 1 C10 0.8174 (5) 0.7963 (4) -1.1542 (2) 0.0297 (11) 1 C12 0.9621 (5) 0.7945 (5) -1.1506 (3) 0.0340 (12) 1 C13 1.0169 (4) 0.7269 (4) -1.0876 (3) 0.0270 (10) 1 C16 1.1059 (4) 0.5829 (4) -0.9737 (3) 0.0276 (10) 1 C17 0.9892 (5) 0.7174 (5) -0.8798 (3) 0.0361 (12) 1 C18 0.9159 (6) 0.8202 (5) -0.8625 (3) 0.0421 (13) 1 C19 0.8747 (6) 0.8943 (5) -0.9186 (3) 0.0415 (13) 1 C20 0.9089 (5) 0.8666 (5) -0.9924 (3) 0.0335 (11) 1 C21 0.9843 (4) 0.7629 (4) -1.0106 (3) 0.0288 (11) 1 C22 1.0235 (4) 0.6886 (4) -0.9546 (3) 0.0268 (10) 1 C24 0.5598 (4) 0.6403 (4) -0.8853 (2) 0.0241 (10) 1 C27 0.3420 (4) 0.6517 (5) -0.8139 (3) 0.0332 (11) 1 C28 0.3090 (5) 0.6216 (5) -0.7321 (3) 0.0321 (11) 1 C29 0.4138 (5) 0.5286 (4) -0.7166 (3) 0.0282 (10) 1 C30 0.4238 (5) 0.4550 (5) -0.6561 (3) 0.0341 (11) 1 C31 0.5353 (5) 0.3777 (4) -0.6531 (3) 0.0319 (11) 1 C32 0.6361 (5) 0.3778 (4) -0.7072 (3) 0.0331 (11) 1 C33 0.6276 (5) 0.4509 (4) -0.7683 (3) 0.0274 (10) 1 C68 0.9115 (4) 0.8860 (4) -0.6627 (2) 0.0242 (10) 1 C35 0.9589 (4) 0.9938 (4) -0.6941 (2) 0.0227 (9) 1 C36 1.0980 (4) 0.9883 (4) -0.6700 (3) 0.0278 (10) 1 C37 1.1047 (5) 0.9134 (4) -0.6010 (3) 0.0283 (10) 1 C34 0.5136 (4) 0.5243 (4) -0.7723 (2) 0.0244 (10) 1 C38 0.9986 (4) 0.8363 (4) -0.6054 (2) 0.0237 (10) 1 C39 0.9976 (5) 0.7752 (4) -0.6777 (2) 0.0273 (10) 1 C41 0.8734 (4) 1.0998 (4) -0.6625 (2) 0.0231 (9) 1 C43 0.6858 (4) 1.1028 (4) -0.7351 (2) 0.0234 (9) 1

Experimental Section

177

C45 0.5400 (5) 1.0987 (4) -0.7316 (3) 0.0293 (11) 1 C46 0.4876 (4) 1.0317 (4) -0.6670 (2) 0.0254 (10) 1 C49 0.3993 (4) 0.9187 (4) -0.5397 (2) 0.0264 (10) 1 C50 0.5254 (5) 0.7401 (5) -0.5854 (3) 0.0327 (11) 1 C51 0.6038 (5) 0.6779 (5) -0.6403 (3) 0.0335 (11) 1 C52 0.6428 (5) 0.7319 (5) -0.7063 (3) 0.0332 (11) 1 C53 0.6037 (4) 0.8471 (4) -0.7162 (3) 0.0258 (10) 1 C54 0.5258 (4) 0.9102 (4) -0.6604 (2) 0.0242 (10) 1 C55 0.4865 (4) 0.8534 (4) -0.5957 (2) 0.0262 (10) 1 C57 0.9391 (4) 0.7862 (4) -0.5361 (2) 0.0244 (10) 1 C60 1.1476 (6) 0.6592 (6) -0.5249 (3) 0.0552 (19) 1 C61 1.1865 (5) 0.5675 (5) -0.4691 (3) 0.0401 (13) 1 C62 1.0840 (5) 0.5812 (5) -0.4069 (3) 0.0308 (11) 1 C63 1.0766 (5) 0.5320 (5) -0.3370 (3) 0.0377 (12) 1 C64 0.9682 (6) 0.5610 (5) -0.2877 (3) 0.0400 (13) 1 C65 0.8667 (5) 0.6363 (5) -0.3095 (3) 0.0366 (12) 1 C66 0.8711 (5) 0.6888 (4) -0.3793 (3) 0.0313 (11) 1 C67 0.9818 (5) 0.6626 (4) -0.4271 (3) 0.0270 (10) 1 H7 0.2911 0.8633 -1.1031 0.06 (2) 1 H40 1.1933 0.9830 -0.7642 0.09 (3) 1 H9 0.8178 0.6527 -1.1045 0.034 1 H15 1.1879 0.5033 -1.0604 0.036 1 H42 0.6852 1.0943 -0.6292 0.027 1 H48 0.3138 1.0705 -0.5239 0.033 1 H1 0.6740 0.7471 -0.9926 0.037 1 H2 0.5391 0.8559 -1.0954 0.029 1 H3 0.3929 0.6975 -1.1022 0.036 1 H4A 0.3127 0.7008 -0.9623 0.036 1 H4B 0.4113 0.6009 -0.9968 0.036 1 H6A 0.5498 0.8802 -0.9024 0.043 1 H6B 0.4416 0.9024 -0.9575 0.043 1 H8A 0.6022 0.7058 -1.1703 0.031 1 H8B 0.6162 0.6219 -1.1024 0.031 1 H12A 1.0066 0.7621 -1.1955 0.041 1 H12B 0.9813 0.8711 -1.1454 0.041 1 H17 1.0151 0.6655 -0.8408 0.043 1 H18 0.8932 0.8409 -0.8114 0.051 1 H19 0.8230 0.9654 -0.9062 0.050 1 H20 0.8796 0.9178 -1.0310 0.040 1 H27A 0.3309 0.7325 -0.8227 0.040 1 H27B 0.2918 0.6156 -0.8463 0.040 1 H28A 0.3095 0.6879 -0.7022 0.039 1 H28B 0.2255 0.5964 -0.7260 0.039 1 H30 0.3548 0.4575 -0.6174 0.041 1 H31 0.5434 0.3222 -0.6137 0.038 1 H32 0.7140 0.3271 -0.7025 0.040 1 H33 0.6982 0.4495 -0.8059 0.033 1 H68 0.8244 0.8979 -0.6407 0.029 1 H35 0.9610 0.9956 -0.7474 0.027 1 H36 1.1146 1.0641 -0.6595 0.033 1 H37A 1.1884 0.8697 -0.6024 0.034 1 H37B 1.0909 0.9598 -0.5572 0.034 1 H39A 0.9462 0.7159 -0.6862 0.033 1 H39B 1.0550 0.7844 -0.7211 0.033 1 H41A 0.9019 1.1646 -0.6888 0.028 1 H41B 0.8882 1.1044 -0.6109 0.028 1 H45A 0.4957 1.1748 -0.7281 0.035 1 H45B 0.5199 1.0636 -0.7760 0.035 1 H50 0.4981 0.7045 -0.5395 0.039 1

Experimental Section

178

H51 0.6301 0.5982 -0.6320 0.040 1 H52 0.6963 0.6891 -0.7446 0.040 1 H53 0.6302 0.8858 -0.7610 0.031 1 H60A 1.1508 0.6314 -0.5748 0.066 1 H60B 1.2018 0.7185 -0.5236 0.066 1 H61A 1.1893 0.4953 -0.4932 0.048 1 H61B 1.2696 0.5749 -0.4529 0.048 1 H63 1.1455 0.4772 -0.3224 0.045 1 H64 0.9640 0.5303 -0.2381 0.048 1 H65 0.7899 0.6523 -0.2766 0.044 1 H66 0.7999 0.7416 -0.3935 0.038 1

Table 2. Anisotropic displacement parameters (Å2) U11 U12 U13 U22 U23 U33

O7 0.037 (2) 0.0143 (18) -0.0045 (15) 0.052 (2) 0.0026 (16) 0.0239 (17) O11 0.034 (2) -0.0051 (17) -0.0035 (15) 0.039 (2) 0.0185 (16) 0.041 (2) O23 0.0316 (19) 0.0016 (15) -0.0103 (15) 0.0293 (19) 0.0078 (15) 0.0391 (19) O25 0.0247 (18) 0.0019 (16) 0.0017 (14) 0.042 (2) 0.0070 (15) 0.0340 (18) O40 0.0305 (19) 0.0037 (16) 0.0121 (16) 0.039 (2) 0.0129 (17) 0.042 (2) O44 0.0307 (18) -0.0008 (15) 0.0008 (13) 0.042 (2) 0.0060 (14) 0.0198 (16) O56 0.034 (2) -0.0105 (17) 0.0025 (14) 0.054 (2) 0.0029 (15) 0.0207 (16) O58 0.030 (2) 0.0146 (19) 0.0130 (17) 0.063 (3) 0.026 (2) 0.047 (2) N9 0.027 (2) -0.0008 (17) -0.0033 (16) 0.028 (2) 0.0084 (16) 0.029 (2) N14 0.0174 (19) -0.0059 (18) 0.0030 (15) 0.042 (3) 0.0031 (18) 0.030 (2) N15 0.025 (2) -0.0020 (17) -0.0019 (16) 0.032 (2) -0.0016 (17) 0.034 (2) N26 0.0179 (18) 0.0034 (16) -0.0008 (14) 0.033 (2) 0.0027 (16) 0.0221 (18) N42 0.0226 (19) -0.0002 (15) -0.0006 (14) 0.024 (2) 0.0038 (14) 0.0211 (18) N47 0.022 (2) -0.0010 (17) -0.0015 (16) 0.033 (2) -0.0003 (17) 0.030 (2) N48 0.0209 (19) -0.0022 (17) 0.0044 (15) 0.034 (2) -0.0074 (16) 0.027 (2) N59 0.021 (2) 0.0026 (17) 0.0049 (16) 0.029 (2) 0.0097 (17) 0.030 (2) C1 0.032 (3) -0.006 (2) -0.0012 (19) 0.036 (3) 0.0009 (19) 0.024 (2) C2 0.027 (2) -0.0026 (19) -0.0016 (18) 0.022 (2) 0.0059 (17) 0.023 (2) C3 0.028 (2) -0.001 (2) -0.0037 (18) 0.036 (3) 0.0001 (19) 0.024 (2) C4 0.025 (2) -0.005 (2) -0.0050 (19) 0.037 (3) 0.007 (2) 0.029 (2) C5 0.023 (2) -0.0018 (19) -0.0047 (17) 0.029 (2) 0.0004 (18) 0.023 (2) C6 0.051 (3) -0.007 (2) -0.003 (2) 0.030 (3) 0.001 (2) 0.027 (2) C8 0.026 (2) -0.0019 (19) 0.0006 (18) 0.026 (2) 0.0035 (18) 0.023 (2) C10 0.026 (2) -0.001 (2) 0.0002 (18) 0.036 (3) 0.006 (2) 0.025 (2) C12 0.029 (3) -0.008 (2) 0.003 (2) 0.041 (3) 0.009 (2) 0.031 (3) C13 0.022 (2) -0.006 (2) 0.0001 (17) 0.033 (3) 0.0056 (19) 0.026 (2) C16 0.020 (2) -0.0055 (19) -0.0047 (18) 0.028 (3) 0.003 (2) 0.035 (3) C17 0.037 (3) -0.004 (2) -0.005 (2) 0.042 (3) 0.001 (2) 0.029 (3) C18 0.045 (3) 0.001 (3) -0.002 (2) 0.048 (3) -0.010 (2) 0.032 (3) C19 0.042 (3) 0.001 (2) -0.006 (2) 0.035 (3) -0.005 (2) 0.047 (3) C20 0.029 (3) 0.000 (2) -0.006 (2) 0.035 (3) -0.001 (2) 0.035 (3) C21 0.018 (2) -0.0061 (19) 0.0025 (19) 0.028 (3) 0.002 (2) 0.040 (3) C22 0.021 (2) -0.0064 (19) -0.0013 (18) 0.029 (3) 0.0016 (19) 0.031 (2) C24 0.021 (2) -0.0011 (19) -0.0061 (17) 0.027 (2) -0.0024 (18) 0.025 (2) C27 0.018 (2) 0.002 (2) 0.0004 (19) 0.045 (3) 0.003 (2) 0.034 (3) C28 0.024 (2) -0.001 (2) 0.0020 (19) 0.038 (3) 0.006 (2) 0.033 (3) C29 0.027 (2) -0.006 (2) -0.0013 (19) 0.029 (3) 0.0014 (19) 0.029 (2) C30 0.038 (3) -0.008 (2) 0.000 (2) 0.038 (3) 0.002 (2) 0.027 (2) C31 0.045 (3) -0.006 (2) -0.010 (2) 0.021 (2) 0.0067 (19) 0.031 (3) C32 0.036 (3) -0.001 (2) -0.007 (2) 0.023 (3) 0.003 (2) 0.040 (3) C33 0.032 (3) -0.001 (2) -0.0035 (19) 0.022 (2) -0.0038 (18) 0.028 (2) C68 0.025 (2) -0.0026 (19) -0.0031 (17) 0.028 (2) 0.0033 (17) 0.020 (2) C35 0.026 (2) -0.0037 (19) -0.0039 (17) 0.025 (2) 0.0017 (17) 0.018 (2) C36 0.021 (2) -0.0051 (19) -0.0032 (19) 0.029 (3) 0.0052 (19) 0.034 (2) C37 0.025 (2) -0.005 (2) -0.0052 (18) 0.030 (3) 0.0038 (19) 0.031 (2)

Experimental Section

179

C34 0.025 (2) -0.0039 (19) -0.0053 (18) 0.024 (2) 0.0000 (18) 0.025 (2) C38 0.021 (2) -0.0017 (19) -0.0016 (17) 0.026 (2) 0.0028 (18) 0.023 (2) C39 0.031 (2) -0.002 (2) -0.0042 (19) 0.023 (2) -0.0003 (18) 0.028 (2) C41 0.024 (2) 0.0015 (19) -0.0032 (16) 0.027 (2) -0.0005 (17) 0.018 (2) C43 0.025 (2) 0.0011 (18) -0.0016 (17) 0.021 (2) -0.0004 (17) 0.023 (2) C45 0.027 (2) 0.002 (2) -0.0045 (19) 0.033 (3) 0.0048 (19) 0.027 (2) C46 0.021 (2) -0.0009 (19) -0.0060 (17) 0.030 (3) -0.0033 (18) 0.025 (2) C49 0.022 (2) -0.009 (2) -0.0041 (17) 0.036 (3) -0.0031 (19) 0.023 (2) C50 0.031 (3) -0.008 (2) -0.008 (2) 0.040 (3) 0.002 (2) 0.028 (2) C51 0.037 (3) -0.006 (2) -0.004 (2) 0.028 (3) -0.001 (2) 0.036 (3) C52 0.031 (3) -0.002 (2) 0.000 (2) 0.035 (3) -0.008 (2) 0.033 (3) C53 0.020 (2) -0.0064 (19) -0.0050 (18) 0.028 (3) -0.0018 (19) 0.031 (2) C54 0.021 (2) 0.0002 (19) -0.0047 (17) 0.030 (3) -0.0080 (18) 0.021 (2) C55 0.022 (2) -0.005 (2) -0.0028 (18) 0.033 (3) 0.0002 (19) 0.023 (2) C57 0.024 (2) 0.0013 (19) -0.0004 (17) 0.025 (2) 0.0028 (17) 0.022 (2) C60 0.032 (3) 0.021 (3) 0.014 (3) 0.070 (4) 0.030 (3) 0.052 (3) C61 0.035 (3) 0.010 (3) 0.001 (2) 0.043 (3) 0.007 (2) 0.037 (3) C62 0.027 (2) -0.008 (2) -0.0053 (19) 0.043 (3) 0.004 (2) 0.024 (2) C63 0.039 (3) -0.005 (2) -0.010 (2) 0.034 (3) 0.010 (2) 0.042 (3) C64 0.047 (3) -0.016 (3) -0.007 (2) 0.052 (4) 0.009 (2) 0.023 (2) C65 0.042 (3) -0.015 (3) 0.002 (2) 0.045 (3) -0.001 (2) 0.024 (2) C66 0.035 (3) -0.006 (2) 0.000 (2) 0.029 (3) 0.0013 (19) 0.031 (2) C67 0.029 (2) -0.003 (2) 0.0007 (19) 0.026 (2) 0.0017 (18) 0.025 (2)

Table 3 . Geometric parameters (Å, °) O7—C3 1.432 (6) O11—C10 1.244 (6) O23—C16 1.251 (6) O25—C24 1.226 (5) O40—C36 1.433 (6) O44—C43 1.226 (5) O56—C49 1.242 (6) O58—C57 1.221 (6) N9—C10 1.326 (6) N9—C8 1.447 (6) N14—C13 1.305 (6) N14—N15 1.371 (6) N15—C16 1.359 (6) N26—C24 1.358 (6) N26—C34 1.436 (6) N26—C27 1.474 (6) N42—C43 1.329 (6) N42—C41 1.456 (6) N47—C46 1.295 (6) N47—N48 1.365 (6) N48—C49 1.359 (7) N59—C57 1.349 (6) N59—C67 1.423 (6) N59—C60 1.484 (6) C1—C6 1.504 (7) C1—C2 1.515 (6) C1—C5 1.521 (7) C2—C8 1.519 (6) C2—C3 1.551 (7) C3—C4 1.536 (6) C4—C5 1.513 (7) C5—C24 1.491 (6) C5—C6 1.514 (7) C10—C12 1.526 (7)

C12—C13 1.491 (6) C13—C21 1.459 (7) C16—C22 1.455 (7) C17—C18 1.378 (8) C17—C22 1.403 (7) C18—C19 1.388 (8) C19—C20 1.383 (7) C20—C21 1.400 (7) C21—C22 1.382 (7) C27—C28 1.533 (7) C28—C29 1.494 (7) C29—C34 1.385 (6) C29—C30 1.388 (7) C30—C31 1.395 (7) C31—C32 1.378 (7) C32—C33 1.391 (7) C33—C34 1.393 (7) C68—C38 1.501 (6) C68—C39 1.508 (6) C68—C35 1.511 (6) C35—C41 1.533 (6) C35—C36 1.554 (6) C36—C37 1.517 (6) C37—C38 1.536 (7) C38—C57 1.497 (6) C38—C39 1.504 (7) C43—C45 1.536 (6) C45—C46 1.502 (7) C46—C54 1.447 (7) C49—C55 1.464 (6) C50—C55 1.364 (7) C50—C51 1.389 (7) C51—C52 1.397 (7) C52—C53 1.385 (7)

Experimental Section

180

C53—C54 1.403 (6) C54—C55 1.394 (7) C60—C61 1.507 (7) C61—C62 1.484 (7) C62—C63 1.380 (7) C62—C67 1.415 (7) C63—C64 1.394 (8) C64—C65 1.379 (8) C65—C66 1.394 (7) C66—C67 1.392 (7) O7—H7 0.8500 O40—H40 0.8500 N9—H9 0.8800 N15—H15 0.8800 N42—H42 0.8800 N48—H48 0.8800 C1—H1 0.9600 C2—H2 0.9601 C3—H3 0.9600 C4—H4A 0.9599 C4—H4B 0.9600 C6—H6A 0.9600 C6—H6B 0.9601 C8—H8A 0.9600 C8—H8B 0.9600 C12—H12A 0.9600 C12—H12B 0.9601 C17—H17 0.9601 C18—H18 0.9599 C19—H19 0.9599 C20—H20 0.9600

C27—H27A 0.9601 C27—H27B 0.9599 C28—H28A 0.9600 C28—H28B 0.9600 C30—H30 0.9599 C31—H31 0.9601 C32—H32 0.9600 C33—H33 0.9599 C68—H68 0.9599 C35—H35 0.9599 C36—H36 0.9599 C37—H37A 0.9600 C37—H37B 0.9599 C39—H39A 0.9600 C39—H39B 0.9600 C41—H41A 0.9601 C41—H41B 0.9599 C45—H45A 0.9600 C45—H45B 0.9601 C50—H50 0.9599 C51—H51 0.9600 C52—H52 0.9600 C53—H53 0.9600 C60—H60A 0.9600 C60—H60B 0.9600 C61—H61A 0.9600 C61—H61B 0.9600 C63—H63 0.9600 C64—H64 0.9600 C65—H65 0.9600 C66—H66 0.9600

C10—N9—C8 123.9 (4) C13—N14—N15 117.1 (4) C16—N15—N14 127.4 (4) C24—N26—C34 125.7 (4) C24—N26—C27 125.6 (4) C34—N26—C27 108.6 (4) C43—N42—C41 123.5 (4) C46—N47—N48 116.7 (4) C49—N48—N47 128.1 (4) C57—N59—C67 125.8 (4) C57—N59—C60 125.0 (4) C67—N59—C60 109.1 (4) C6—C1—C2 118.3 (4) C6—C1—C5 60.1 (3) C2—C1—C5 109.5 (4) C1—C2—C8 110.4 (4) C1—C2—C3 104.9 (4) C8—C2—C3 110.3 (4) O7—C3—C4 109.2 (4) O7—C3—C2 112.0 (4) C4—C3—C2 106.2 (4) C5—C4—C3 107.2 (4) C24—C5—C4 121.5 (4) C24—C5—C6 118.7 (4) C4—C5—C6 115.6 (4) C24—C5—C1 118.1 (4) C4—C5—C1 107.0 (4) C6—C5—C1 59.4 (3)

C1—C6—C5 60.5 (3) N9—C8—C2 115.6 (4) O11—C10—N9 122.8 (5) O11—C10—C12 120.9 (5) N9—C10—C12 116.3 (4) C13—C12—C10 115.0 (4) N14—C13—C21 122.4 (4) N14—C13—C12 116.2 (4) C21—C13—C12 121.4 (4) O23—C16—N15 120.8 (4) O23—C16—C22 124.1 (4) N15—C16—C22 115.1 (4) C18—C17—C22 119.6 (5) C17—C18—C19 120.4 (5) C20—C19—C18 120.2 (5) C19—C20—C21 119.9 (5) C22—C21—C20 119.7 (5) C22—C21—C13 118.3 (4) C20—C21—C13 121.9 (5) C21—C22—C17 120.2 (5) C21—C22—C16 119.4 (4) C17—C22—C16 120.2 (4) O25—C24—N26 121.3 (4) O25—C24—C5 122.6 (4) N26—C24—C5 116.1 (4) N26—C27—C28 104.8 (4) C29—C28—C27 103.2 (4) C34—C29—C30 119.9 (5)

Experimental Section

181

C34—C29—C28 110.5 (4) C30—C29—C28 129.6 (5) C29—C30—C31 118.6 (5) C32—C31—C30 120.9 (5) C31—C32—C33 121.2 (5) C32—C33—C34 117.3 (5) C38—C68—C39 60.0 (3) C38—C68—C35 109.0 (4) C39—C68—C35 117.4 (4) C68—C35—C41 111.0 (4) C68—C35—C36 104.6 (4) C41—C35—C36 111.1 (4) O40—C36—C37 108.7 (4) O40—C36—C35 111.7 (4) C37—C36—C35 106.9 (4) C36—C37—C38 105.2 (4) C29—C34—C33 122.0 (4) C29—C34—N26 109.2 (4) C33—C34—N26 128.8 (4) C57—C38—C68 118.3 (4) C57—C38—C39 118.4 (4) C68—C38—C39 60.2 (3) C57—C38—C37 120.3 (4) C68—C38—C37 108.0 (4) C39—C38—C37 116.3 (4) C38—C39—C68 59.8 (3) N42—C41—C35 113.8 (4) O44—C43—N42 122.9 (4) O44—C43—C45 121.3 (4) N42—C43—C45 115.8 (4) C46—C45—C43 114.5 (4) N47—C46—C54 122.8 (4) N47—C46—C45 116.1 (4) C54—C46—C45 121.0 (4) O56—C49—N48 120.8 (4) O56—C49—C55 124.0 (5) N48—C49—C55 115.1 (4) C55—C50—C51 120.3 (5) C50—C51—C52 119.7 (5) C53—C52—C51 119.6 (4) C52—C53—C54 120.6 (5) C55—C54—C53 118.4 (4) C55—C54—C46 119.3 (4) C53—C54—C46 122.4 (4) C50—C55—C54 121.3 (4) C50—C55—C49 120.8 (4) C54—C55—C49 117.9 (4) O58—C57—N59 120.5 (4) O58—C57—C38 122.0 (4) N59—C57—C38 117.5 (4) N59—C60—C61 105.6 (4) C62—C61—C60 105.6 (4) C63—C62—C67 119.2 (5) C63—C62—C61 131.4 (5) C67—C62—C61 109.4 (4) C62—C63—C64 120.0 (5) C65—C64—C63 120.0 (5) C64—C65—C66 121.7 (5) C67—C66—C65 117.8 (5) C66—C67—C62 121.1 (4)

C66—C67—N59 129.5 (4) C62—C67—N59 109.5 (4) C3—O7—H7 109.5 C36—O40—H40 109.5 C10—N9—H9 118.1 C8—N9—H9 118.1 C16—N15—H15 116.3 N14—N15—H15 116.3 C43—N42—H42 118.2 C41—N42—H42 118.2 C49—N48—H48 116.0 N47—N48—H48 116.0 C6—C1—H1 128.4 C2—C1—H1 112.8 C5—C1—H1 109.1 C1—C2—H2 112.0 C8—C2—H2 109.3 C3—C2—H2 109.8 O7—C3—H3 109.6 C4—C3—H3 110.6 C2—C3—H3 109.1 C5—C4—H4A 110.7 C3—C4—H4A 109.3 C5—C4—H4B 110.9 C3—C4—H4B 109.1 H4A—C4—H4B 109.5 C1—C6—H6A 109.2 C5—C6—H6A 124.9 C1—C6—H6B 108.9 C5—C6—H6B 125.3 H6A—C6—H6B 109.5 N9—C8—H8A 109.2 C2—C8—H8A 106.1 N9—C8—H8B 109.8 C2—C8—H8B 106.5 H8A—C8—H8B 109.5 C13—C12—H12A 106.7 C10—C12—H12A 109.4 C13—C12—H12B 106.8 C10—C12—H12B 109.3 H12A—C12—H12B 109.5 C18—C17—H17 119.9 C22—C17—H17 120.4 C17—C18—H18 120.0 C19—C18—H18 119.6 C20—C19—H19 119.8 C18—C19—H19 119.9 C19—C20—H20 120.0 C21—C20—H20 120.1 N26—C27—H27A 109.6 C28—C27—H27A 112.2 N26—C27—H27B 109.1 C28—C27—H27B 111.5 H27A—C27—H27B 109.5 C29—C28—H28A 112.6 C27—C28—H28A 109.2 C29—C28—H28B 112.1 C27—C28—H28B 110.1 H28A—C28—H28B 109.5 C29—C30—H30 120.2

Experimental Section

182

C31—C30—H30 121.2 C32—C31—H31 118.2 C30—C31—H31 120.9 C31—C32—H32 119.6 C33—C32—H32 119.3 C32—C33—H33 120.3 C34—C33—H33 122.3 C38—C68—H68 109.4 C39—C68—H68 128.9 C35—C68—H68 113.2 C68—C35—H35 111.3 C41—C35—H35 109.2 C36—C35—H35 109.6 O40—C36—H36 109.7 C37—C36—H36 111.0 C35—C36—H36 108.9 C36—C37—H37A 108.7 C38—C37—H37A 111.5 C36—C37—H37B 110.0 C38—C37—H37B 111.8 H37A—C37—H37B 109.5 C38—C39—H39A 125.6 C68—C39—H39A 109.7 C38—C39—H39B 124.6 C68—C39—H39B 109.0 H39A—C39—H39B 109.5 N42—C41—H41A 109.5 C35—C41—H41A 106.9 N42—C41—H41B 109.9 C35—C41—H41B 107.2 H41A—C41—H41B 109.5

C46—C45—H45A 107.4 C43—C45—H45A 109.5 C46—C45—H45B 106.8 C43—C45—H45B 109.1 H45A—C45—H45B 109.5 C55—C50—H50 118.8 C51—C50—H50 120.9 C50—C51—H51 119.1 C52—C51—H51 121.1 C53—C52—H52 120.3 C51—C52—H52 120.1 C52—C53—H53 120.8 C54—C53—H53 118.6 N59—C60—H60A 110.6 C61—C60—H60A 113.4 N59—C60—H60B 108.0 C61—C60—H60B 109.6 H60A—C60—H60B 109.5 C62—C61—H61A 111.1 C60—C61—H61A 107.4 C62—C61—H61B 112.0 C60—C61—H61B 111.1 H61A—C61—H61B 109.5 C62—C63—H63 120.2 C64—C63—H63 119.8 C65—C64—H64 119.3 C63—C64—H64 120.7 C64—C65—H65 119.9 C66—C65—H65 118.5 C67—C66—H66 121.5 C65—C66—H66 120.6

Experimental Section

183

C13—N14—N15—C16 3.8 (7) C46—N47—N48—C49 2.3 (7) C6—C1—C2—C8 -169.4 (4) C5—C1—C2—C8 -103.6 (5) C6—C1—C2—C3 -50.5 (6) C5—C1—C2—C3 15.2 (5) C1—C2—C3—O7 96.7 (4) C8—C2—C3—O7 -144.5 (4) C1—C2—C3—C4 -22.5 (5) C8—C2—C3—C4 96.4 (4) O7—C3—C4—C5 -99.2 (5) C2—C3—C4—C5 21.7 (5) C3—C4—C5—C24 -152.1 (4) C3—C4—C5—C6 51.3 (5) C3—C4—C5—C1 -12.3 (5) C6—C1—C5—C24 -108.4 (5) C2—C1—C5—C24 139.4 (4) C6—C1—C5—C4 110.1 (4) C2—C1—C5—C4 -2.0 (5) C2—C1—C5—C6 -112.1 (5) C2—C1—C6—C5 97.3 (5) C24—C5—C6—C1 107.4 (5) C4—C5—C6—C1 -95.3 (4) C10—N9—C8—C2 -66.5 (6) C1—C2—C8—N9 -53.5 (5) C3—C2—C8—N9 -169.0 (4) C8—N9—C10—O11 -2.1 (7) C8—N9—C10—C12 178.8 (4) O11—C10—C12—C13 154.6 (5) N9—C10—C12—C13 -26.3 (7) N15—N14—C13—C21 1.0 (7) N15—N14—C13—C12 -175.9 (4) C10—C12—C13—N14 109.6 (5) C10—C12—C13—C21 -67.3 (6) N14—N15—C16—O23 177.1 (4) N14—N15—C16—C22 -3.3 (7) C22—C17—C18—C19 0.8 (9) C17—C18—C19—C20 -1.2 (9) C18—C19—C20—C21 0.6 (9) C19—C20—C21—C22 0.3 (8) C19—C20—C21—C13 176.5 (5) N14—C13—C21—C22 -5.8 (7) C12—C13—C21—C22 171.0 (5) N14—C13—C21—C20 178.0 (5) C12—C13—C21—C20 -5.2 (7) C20—C21—C22—C17 -0.7 (7) C13—C21—C22—C17 -177.0 (5) C20—C21—C22—C16 -177.7 (5) C13—C21—C22—C16 6.0 (7) C18—C17—C22—C21 0.2 (8) C18—C17—C22—C16 177.1 (5) O23—C16—C22—C21 177.6 (5) N15—C16—C22—C21 -1.9 (7) O23—C16—C22—C17 0.7 (8) N15—C16—C22—C17 -178.8 (4) C34—N26—C24—O25 -5.6 (7) C27—N26—C24—O25 170.6 (5) C34—N26—C24—C5 174.1 (4) C27—N26—C24—C5 -9.8 (7) C4—C5—C24—O25 120.4 (5) C6—C5—C24—O25 -83.8 (6)

Experimental Section

184

C1—C5—C24—O25 -15.2 (7) C4—C5—C24—N26 -59.3 (6) C6—C5—C24—N26 96.5 (5) C1—C5—C24—N26 165.1 (4) C24—N26—C27—C28 -159.0 (5) C34—N26—C27—C28 17.7 (5) N26—C27—C28—C29 -19.0 (5) C27—C28—C29—C34 14.6 (6) C27—C28—C29—C30 -168.2 (5) C34—C29—C30—C31 -0.2 (8) C28—C29—C30—C31 -177.2 (5) C29—C30—C31—C32 3.1 (8) C30—C31—C32—C33 -3.2 (8) C31—C32—C33—C34 0.5 (8) C38—C68—C35—C41 -104.4 (4) C39—C68—C35—C41 -169.7 (4) C38—C68—C35—C36 15.5 (5) C39—C68—C35—C36 -49.8 (5) C68—C35—C36—O40 94.0 (4) C41—C35—C36—O40 -146.2 (4) C68—C35—C36—C37 -24.8 (5) C41—C35—C36—C37 95.0 (4) O40—C36—C37—C38 -96.4 (4) C35—C36—C37—C38 24.3 (5) C30—C29—C34—C33 -2.5 (8) C28—C29—C34—C33 175.0 (5) C30—C29—C34—N26 178.5 (4) C28—C29—C34—N26 -4.0 (6) C32—C33—C34—C29 2.4 (7) C32—C33—C34—N26 -178.8 (5) C24—N26—C34—C29 167.7 (4) C27—N26—C34—C29 -9.0 (5) C24—N26—C34—C33 -11.2 (8) C27—N26—C34—C33 172.1 (5) C39—C68—C38—C57 -108.4 (5) C35—C68—C38—C57 140.3 (4) C35—C68—C38—C39 -111.3 (4) C39—C68—C38—C37 110.5 (4) C35—C68—C38—C37 -0.7 (5) C36—C37—C38—C57 -154.9 (4) C36—C37—C38—C68 -14.7 (5) C36—C37—C38—C39 50.3 (5) C57—C38—C39—C68 108.1 (4) C37—C38—C39—C68 -96.6 (4) C35—C68—C39—C38 97.1 (4) C43—N42—C41—C35 -66.1 (6) C68—C35—C41—N42 -52.2 (5) C36—C35—C41—N42 -168.1 (3) C41—N42—C43—O44 -0.8 (7) C41—N42—C43—C45 179.2 (4) O44—C43—C45—C46 149.3 (4) N42—C43—C45—C46 -30.7 (6) N48—N47—C46—C54 1.1 (7) N48—N47—C46—C45 -176.5 (4) C43—C45—C46—N47 113.3 (5) C43—C45—C46—C54 -64.4 (6) N47—N48—C49—O56 178.4 (4) N47—N48—C49—C55 -2.0 (7) C55—C50—C51—C52 0.0 (8) C50—C51—C52—C53 0.5 (8) C51—C52—C53—C54 0.3 (7)

Experimental Section

185

C52—C53—C54—C55 -1.5 (7) C52—C53—C54—C46 177.2 (4) N47—C46—C54—C55 -4.6 (7) C45—C46—C54—C55 172.9 (4) N47—C46—C54—C53 176.7 (4) C45—C46—C54—C53 -5.7 (7) C51—C50—C55—C54 -1.3 (7) C51—C50—C55—C49 177.3 (5) C53—C54—C55—C50 2.0 (7) C46—C54—C55—C50 -176.7 (4) C53—C54—C55—C49 -176.6 (4) C46—C54—C55—C49 4.7 (6) O56—C49—C55—C50 -0.7 (7) N48—C49—C55—C50 179.7 (4) O56—C49—C55—C54 177.9 (4) N48—C49—C55—C54 -1.7 (6) C67—N59—C57—O58 -10.0 (8) C60—N59—C57—O58 172.8 (6) C67—N59—C57—C38 170.7 (4) C60—N59—C57—C38 -6.5 (8) C68—C38—C57—O58 -22.5 (7) C39—C38—C57—O58 -92.0 (6) C37—C38—C57—O58 113.7 (6) C68—C38—C57—N59 156.7 (4) C39—C38—C57—N59 87.2 (5) C37—C38—C57—N59 -67.1 (6) C57—N59—C60—C61 -174.1 (5) C67—N59—C60—C61 8.3 (7) N59—C60—C61—C62 -9.6 (7) C60—C61—C62—C63 -170.2 (6) C60—C61—C62—C67 7.9 (7) C67—C62—C63—C64 1.2 (8) C61—C62—C63—C64 179.1 (6) C62—C63—C64—C65 1.9 (9) C63—C64—C65—C66 -2.6 (9) C64—C65—C66—C67 0.2 (8) C65—C66—C67—C62 3.0 (8) C65—C66—C67—N59 -176.1 (5) C63—C62—C67—C66 -3.7 (8) C61—C62—C67—C66 177.9 (5) C63—C62—C67—N59 175.6 (5) C61—C62—C67—N59 -2.8 (6) C57—N59—C67—C66 -1.9 (9) C60—N59—C67—C66 175.6 (6) C57—N59—C67—C62 178.9 (5) C60—N59—C67—C62 -3.6 (6)

Experimental Section

186

5.2.2 Biological Evaluation of Bicyclo[3.1.0]hexane -based Chemical

Library

Reporter Gene Assay Screening for Inhibitory Effect s on Cellular Signaling Pathways:

The assays were performed by Dr. Dorian Fabbro and Dr. Daniel D’Orazio. For the EGF and

TNFα-involving pathways a reporter gene assay with AP-1-bla ME180 cells containing the β-

lactamase reporter gene under the control of the AP-1 promoter was used. The cell line

(catalogue nr. K1185) was purchased by Invitrogen, and the assay was modified and

validated for the use to profile kinase inhibitors that interact with cellular pathways leading to

the activation of AP-1 driven transcription based on the Invitrogen manual

(https://www.invitrogen.com/content/sfs/manuals/cellsensor_ap1blame180_man.pdf).

Protease Inhibition Assays : Either thrombin (100nM) in PBS (20 mM NaH2PO4, 30 mM

Na2HPO4, 100 mMNaCl, pH 7.4) or urokinase (62 nM) in PBS or cathepsin B (5.4 nM) in

0.1% (v/v) Brij 35 solution and a solution of (L)-Cysteine (8 mM) in PBS (352 mM KH2PO4,

48mM Na2HPO4, 4 mM ethylenediamine tetraacetic acid, pH 6.0) was incubated with 84

library members at a concentration of 10 µM in 384-well microtiter plates for 30 min at room

temperature. For the thrombin and urokinase assays, the reaction was started by the

addition of the fluorogenic substrate Z-GGR-AMC (Bachem, Bubendorf, Switzerland)

dissolved in PBS and 1% DMSO to a final concentration of 0.25 mM in a total volume of 50

µl. As positive control, for thrombin a 0.5 µM solution of the known inhibitor hirudin fragment

54-65 was employed. As a positive control for urokinase, a 120 µM solution of known

inhibitor benzamidine was used. For the cathepsin B assay, the reaction was started by the

addition of the fluorogenic substrate Arg-Arg-7-AMC (Bachem, Bubendorf, Switzerland)

dissolved in 0.1% Brij 35 solution and 1% DMSO to a final concentration of 0.15 mM in a

total volume of 50 µl. As a positive control a 10.3 µM solution of known inhibitor cystatin was

used.

The change of fluorescence signal (ex: 360 nm, em: 460 nm, cutoff: 420 nm) was recorded

over 30 min using a SpectraMaxmicroplate reader (Molecular Devices).

Every assay was performed as a triplicate.

Inhibition of Mycobacterium Tuberculosis: The assay was carried out by Dr. Ruben

Haartkoorn. Two bacterial strains were used, H37Rv (growing) and 18b (non-growing), in a

standard resazurine assay as described in 166.

Experimental Section

187

Cytoxicity Screening : The measurements were carried out by Dr. Andrea Chicca. The

cytotoxicity of the library compounds against MCF-7 (breast cancer) and U937 (lymphoma)

cells was determined after 72 hours incubation time in a WST-1 based cell viability assay as

described in 170. All compounds were tested in a concentration of 5 µM. Every test was

performed as a triplicate.

5.2.3 Synthesis of Miscellaneous Derivatives and Bi cyclo[3.1.0]hexane-

based 5-Alkynyl-deoxypyrimidine Nucleoside Analogue 243

COOMe

HO

N3

253

(1S,3S,4R,5S)-methyl 4-(azidomethyl)-3-hydroxybicyc lo[3.1.0]hexane-1-carboxylate

192 (850 mg, 2.612 mmol) was dissolved in THF (20 ml), then TBAF (3.3 ml, 3.134 mmol)

was added slowly under stirring. The reaction mixture was stirred for 1 h at RT. The reaction

was quenched by the addition of saturated aqueous NH4Cl-solution and the water phase was

extracted with EtOAc (3x 50 ml). The combined organic phases were dried over MgSO4 and

evaporated. The residue was purified by flash column chromatography (EtOAc/hex 1:1) to

give 495 mg (2.344 mmol, 90%) of the product as a colorless oil.

Rf = 0.31 (Hex/EtOAc 1:1).

1H-NMR (400 MHz, CDCl3): δ = 4.19 (dd, J = 7.0, 1.2 Hz, 1H), 3.64 (s, 3H), 3.28 (dd, J =

12.5, 6.6 Hz, 1H), 3.20 (dd, J = 12.5, 7.6 Hz, 1H), 2.62 (ddd, J = 14.6, 7.0, 1.8 Hz, 1H), 2.16

(t, J = 7.2 Hz, 1H), 1.95 (s, br, 1H), 1.86 (dd, J = 14.7, 1.2 Hz, 1H), 1.76 (ddd, J = 9.2, 5.2,

1.2 Hz, 1H), 1.53, (ddd, J = 9.0, 4.6, 1.8 Hz, 1H), 1.40, (t, J = 5.0 Hz, 1H).

13C-NMR (100 MHz, CDCl3): δ = 174.4, 76.3, 53.9, 52.1, 49.9, 36.8, 32.5, 31.4, 21.2.

IR (film): ν 3440, 2949, 2101, 1699, 1439, 1272, 1150, 1071.

HR-MS (ESI): calcd for C9H13N3NaO3 [M-H]+: 234.0849, found: 234.0844.

Experimental Section

188

(1S,3S,4R,5S)-methyl 4-(azidomethyl)-3-methoxybicy clo[3.1.0]hexane-1-carboxylate

NaH (38 mg, 60% dispersion in oil) was added to a solution of 240 (40 mg, 0.189 mmol) in 3

ml of DMF and the reaction mixture was stirred for 30 min at RT. The reaction mixture turned

slightly orange. CH3I (134 mg, 0.945 mmol) was added and the reaction mixture was stirred

at RT for 20 h. The mixture was poured on water and extracted with EtOAc (3x30 ml), the

combined organic phases were washed with brine, dried over MgSO4 and evaporated. The

residue, an orange oil, was purified by flash column chromatography (Hex/EtOAc 5:1) to give

27 mg (0.120 mmol, 63%) of the product as a colorless oil.

Rf = 0.37 (Hex/EtOAc 3:1)

1H-NMR (400 MHz, CDCl3): δ = 3.65 (s, 3H), 3.62 (d, J = 7.4 Hz, 1H), 3.28 (dd, J = 12.5, 6.6

Hz, 1H), 3.19 (s, 3H), 3.17 (dd, J = 12.5, 7.9 Hz, 1H), 2.48 (ddd, J = 14.7, 7.0, 1.8 Hz, 1H),

2.28 (t, J = 7.2 Hz, 1H), 2.00 (d, J = 14.6 Hz, 1H), 1.74 (ddd, 8.9, 5.2, 1.0 Hz, 1H), 1.46 (ddd,

J = 9.0, 4.4, 1.8 Hz, 1H), 1.26 (t, J = 4.8 Hz, 1H).

13C-NMR (100 MHz, CDCl3): δ = 174.4, 85.4, 56.5, 53.9, 51.9, 46.2, 32.8, 31.9, 30.9, 20.3.

MS (ESI): calcd for C10H16N3O3 [M-H]+: 226.12, found: 226.05 (I = 100%).

249

To a stirred solution of 195 (30 mg) in THF(0.5 ml) thionyl chloride (90 ul) was added

dropwise at rt and the resulting mixture was refluxed for 2 h. After cooling the mixture was

added dropwise to NH3 (aq., 0.5 ml) at 0°C. The solution was dried, filtr ated, the filter cake

was washed and the filtrate was added dropwise to a stirred solution of oxalyl chloride (13 ul)

with DMF (12 ul) in 0.5 ml of THF. The solvents were evaporated to afford 16 mg (>100%) of

249.

1H-NMR (400 MHz, MeOD): δ = 4.18 (d, J = 5.6 Hz, 1H), 3.48-3.38 (m , 2H), 2.69 (s, br, 1H),

2.43 (dd, J = 13.7, 6.6 Hz, 1H), 2.09 (t, J = 6.4 Hz, 1H), 1.92-1.81 (m, 2H), 1.47-1.35 (m, 2H).

13C-NMR (100 MHz, MeOD): δ = 118.7, 75.9, 64.7, 53.5, 32.9, 31.8, 21.4.

Experimental Section

189

252

(((1S,3S,4R,5S)-4-(azidomethyl)-1-iodobicyclo[3.1.0 ]hexan-3-yl)oxy)(tert-

butyl)dimethylsilane

A solution of 3 (50 mg, 0.160 mmol), iodosobenzene diacetate (56 mg, 0.176 mmol) and

iodine (44 mg, 0.176 mmol) was stirred placed above a 500 W tungsten lamp. The mixture

was refluxed and illuminated for 1 h, then additional iodosobenzene diacetate (56 mg) and

iodine (44 mg) were added and the reaction mixture was refluxed for additional 1.5 h. The

reaction mixture was cooled to RT and diluted with 10% aqueous sodium thiosulfate solution

(5 ml) and Et2O (10 ml), the phases were separated and the organic phase was dried over

MgSO4 and evaporated. The residue was purified by flash column chromatography (H/EE

100:1) to give 33 mg (0.084 mmol, 52%) of the product as colorless crystals.

1H-NMR (400 MHz, CDCl3): δ = 3.91 (d, J = 6.5 Hz, 1H), 3.40 (dd, J = 12.5, 5.8 Hz, 1H), 3.18

(dd, J = 12.2, 8.4 Hz, 1H), 2.51 (ddd, J = 14.1, 6.7, 2.0 Hz, 1H), 2.26 (d, J = 14.1 Hz, 1H),

2.11 (dd, J= 8.6, 6.0 Hz, 1H), 1.54-1.48 (m , 2H), 1.09 (ddd, J = 10.6, 7.0, 2.0 Hz, 1H), 0.85

(s, 9H), 0.03 (s, 6H).

13C-NMR (100 MHz, CDCl3): δ = 76.3, 54.2, 50.9, 49.5, 31.5, 25.7, 22.8, 17.8, 1.1, -4.8, -4.9.

IR (film): ν 2953, 2927, 2856, 2363, 2099, 1462, 1385, 1257, 1135, 1089, 1026, 930, 852,

837, 777, 744, 682.

MS (ESI): calcd for C13H25IN3OSi [M-H]+: 394.08, found: 394.05 (I = 40%).

251

tert-butyl(((1S,2R,3S,5S)-3-((tert-butyldimethylsil yl)oxy)-5-iodobicyclo[3.1.0]hexan-2-

yl)methoxy) dimethylsilane

A solution of 195 (60 mg, 0.150 mmol), iodosobenzene diacetate (53 mg, 0.165 mmol) and

iodine (42 mg, 0.165 mmol) in cyclohexane (2 ml) was stirred placed above a 500 W

tungsten lamp. The mixture was refluxed and illuminated for 2 h, then additional

iodosobenzene diacetate (53 mg) and iodine (42 mg) were added and the reaction mixture

was refluxed for additional 1.5 h. The reaction mixture was cooled to RT and diluted with 5%

Experimental Section

190

aqueous sodium thiosulfate solution (5 ml) and Et2O (20 ml). The phases were separated,

the organic phase was dried over MgSO4 and evaporated. The residue was purified by flash

column chromatography (Hex/EtOAc 200:1) to afford 28 mg (0.058 mmol, 39%) of the

product.

1H-NMR (400 MHz, CDCl3): 3.97 (d, J = 6.5 Hz, 1H), 3.61 (dd, J = 10.2, 5.7 Hz, 1H), 3.43

(dd, J = 10.2, 7.9 Hz, 1H), 2.47 (ddd, J = 13.8, 6.7, 2.0 Hz, 1H), 2.23 (d, J = 13.9 Hz, 1H),

2.06 (dd, J= 8.1, 5.7 Hz, 1H), 1.51-1.44 (m , 2H), 1.06 (ddd, J = 10.6, 7.0, 1.5 Hz, 1H), 0.93

(s, 9H), 0.85 (s, )H), 0.09 (s, 3H), 0.09 (s, 3H), 0.01 (s, 6H).

13C-NMR (100 MHz, CDCl3): 75.8, 64.7, 53.4, 49.6, 30.8, 26.0, 25.7, 22.4, 18.9, 17.5, 2.4, -

4.8, -4.9. -5.3, -5.4.

IR (film): ν 2953, 2927, 2886, 2856, 1462, 1385, 1253, 1136, 1115, 1076, 1005, 933, 835,

775, 689.

MS (ESI): calcd for C19H40IO2Si2 [M-H]+: 483.16, found: 483.05 (I = 50%).

245

(E)-N-(((1S,3S,4R,5S)-3-((tert-butyldimethylsilyl)o xy)-4-(((tert-

butyldimethylsilyl)oxy)methyl)bicyclo[3.1.0]hexan-1 -yl)carbamoyl)-3-

methoxyacrylamide

245 (271 mg, 95%) was prepared from 213 mg of 4 and freshly prepared 211 (from 1.2 g of

217) analogeously to the procedure described for 212.

Rf = 0.38 (Hex/EtOAc 2:1).

1H-NMR (400 MHz, CDCl3): δ = 9.30 (s, 1H), 8.80 (s, 1H), 7.64 (d, J = 12.4 Hz, 1H), 5.28 (d,

J = 12.4 Hz, 1H), 4.14 (d, J = 6.6 Hz, 1H), 3.70 (s, 3H), 3.63 (dd, J = 10.2, 5.6 Hz, 1H), 3.53

(dd, J = 10.2, 9.4 Hz, 1H), 2.33 (ddd, J = 13.4, 6.6, 1.7 Hz, 1H), 2.00-1.94 (m, 2H), 1.32 (t, J

= 4.5 Hz, 1H), 1.26-1.21 (m, 2H), 0.88 (s, 9H), 0.83 (s, 9H), 0.04 (s, 6H), -0.01 (s, 3H), -0.02

(s, 3H).

13C-NMR (100 MHz, CDCl3 ): δ = 167.6, 163.7, 155.4, 97.9, 74.4, 65.1, 57.4, 53.8, 40.8, 39.7,

26.8, 26.0, 25.9, 18.6, 18.3, 17.8, -4.7, -4.8, -5.3, -5.4.

MS (ESI): calcd for C24H46N2NaO5Si2 [M-H]+: 521.28, found: 521.30 (I = 100%).

Experimental Section

191

153

1-((1S,3S,4R,5S)-3-hydroxy-4-(hydroxymethyl)bicyclo [3.1.0]hexan-1-yl)pyrimidine-

2,4(1H,3H)-dione

153 (48 mg, 75%) was prepared from 135 mg of 245 with 2.7 ml of H2SO4 (aq., 1 M) in 1 ml

of dioxane analogeously to the procedure described for 213a.

Rf = 0.40 (EtOAc/MeOH 6:1).

1H-NMR (400 MHz, MeOD): δ = 7.66 (d, J = 10.1 Hz, 1H), 5.61 (d, J = 10.1 Hz, 1H), 4.21 (d,

J = 6.6 Hz, 1H), 3.75 (dd, J = 10.1, 5.6 Hz, 1H), 3.67 (dd, J = 10.2, 5.6 Hz, 1H), 2.38 (ddd, J

= 13.4, 6.6, 1.7 Hz, 1H), 2.12-2.05 (m, 2H), 1.73 (ddd, J = 10.2, 6.6, 1.0 Hz, 1H), 1.48 (t, J =

4.5 Hz, 1H), 1.15 (ddd, J = 13.4, 6.6, 1.7 Hz, 1H).

13C-NMR (100 MHz, MeOD): δ = 165.5, 152.9, 147.8, 101.8, 74.9, 69.7, 64.1, 53.6, 49.5,

47.4, 40.6, 28.3, 19.2.

MS (ESI): calcd for C11H15N2O2 [M-H]+: 239.10, found: 239.12 (I = 100%).

247

1-((1S,3S,4R,5S)-3-hydroxy-4-(hydroxymethyl)bicyclo [3.1.0]hexan-1-yl)-5-

iodopyrimidine-2,4(1H,3H)-dione

Iodine (14 mg, 0.053 mmol) and CAN (29 mg, 0.053 mmol) were added to a solution of 153

(25 mg, 0.105 mmol) in AcOH (3 ml). The mixture was stirred at 80°C for 4 h. The solvent

was evaporated and the residue was purified by flash column chromatography

(EtOAc/MeOH 20:1) to yield 23 mg (0.063 mmol, 60%) of the product as a yellow solid.

Rf = 0.38 (Hex/EtOAc 20:1)

1H-NMR (400 MHz, MeOD): δ = 8.20 (s, 1H), 4.27-4.24 (d, J = 7.5 Hz, 1H), 3.83-3.78 (dd, J =

10.5, 5.0 Hz, 1H), 3.77-3.72 (dd, J = 10.5, 5.0 Hz, 1H), 2.44-2.37 (ddd, J = 13.3, 7.0, 2.3 Hz,

Experimental Section

192

1H), 2.16-2.07 (m, 2H), 1.81-1.76 (ddd, J = 9.7, 4.8, 1.2 Hz, 1H), 1.53-1.48 (t, J = 5.2 Hz,

1H), 1.24-1.19 (m, 1H).

13C-NMR (300 MHz, MeOD): δ = 163.2, 153.1, 151.7, 75.6, 67.9, 64.9, 54.3, 50.2, 41.2, 28.9,

19.1.

IR (film): ν 2985, 2905, 2364, 2335, 1699, 1409, 1295, 1246, 1053, 439.

MS (ESI): calcd for C11H14IN2O2 [M-H]+: 365.00, found: 364.78 (I = 100%).

243

5-(dodec-1-yn-1-yl)-1-((1S,3S,4R,5S)-3-hydroxy-4-(h ydroxymethyl)bicyclo[3.1.0]hexan-

1-yl) pyrimidine-2,4(1H,3H)-dione

247 (20 mg, 0.055 mmol) was dissolved in DMF (2 ml), Pd(PPh3)4 (7 mg, 0.006 mmol), CuI

(2 mg, 0.011 mmol), DIPEA (19 ul, 0.165 mmol) and 1-Dodecyne (35 ul, 0.165 mmol) were

added and the reaction mixture was stirred overnight at RT. Two drops of 5% disodium salt

of an aqueous EDTA solution was added (colors turns from orange to brown) and the solvent

was evaporated. The residue was purified by flash column chromatography (EtOAc/MeOH

20:1) to afford 17 mg of product that was still containing some minor impurities. Subsequent

purification by preparative HPLC yielded 3 mg (0.008 mmol, 14%) of the product as a white

solid.

Rf = 0.22 (EtOAc/MeOH 20:1)

1H-NMR (400 MHz, MeOD): δ = 7.53 (s, 1H), 4.45-4.41 (d, J = 6.5 Hz, 1H), 3.91-3.87 (dd, J =

11.3, 2.7 Hz, 1H), 3.78-3.74 (dd, J = 11.3, 3.8 Hz, 1H), 2.54-2.47 (ddd, J = 13.4, 6.6, 2.3 Hz,

1H), 2.40-2.35 (t, J = 7.2 Hz, 2H), 2.15-2.13 (t, J = 3.3 Hz, 1H), 2.08-2.02 (d, J = 13.4 Hz,

1H), 1.84-1.79 (dd, J = 9.6, 4.7 Hz, 1H), 1.60-1.51 (m, 3H), 1.42-1.34 (m, 2H), 1.25 (br s,

12H), 1.12-1.07 (ddd, J = 9.8, 5.5, 2.4 Hz, 1H), 0.89-0.84 (t, J = 6.8 Hz, 3H).

13C-NMR (100 MHz, MeOD): δ = 167.0, 154.9, 152.0, 104.3, 97.7, 78.5, 75.0, 68.4, 56.6,

52.9, 44.0, 35.9, 33.3, 33.1, 33.0, 32.8, 32.5, 32.2, 31.5, 26.4, 22.7, 21.3, 16.9.

MS (ESI): calcd for C23H33N2O4 [M-H]-: 401.24, found: 401.04 (I = 75%).

Experimental Section

193

Biological Evaluation of 243 :

The test for inhibiting activity on mycobacterium tuberculosis was carried out by Dr. Thomas

Keller. The compound was found to be inactive.

Experimental Section

194

5.2.4 Synthesis of Bicyclo[3.1.0]hexane based Pento statin- and (S)-

Adenosylhomocysteine Analogues

5.2.4.1 Preparation of lactone (-)-44 (enantiomeric series)

In general, for the preparation of 161, procedures were followed as described in 117b and in

117a.

(1R,6S)-6-(methoxycarbonyl)cyclohex-3-enecarboxyli c acid

1,2,3,6-tetrahydrophthalic anhydride (83 g) was dispersed in MeOH, then K2CO3 (15 g) was

slowly added and the reaction mixture was stirred for 15 h at RT. The solvent was

evaporated and the residue was extracted with EtOAc. The organic phases were combined,

dried over MgSO4 and evaporated to yield 100.5 g (100%) of 175 (rac) as a white solid.

1H-NMR (400 MHz, CDCl3): δ = 5.62 (m, 2H), 3.65 (s, 3H), 3.08-3.02 (m, 2H), 2.59-2.50 (m,

2H), 2.39-2.30 (m, 2H).

13C-NMR (100 MHz, CDCl3): δ = 179.7, 173.8, 125.3, 125.1, 52.0, 39.6, 39.5, 25.8, 25.6.

COOMe

COO HNH2

HOH

Bn

264

175 (rac) (100 g) and (-)-ephedrine (90 g) each dissolved in 270 ml of EtOH at 70°C were

slowly combined. After 12 h at RT the crystals that had formed were titrated with cold

EtOH/EtOAc and filtrated and then recrystallized from 380 ml of EtOH , then a second and a

third time recrystallized from 250 ml of EtOH to afford 45 g (47 %, corresponding to the

racemate) of 264 as white crystals.

Mp. 147°C.

Experimental Section

195

[αααα]D20 = -35.7° (c = 2, MeOH).

1H-NMR (400 MHz, CDCl3): δ = 8.03 (s, br, 3H), 7.36-7.19 (m, 5H), 5.68-5.57 (m, 2H), 5.23

(d, J = 2 Hz, 1H), 3.59 (s, 3H), 3.10 (dd, J = 6.8, 2.2 Hz, 1H), 2.97-2.90 (m, 2H), 2.65 (s, 3H),

2.60-2.47 (m, 2H), 2.33-2.23 (m, 2H), 1.03 (d, J = 6.6 Hz, 3H).

13C-NMR (100 MHz, CDCl3): δ = 180.5, 175.3, 140.5, 128.2, 127.3, 126.3, 125.8, 125.1, 70.9,

61.6, 51.5, 41.8, 40.3, 31.3, 27.1, 26.1, 9.2.

264 was dissolved in cold 2 N H2SO4 (~200 ml) and extracted with Et2O (2x 400 ml), the

organic phases were washed with 2 N H2SO4 (~50 ml), then with water (~50 ml), dried over

MgSO4 and evaporated to give 24.3 g (47%, corresponding to the racemate) of (-)-175 as a

colorless oil.

[αααα]D20 = -10.6° (c = 2.18, acetone).

1H-NMR (400 MHz, CDCl3): δ = 5.61 (m, 2H), 3.67 (s, 3H), 3.08-3.00 (m, 2H), 2.60-2.51 (m,

2H), 2.39-2.30 (m, 2H).

13C-NMR (100 MHz, CDCl3): δ = 179.7, 173.7, 125.2, 125.0, 52.0, 39.6, 39.5, 25.8, 25.6.

177

(3aS,7aR)-3a,4,7,7a-tetrahydroisobenzofuran-1(3H)-o ne

To a solution of 175 (75.1 g) in dichloromethane, oxalylchloride (52 ml) was added slowly at

0°C under stirring. After gas development had cease d (1-2 h) the reaction mixture was stirred

at RT under Ar overnight. The mixture was concentrated under educed pressure to afford

80.6 g (98%) of the acid chloride derivative as a slightly yellow liquid.

1H-NMR (400 MHz, CDCl3): δ = 5.71 (m, 2H), 3.69 (s, 3H), 3.50-3.14 (m, 2H), 2.68-2.42 (m,

4H).

13C-NMR (100 MHz, CDCl3): δ = 174.3, 172.4, 125.5, 123.7, 52.3, 40.5, 26.0.

Experimental Section

196

NaBH4 (28.6 g) was dissolved in 1600 ml of EtOH and cooled to -40°C , then the acid

chloride derivative (80.6 g) in 160 ml of dry THF was added slowly under stirring. After gas

evaporation had ceased the reaction mixture was stirred for 2 h at -40°C, then it was

quenched at this temperature with 4N H2SO4 until a pH of 2 was reached. The solvents were

evaporated and the residue was mixed with 600 ml of water and extracted with Et2O. The

organic phases were washed with aq. NaHCO3 and brine, dried over MgSO4 and

concentrated under reduced pressure to yield approx. 80 g (the product was not completely

dried) of a mixture of 177 and the hydroxyl-methylester derivative. The mixture was dissolved

in 480 ml of toluene, 10-campher sulfonic acid (160 mg) was added and the reaction mixture

was refluxed in the water separator for approx. 3 h. The mixture was concentrated under

reduced pressure and purified by a Kugelrohrdistillation to give 51.9 g (94%, over 2 steps) of

the product as a colorless oil.

[αααα]D20 = +59.50° (c = 2, EtOAc)

ee ≥ 85%. The enantiomeric purity was determined by HPLC analysis using a DAICEL chiral

column.

1H-NMR (400 MHz, CDCl3): δ = 5.61 (m, 2H), 3.67 (s, 3H), 3.08-3.00 (m, 2H), 2.60-2.51 (m,

2H), 2.39-2.30 (m, 2H).

13C-NMR (100 MHz, CDCl3): δ = 179.3, 130.0, 125.1, 72.5, 37.2, 32.1, 24.9, 22.2.

From 177, lactone 161 was prepared according to procedures described in 117b and 117a, in 6

steps with an overall yield of 23%.

161

(3aS,4R,5S,6aR)-5-hydroxy-4-(hydroxymethyl)hexahydr o-1H-cyclopenta[c]furan-1-one

To a solution of 182 (5.2 g) and trimethylborate (9.4 ml, 0.084 mol, distilled prior to use) in 75

ml of THF a solution of boran-dimethylsulfid complex (5.3 ml, 10-10.2 M solution of SMe2 in

BH3) in 25 ml of THF was added drop by drop at 5°C. Af ter gas evaporation ceased the

reaction mixture was slowly, during 45 min, warmed to 15°C. The reaction was quenched

with 35 ml of H2O that was added slowly and the mixture was evaporated to dryness. The

residue was dissolved in 100 ml of MeOH and evaporated to remove boric acid (3x). The

Experimental Section

197

crude product was purified by flash column chromatography (EtOAc/MeOH 6:1) to afford 4 g

(83 %) of the product as a colorless oil.

Rf = 0.16 (EtOAc/MeOH 9:1)

[αααα]D20 = -54.60° ± 0.96° (c = 1.07, MeOH)

1H-NMR (400 MHz, D6-acetone): δ = 4.45 (dd, J = 9.1, 8.0 Hz, 1H), 4.22 (dd, J = 9.1, 2.8 Hz,

1H), 4.15-4.11 (m, 1H), 3.60 (dd, J = 10.7, 6.3 Hz, 1H), 3.51 (dd, J = 10.7, 6.8 Hz), 2.97 (tdd,

J = 11.5, 9.9, 3.3 Hz, 1H), 2.81 (dddd, J = 10.5, 8.1, 5.3, 2.7 Hz, 1H), 2.24 (ddd, J = 13.4,

9.8, 5.6 Hz, 1H), 2.00 (q, J = 5.6 Hz, 1H), 1.92 (dt, J = 13.6, 3.5 Hz, 1H)

13C-NMR (100 MHz, D6-acetone): δ = 181.3, 75.6, 75.3, 63.5, 57.7, 42.6, 41.7, 38.1.

HR-MS (ESI): calcd for C8H12O4 [M+H]+: 172.0808, found: 173.0800.

5.2.4.2 Preparation of Advanced Intermediate (+)-4

183

(1R,2S,3R,4S)-methyl 2-(bromomethyl)-4-hydroxy-3-

(hydroxymethyl)cyclopentanecarboxylate

To a solution of 161 (4 g, 0.023 mol) in MeOH (120 ml) bromotrimethylsilane (17.8 g, 0.116

mol) was added slowly during 10 min at 0°C. Then Zn Br2 (0.52 g, 0.002 mol) was added and

the reaction mixture was stirred at RT for 25 h. The solvent was evaporated at a water bath

temperature below 30°C and the crude product, an or ange oil, was purified by flash column

chromatography (EtOAc/MeOH 12:1) to yield 3.26 g (0.012 mol, 53%) of the product as a

slightly yellow oil.

Rf = 0.45 (EtOAc/MeOH 12:1)

1H-NMR (400 MHz, CDCl3): δ = 5.00 (s, br, 2H), 4.29-4.23 (m, 1H), 3.96-3.89 (m, 1H), 3.75

(s, 3H), 3.75-3.69 (m, 1H), 3.60-3.55 (m, 1H), 3.47-3.41 (m, 1H), 3.16-3.09 (m, 1H), 2.43-

2.34 (m, 1H), 2.26-2.19 (m, 1H), 2.15-2.10 (m, 1H), 2.20-1.96 (m, 1H).

13C-NMR (300 MHz, CDCl3): δ = 176.3, 76.1, 63.4, 54.3, 52.5, 45.9, 45.3, 37.1, 33.4.

Experimental Section

198

HR-MS (ESI): calcd for C9H16BrO4 [M+H]+ : 267.0232, found: 267.0231.

184

(1R,2S,3R,4S)-methyl 2-(bromomethyl)-4-((tert-butyl dimethylsilyl)oxy)-3-(((tert-

butyldimethylsilyl)oxy)methyl)cyclopentanecarboxyla te

To a solution of 183 (430 mg, 1.610 mmol) in 30 ml of DMF MTBSA (3 g, 0.016 mol) was

added drop by drop (over 10min) under cooling to 0°C. The reaction mixture was stirred for

24 h at room temperature. The CH2Cl2 was evaporated and the resulting oil was dried under

high vacuum conditions for 12 h to remove excessive MTBSA. The crude product was

purified by flash column chromatography (CH2Cl2) to afford 463 mg (0.934 mmol, 58%) of the

product as a very slightly yellow oil.

Rf = 0.65 (CH2Cl2).

[αααα]D20 = -11.83° ± 0.06° (c = 1, EtOAc).

1H-NMR (400 MHz, CDCl3): δ = 4.04-3.97 (m, 1H), 3.68 (s, 3H), 3.66-3.61 (m, 2H), 3.58-

3.54 (m, 2H), 2.94-2.86 (m, 1H), 2.16-2.07 (m, 1H),1.95-1.85 (m, 2H), 0.88 (s, 9H), 0.85 (s,

9H), 0.04 (s, 3H), 0.03 (s, 3H), 0.02 (s, 3H), 0.01 (s, 3H).

13C-NMR (300 MHz, CDCl3): δ = 1714.3, 72.5, 61.5, 54.2,51.6, 43.6, 43.2, 37.9, 34.7, 26.1,

25.7, 18.1, 17.6, -4.4, -4.8, -5.5, -5.9.

IR (film): ν 2929, 2857, 1735, 1471, 1435, 1361, 1252, 1109, 883, 833, 774, 669.

HR-MS (ESI): calcd for C21H43BrO4Si2 [M+H]+:495.1956, found: 495.1940.

185

(1S,3S,4R,5S)-methyl 3-((tert-butyldimethylsilyl)ox y)-4-(((tert-

butyldimethylsilyl)oxy)methyl) bicyclo[3.1.0]hexane -1-carboxylate

184 (90 mg, 0.181 mmol) was dissolved in 1 ml of tert-BuOH, then tert-BuOK (23 mg, 0.200

mmol) dissolved in 1 ml of tert-BuOH was added slowly under stirring. The resulting

Experimental Section

199

suspension was stirred for 1.5 h at room temperature. The reaction mixture was poured on

40 ml of Et2O and washed with ice water (3x 7 ml). The phases were separated and the

organic phase was dried over MgSO4 and evaporated. Purification by flash column

chromatography (CH2Cl2) yielded 71 mg (0.170 mmol, 94%) of the product as a colorless oil.

Rf = 0.58 (CH2Cl2).

[αααα]D24 = -20.21° (c = 1.35, CHCl 3).

1H-NMR (400 MHz, CDCl3): δ = 4.16 (d, J = 6.6 Hz, 1H), 3.62 (s, 3H), 3.51 (dd, J = 9.9, 5.7

Hz, 1H), 3.33 (dd, J= 10.1, 7.8 Hz, 1H), 2.49 (ddd, J = 14.2, 6.7, 1.6 Hz, 1H), 2.06 (dd, J =

7.8, 5.7 Hz, 1H), 1.79 (d, J = 14 Hz, 1H), 1.69 (ddd, 8.8, 5.0, 1.3 Hz, 1H), 1.46 (dd, J = 5.4,

4.2 Hz, 1H), 1.42-1.38 (m, 1H), 0.88 (s, 9H), 0.84 (s, 9H), 0.03 (s, 3H), 0.02 (s, 3H), 0.01 (s,

6H).

13C-NMR (100 MHz, CDCl3): δ = 175.1, 75.8, 64.8, 53.6, 50.7, 36.3, 31.9, 30.9, 25.9, 25.8,

20.2, 18.4, 17.3, -4.7, -4.8, -5.5.

IR (film): ν 2930, 2857, 2357, 1726, 1467, 1439, 1364, 1253, 1147, 1079, 1004, 939, 835,

776, 748, 662.

HR-MS (ESI): calcd for C21H43O4Si2 [M+H]+: 415.2694, found: 415.2693.

195

(1S,3S,4R,5S)-3-((tert-butyldimethylsilyl)oxy)-4-(( (tert-

butyldimethylsilyl)oxy)methyl)bicyclo [3.1.0] hexan e-1-carboxylic acid

185 (0.97 g) was dissolved in EtOH (15 ml) and KOH (0.39 g) was added. The reaction

mixture was heated in the microwave at 100°C for 2. 5 h, then it was stirred at RT overnight.

The solvent was evaporated and the residue was mixed with 50 ml of EtOAc and 15 ml of

iced water, then 2 N aqueous HCl was added under vivid stirring until the pH reached 2. The

phases were separated and the water phase was extracted with EtOAc (2x 50 ml). The

combined organic phases were dried and evaporated. The residue was purified by flash

column chromatography (CH2Cl2/Et2O 4:1--> CH2Cl2) to yield 0.84 g (90%) of the product as

a slightly yellow solid.

Rf = 0.13 (CH2Cl2), 0.41 (Et2O)

[a] 24D = -24.65° (c = 1, CHCl 3)

Experimental Section

200

1H-NMR (400 MHz, CDCl3): δ = 4.17 (d, J = 6.5 Hz, 1H), 3.52 (dd, J = 10.2, 5.6 Hz, 1H),

3.33 (dd, J = 10.0, 7.8 Hz, 1H), 2.48 (ddd, J = 14.0, 6.7, 1.0 Hz, 1H), 2.08 (dd, J = 7.9, 5.6

Hz, 1H), 1.81-1.75 (m, 2H), 1.55 (dd, J = 5.3, 4.0 Hz, 1H), 1.49 (ddd, J = 9.2, 4.2, 1.4 Hz,

1H), 0.88 (s, 9H), 0.84 (s, 9H), 0.03 (s, 3H), 0.02 (s, 3H), 0.01 (s, 3H), 0.01 (s, 3H).

13C-NMR (100 MHz, CDCl3): δ = 180.5, 75.0, 64.1, 53.2, 36.9, 33.1, 30.1, 25.9, 25.8, 21.3,

18.3, 17.8, -4.7, -4.9, -5.5.

IR (film): ν 2931, 2857, 2361, 1686, 1467, 1254, 1082, 1044, 917, 837, 776, 660.

HR-MS (ESI): calcd for C20H41Na2O4Si2 [M+H]+: 445.2177, found: 445.2178.

196

benzyl ((1S,3S,4R,5S)-3-((tert-butyldimethylsilyl)o xy)-4-(((tert-

butyldimethylsilyl)oxy)methyl) bicyclo[3.1.0]hexan- 1-yl)carbamate

195 (0.69 g) was dissolved in toluene (17 ml) and cooled to 0°C, then DPPA (398 ul) and

Et3N (333 ul) were added and the reaction mixture was stirred for 1 h at 0°C, then 1.5 h at RT

and for 2 h at 80°C. The colorless solution turned slightly yellow upon formation of the

isocyanate. The reaction mixture was cooled to RT, then BnOH (455 ul) and Di-n-

butyltindilaurate (114 mg) were added and the mixture was stirred at 80°C for 2 h, then at

100°C for 15 min. The mixture was cooled to RT, dil uted with Et2O (100 ml) and washed with

saturated aqueous NaHCO3 solution (2x 15 ml) and water (2x 30 ml). The combined organic

phases were dried and evaporated to yield the crude product as a yellow oil that was purified

by flash column chromatography (CH2Cl2 --> CH2Cl2/MeOH 15:1) to afford 0.77 g (88%) of

the product as a colorless oil.

Rf = 0.26 (CH2Cl2)

[a] 25D = -5.66° (c = 1.3 in CHCl 3)

1H-NMR (400 MHz, CDCl3): δ = 7.38-7.29 (m, 5H), 5.07 (s, br, 2H), 4.16 (d, J = 5.5 Hz, 1H),

3.72-3.52 (m, 2H), 2.39-2.29 (m, 1H), 2.01-1.95 (m, 1H), 1.92 (d, J = 13.3 Hz, 1H), 1.59-1.50

(m, 1H), 1.34-1.30 (m, 1H), 0.92 (s, 9H), 0.86 (s, 9H), 0.84-0.81 (m , 1H), 0.08 (m, 6H), 0.02

(s, 3H), 0.02 (s, 3H).

13C-NMR (100 MHz, CDCl3): δ = 136.6, 128.6, 128.0, 74.3, 64.7, 54.0, 29.4, 26.0, 25.6, 18.3,

18.0, -4.6, -4.8, -5.3, -5.4.

Experimental Section

201

IR (film): ν 2928, 2856, 2359, 1714, 1498, 1462, 1255, 1219, 1083, 835, 774, 666.

MS (ESI): calcd for C27H48NO4Si2 [M+H]+: 506.31, found: 506.32 (I = 100%).

TBSO

TBSONH2

4

(1S,3S,4R,5S)-3-((tert-butyldimethylsilyl)oxy)-4-(( (tert-

butyldimethylsilyl)oxy)methyl)bicyclo[3.1.0] hexan- 1-amine

To a solution of 196 (25 mg) in toluene/MeOH 1:1 (0.5 ml) 10% Pd/C (11 mg) was added and

the reaction mixture was stirred vigorously under H2 (1 bar) for 4 h at RT. The reaction

mixture was directly put on the column and purified by flash chromatography (EtOAc/MeOH

20:1) to afford 16 mg (88%) of the product as a yellow oil.

Rf = 0.24 (EtOAc/MeOH 20:1)

[a] 24D = +11.42° (c = 0.8 in MeOH)

1H-NMR (400 MHz, MeOD): δ = 4.16 (d, J = 6.5 Hz, 1H), 3.60 (dd, J = 10.1, 5.6 Hz, 1H),

3.40 (dd, J = 10.3, 8.9 Hz, 1H), 2.08 (ddd, J = 13.5. 6.6, 2.0 Hz, 1H), 1.90 (dd, J = 8.9, 5.6

Hz, 1H), 1.86 (d, J = 13.5 Hz, 1H), 1.06 (t, J = 4.3 Hz, 1H), 0.97 (ddd, J = 9.1, 3.9, 1.2 Hz,

1H), 0.92 (s, 9H), 0.86 (s, 9H), 0.69 (ddd, J = 9.1, 4.4, 2.2 Hz, 1H), 0.08 (m, 6H), 0.02 (s,

3H), 0.02 (s, 3H).

13C-NMR (100 MHz, CDCl3): δ = 75.0, 65.5, 54.1, 45.4, 42.9, 27.7, 26.1, 25.6, 18.8, 18.3,

17.7, -4.7, -4.9, -5.4.

HR-MS (ESI): calcd for C19H42NO2Si2 [M-H]+: 372.2749, found: 372.2743.

Experimental Section

202

5.2.4.3 Attempted Preparation of Pentostatin Analog ue 6

162

Compound 162 was prepared by Kurt Hauenstein according to a literature procedure156b

starting from N-Cbz-(L)-vinylglycine methyl ester in an amount of 170 mg.

163

4-((R)-2-azido-1-((tert-butyldimethylsilyl)oxy)ethy l)-1-((1S,3S,4R,5S)-3-((tert-

butyldimethyl silyl)oxy)-4-(((tert-butyldimethylsil yl)oxy)methyl)bicyclo[3.1.0]hexan-1-

yl)-1H-imidazol-5-amine

After co-evaporation with toluene, 4 (22 mg) was dissolved in acetonitrile (0.6 ml) and added

to a sol. of 162 (9 mg) with mol. sieves (4 Å) in acetonitrile. The reaction mixture was refluxed

at 90°C for 2 h. Additional 9 mg of 162 were added and the reaction mixture was stirred for 1

h. The solvent was evaporated and the residue was purified by flash column chromatography

(EtOAc/Hex 2:3, 0.2% Et3N) to afford 27 mg (71%, based on mass recovery) of 163 as a light

brown oil.

Note: Additional peaks in the 1H-NMR indicated a partial decomposition. Purity was

estimated to be approx. 50%.

Rf = 0.51 (Hex/EtOAc 3:2, 0.2% Et3N )

1H-NMR (500 MHz, MeOD): δ = 7.80 (s, 1H), 7.18 (s, 1H), 4.58 (s, 2H), 4.23 – 4.13 (m, 1H),

3.88-3.81 (m, 1H), 3.68 – 3.65 (m, 1H), 3.58-3.54 (m, 1H), 2.25-2.20 (m, 1H), 2.15-2.11 (m,

1H), 1.92-1.88 (m, 1H), 1.76-1.73 (m, 1H), 1.57-1.53 (m, 1H), 1.15-1.12 (m, 1H), 0.96-0.86

(m, >27H), 0.20-0.02 (m , 18H).

MS (ESI): calcd for C30H61N6O3Si3 [M-H]+: 637.41, found: 637.70 (I = 100%).

Experimental Section

203

165

(E)-N'-(4-((R)-2-azido-1-((tert-butyldimethylsilyl) oxy)ethyl)-1-((1S,3S,4R,5S)-3-((tert-

butyldimethyl silyl)oxy)-4-(((tert-butyldimethylsil yl)oxy)methyl)bicyclo[3.1.0]hexan-1-

yl)-1H-imidazol-5-yl)-N,N-dimethylformimidamide

After co-evaporation with toluene (2x 3 ml), 4 (16 mg, 0.043 mmol) was dissolved in

acetonitrile (0.5 ml) and added to a sol. of 162 (20 mg, 0.065 mmol) and 2,2,2-trifluoro-

ethanol (43 ul, 0.004 mmol) with mol. sieves (4 Å) in acetonitrile. The reaction mixture was

refluxed at 90°C for 6 h. Dimethylformamide dimethy l acetal (23 ul, freshly distilled) was

added and the reaction mixture was refluxed for 13 h to give 28 mg (0.040 mmol, 94% based

on mass recovery) of the product as a brown oil.

Note: The 1H-NMR spectrum indicated the presence of both 163 and 165 in an approx. 1:1

mixture. Additional peaks indicated a partial decomposition.

Rf = 0.33 (Hex/EtOAc 10:1, 0.2% Et3N)

[a] 24D = -13.88° (c = 1 in CHCl 3)

1H-NMR (500 MHz, C6D6): δ = 7.33 (s, 1H), 6.99 (s, 1H), 5.66 – 5.47 (m, 1H), 4.21 (d, J = 5.8

Hz, 1H), 4.00 – 3.96 (m, 1H), 3.96 – 3.91 (m, 1H), 3.62 – 3.54 (m, 1H), 3.42 – 3.37 (m, 1H),

3.18 (d, J = 4.7 Hz, 3H), 2.52 (s, 3H), 2.25 (dd, J = 15.3, 7.8 Hz, 1H), 2.21 – 2.10 (m, 2H),

1.83 – 1.71 (m, 1H), 1.66-1.50 (m, 1H), 1.15-1.12 (m, 1H), 1.00 (s, 9H), 0.99 (s, 9H), 0.98 (s,

9H), 0.22 (s, 3H), 0.17 (s, 3H), 0.15 (s, 3H), 0.11 (s, 3H), 0.08 (s, 3H), 0.06 (s, 3H).

IR (film): ν 2929, 2858, 2363, 2336, 1769, 1651, 1458, 1386, 1249, 839, 791, 779, 658.

MS (ESI): calcd for C33H66N7O3Si3 [M-H]+: 692.45, found: 692.60 (I = 100%).

Experimental Section

204

6

(R)-8-(tert-butyldimethylsilyloxy)-3-((1S,3S,4R,5S) -3-(tert-butyldimethylsilyloxy)-4-((tert-

butyldimethylsilyloxy)methyl)bicyclo[3.1.0]hexan-1- yl)-3,6,7,8-tetrahydroimidazo[4,5-

d][1,3]diazepine

After co-evaporation with toluene, 4 (10 mg, 0.027 mmol) was dissolved in 1,2-

dichloroethane (0.3 ml), 162 (12 mg) and trifluoroethanol (27 ul) were added and the reaction

mixture was stirred at 80°C for 4 h. TLC indicated still starting material, so an additional 6 mg

of 162 were added and the mixture was stirred for additional 3 h at 80°C. TLC indicated that

all starting material had been consumed. The solvent was evaporated and the imidazole

intermediate was purified by flash chromatography (Hex/EtOAc 3:2,0.2% Et3N) to afford 4 mg

(0.006 mmol, 23%) of 163 that were immediately reacted with dimethylformamide dimethyl

acetal (15 ul) in dichloroethane (0.3 ml) at 80°C f or 14 h to afford crude 165. The

dichloroethane was evaporated and MeOH (0.3 ml), propanedithiol (19 ul) and Et3N (38 ul)

were added. The reaction mixture was stirred at 50°C for 12 h. Evaporation of the solvent

and purification by flash chromatography (Hex/EtOAc 10:1, 0.2% Et3N) yielded 1.3 mg of the

product (0.002 mmol, 7% over 3 steps).

Rf = 0.4 (EtOAc/MeOH 20:1)

MS (ESI): calcd for C33H66N7O3Si3 [M-H]+: 621.41, found: 621.50 (I = 100%).

1H-NMR indicated the presence of the title compound with 2 of the TBS groups removed, in

small quantity, along with large amounts of grease and additional peaks in the region of δ =

2.6 and 4.0.

Experimental Section

205

5.2.4.4 Preparation of S-Adenosylhomocysteine Analo gue 7

280

9-((1S,3S,4R,5S)-3-((tert-butyldimethylsilyl)oxy)-4 -(((tert-butyldimethylsilyl)oxy)methyl)

bicyclo[3.1.0]hexan-1-yl)-6-chloro-9H-purine

A mixture of 4 (118 mg), 4,6-dichloro-5-formamidopyrimidine (219, 122 mg), Et3N (176 ul) in

1,4-dioxane (3 ml) with mol. sieves was gently refluxed for 24 h at 110°C. The solvent was

evaporated to give approximately 250 mg of crude 279 (brown oil) that was purified by flash

column chromatography (H/EE 2:1) to give 112 mg of 279 as a yellow oil that was

immediately treated with diethoxymethyl acetate (3 ml) and heated to 110°C for 20 h. The

solvents were evaporated and the residue was purified by flash column chromatography

(H/EE 7:1) to yield 72 mg (45% over 2 steps) of the product as a white solid.

Rf = 0.2 (Hex/EtOAc 7:1).

[a] 24D = -30.13° ± 1.86° (c = 1 in CHCl 3).

1H-NMR (400 MHz, CDCl3): δ = 8.71 (s, 1H), 8.19 (s, 1H), 4.30 (d, J = 6.5 Hz), 3.92 (dd, J =

10.2, 7.8 Hz, 1H), 3.85 (dd, J = 10.2, 5.8 Hz, 1H), 2.55 (ddd, J = 13.3, 6.5, 2.0 Hz, 1H), 2.23-

2.16 (m, 2H), 1.81-1.71 (m, 2H), 1.36-1.31 (m, 1H), 0.91 (s, 9H), 0.87 (s, 9H), 0.10 (s,3H),

0.09 (s, 3H), 0.04 (s, 3H), 0.02 (s, 3H).

13C-NMR (100 MHz, CDCl3): δ = 151.5, 150.9, 146.4, 131.5, 74.4, 64.6, 53.0, 43.4, 41.6,

26.5, 26.0, 25.7, 18.5, 17.8, 16.9, -4.7, -4.8, -5.2, -5.3.

IR (film): ν 2953, 2928, 2885, 2857, 2363, 1699, 1589, 1557, 1490, 1472, 1427, 1407, 1389,

1361, 1337, 1254, 1220, 1091, 1070, 1023, 934, 849, 835, 776, 636, 567.

HR-MS (ESI): calcd for C24H42ClN4O2Si2 [M+H]+: 509.2529, found: 509.2535.

Experimental Section

206

167

9-((1S,3S,4R,5S)-3-((tert-butyldimethylsilyl)oxy)-4 -(((tert-

butyldimethylsilyl)oxy)methyl)bicyclo[3.1.0]hexan-1 -yl)-9H-purin-6-amine

A saturated solution of ammonia in methanol was prepared and 280 (25 mg) was dissolved

in 5 ml of it. The reaction mixture was heated to 100°C in the microwave for 4 h. The solvent

was evaporated and the crude product was purified by flash chromatography (CH2Cl2/MeOH

20:1 & 0.5% Et3N) to yield 21 mg (87%) of the product as a colorless oil that turned into a

white solid at -20°C.

Rf = 0.1 (CH2Cl2/MeOH 20:1 0.5% Et3N)

[a] 20D = -23.94° ± 0.38° (c = 1.13 in CHCl 3)

1H-NMR (400 MHz, MeOD): δ = 8.17 (s, 1H), 8.08 (s, 1H), 4.40 (d, J = 7.1 Hz, 1H), 3.99 (dd,

J = 10.3, 8.3 Hz, 1H), 3.91 (dd, J = 10.3, 5.5 Hz, 1H), 2.57 (ddd, J = 13.3, 6.5, 2.1 Hz, 1H),

2.26 (d, J = 13.4 Hz, 1H), 2.18 (dd, J = 8.2, 5.6 Hz, 1H), 1.79 (ddd, J = 9.5, 4.8, 1.1 Hz, 1H),

1.73 (t, J = 5.1 Hz, 1H), 1.41 – 1.35 (m, 1H), 0.95 (s, 9H), 0.91 (s, 9H), 0.15 (s, 3H), 0.14 (s,

3H), 0.09 (s, 3H), 0.08 (s, 3H).

13C-NMR (100 MHz, MeOD): δ = 153.7, 142.8, 76.4, 66.1, 54.9, 44.8, 42.6, 27.4, 26.6, 26.4,

20.2, 18.8, 17.1, -4.3, -4.6.

IR (film): ν 3325, 2952, 2928, 2856, 1614, 1573, 1471, 1390, 1294, 1253, 1190, 1085, 1040,

958, 835, 776, 650.

HR-MS (ESI): calcd for C24H44N5O2Si2 [M+H]+: 490.3028, found: 490.3025.

TBSO

TBSON

N

NN

NHAloc

283

Allyl-(9-((1S,3S,4R,5S)-3-((tert-butyldimethylsilyl )oxy)-4-(((tert-

butyldimethylsilyl)oxy)methyl) bicyclo[3.1.0]hexan- 1-yl)-9H-purin-6-yl)carbamate

To a solution of 167 (10 mg, 0.020mmol) in CH2Cl2 (0.3 ml), N-methylimidazole (5 ul, 0.060

mmol) and allychloroformate (7 ul, 0.060 mmol) were added. The reaction mixture was stirred

Experimental Section

207

for 41 h at RT. The solvent was evaporated to afford the crude product that was purified by

flash chromatography (EtOAc/Hex 1:1) to yield 7 mg (0.012 mmol, 61%) of the product as a

white solid.

Rf = 0.4 (Hex/EtOAc 1:1)

1H-NMR (400 MHz, MeOD): δ = 8.58 (s, 1H), 8.30 (s, 1H), 6.05 (ddt, J = 17.2, 10.6, 5.6 Hz,

1H), 5.43 (dq, J = 17.2, 1.6 Hz, 1H), 5.28 (ddd, J = 10.5, 2.7, 1.3 Hz, 1H), 4.75 (dt, J = 5.6,

1.4 Hz, 2H), 4.42 (d, J = 7.0 Hz, 1H), 4.04 (dd, J = 10.3, 8.9 Hz, 1H), 3.94 (dd, J = 10.3, 5.8

Hz, 1H), 2.65 (ddd, J = 13.3, 6.6, 2.0 Hz, 1H), 2.26 (d, J = 13.4 Hz, 1H), 2.20 (dd, J = 8.8, 5.7

Hz, 1H), 1.82 (ddd, J = 9.4, 4.9, 1.0 Hz, 1H), 1.77 (t, J = 5.1 Hz, 1H), 1.44 (ddd, J = 9.4, 5.1,

2.1 Hz, 1H), 0.97 (s, 9H), 0.92 (s, 9H), 0.16 (s, 3H), 0.15 (s, 3H), 0.10 (s, 3H), 0.09 (s, 3H).

13C-NMR (100 MHz, MeOD): δ = 153.4, 146.6, 120.9, 119.1, 75.7, 67.7, 65.9, 54.6, 44.3,

42.7, 27.9, 26.6, 26.4, 19.4, 18.8, 17.9, -4.3, -4.7.

MS (ESI): calcd for C28H48N5O4Si2 [M+H]+: 574.32, found: 574.30 (I = 100%).

284

Allyl-(9-((1S,3S,4R,5S)-3-((tert-butyldimethylsilyl )oxy)-4-

(hydroxymethyl)bicyclo[3.1.0]hexan-1-yl)-9H-purin-6 -yl)carbamate

283 (15 mg, 0.026 mmol) was dissolved in CH2Cl2/MeOH 1:1 (0.3 ml) and cooled to 0°C.

CSA (15 mg, 0.065 mmol) was added and the reaction mixture was stirred at 0°C for 4 h.

Additional 8 mg of CSA were added and the reaction mixture was stirred for additional 1h.

The reaction was quenched with sat. aq. NaHCO3-sol. until pH reached 8 and evaporated to

give the crude product (78 mg) as a white solid. Purification by flash chromatography

(EtOAc/MeOH 30:1) gave 8 mg (0.017 mmol, 67%) of the product as a yellow oil.

Rf = 0.4 (EtOAc/MeOH 20:1)

MS (ESI): calcd for C22H34N5O4Si [M-H]+: 460.24, found: 460.20 (I = 100%).

Experimental Section

208

285

Allyl-(9-((1S,3S,4R,5S)-3-((tert-butyldimethylsilyl )oxy)-4-((methylbenzenesulfonate)

methyl) bicyclo[3.1.0]hexan-1-yl)-9H-purin-6-yl)ca rbamate

Tosyl chloride (3.2 mg) was added to a stirred solution of 284 (4 mg) and triethylamine (5 ul)

in CHCl3 (0.3 ml) under Argon at 0°C, then DMAP (0.1 mg) was added. The reaction mixture

was warmed to room temperature and stirred for 4 d. Evaporation of the solvent and

purification by flash column chromatography (EtOAc/Hex 3:2) gave 2 mg of the desired

product (36%).

Rf = 0.15 (EtOAc/Hex 3:2).

1H-NMR (400 MHz, CDCl3): δ = 8.65 (s, 1H), 8.03 (s, 1H), 7.86 (d, J = 8.3 Hz, 2H), 7.37 (d, J

= 8.0 Hz, 2H), 6.00 (ddd, J = 16.2, 10.4, 5.7 Hz, 1H), 5.42 (dd, J = 17.2, 1.4 Hz, 1H), 5.30

(dd, J = 10.4, 1.2 Hz, 1H), 4.78 (d, J = 5.8 Hz, 2H), 4.52 – 4.42 (m, 1H), 4.32 (dd, J = 10.2,

5.9 Hz, 1H), 4.21 (d, J = 7.0 Hz, 1H), 2.54 (ddd, J = 13.9, 6.7, 2.3 Hz, 1H), 2.46 (s, 3H), 2.38

(dd, J = 8.4, 5.9 Hz, 1H), 2.15 (d, J = 13.5 Hz, 1H), 1.83 – 1.77 (m, 1H), 1.68 (dd, J = 9.2, 4.8

Hz, 1H), 1.44 – 1.38 (m, 1H), 0.87 (s, 9H), 0.04 (s, 3H), 0.03 (s, 3H).

HR-MS (ESI): calcd for C29H39N5NaO6SSi2 [M-Na]+: 636.2288, found: 636.2285.

((1S,2R,3S,5S)-5-(6-amino-9H-purin-9-yl)-3-((tert-

butyldimethylsilyl)oxy)bicyclo[3.1.0]hexan-2-yl)met hanol

To a solution of 167 (6 mg, 0.012 mmol) in CH2Cl2/MeOH 1:1 (0.2 ml) CSA (10 mg, 0.042

mmol) was added at 0°C and the reaction mixture was stirred for 4 h at 0°C. The reaction

was quenched with sat. aq. NaHCO3-sol. until pH reached 8 and the solvents were

evaporated to afford ~29 mg of the crude product as a slightly yellow solid. Purification by

flash chromatography (EtOAc/MeOH 20:1 & 0.5% Et3N) afforded 4 mg (0.011 mmol, 89%) of

the product as a white solid.

Rf = 0.2 (Hex/EtOAc 20:1 0.5% Et3N)

Experimental Section

209

1H-NMR (400 MHz, MeOD): δ = 8.18 (s, 1H), 8.08 (s, 1H), 4.56 (s, 1H), 4.42 (dd, J = 6.6, 1.3

Hz, 1H), 3.96 (dd, J = 11.5, 3.9 Hz, 1H), 3.82 (dd, J = 11.5, 4.5 Hz, 1H), 3.72 – 3.62 (m, 1H),

3.59 – 3.54 (m, 1H), 2.57 (ddd, J = 13.2, 6.6, 2.3 Hz, 1H), 2.23 (d, J = 13.3 Hz, 1H), 2.17 (t, J

= 4.1 Hz, 1H), 1.84 (ddd, J = 9.6, 4.8, 1.3 Hz, 1H), 1.71 (t, J = 5.1 Hz, 1H), 1.40 (ddd, J = 9.6,

5.2, 2.3 Hz, 1H), 0.92 (s, 9H), 0.09 (s, 3H), 0.09 (s, 3H).

HR-MS (ESI): cacld for C18H30N52Si [M+H]+: 376.2169, found: 376.2158.

MS (ESI): cacld for C18H30N52Si [M+H]+: 376.2169, found: 375.89 (I = 100%).

168

Di-tert-butyl 9-((1S,3S,4R,5S)-3-(tert-butyldimethy lsilyloxy)-4-((tert-

butyldimethylsilyloxy)methyl) bicyclo[3.1.0]hexan-1 -yl)-9H-purin-6-ylcarbamate

167 (40 mg, 0.082 mmol) and DMAP (40 mg) were dissolved in DMF and evaporated under

reduced pressure to remove atmospheric water, Boc2O (71 mg) was added and the reaction

mixture was stirred for 14 h at RT. The reaction mixture as such was purified by flash column

chromatography (Hex/EtOAc 5:1, 0.5% Et3N) to afford the diprotected product 161 (49 mg,

87%).

Rf = 0.3 (Hex/EtOAc 10:1 0.5% Et3N)

[a] 24D = -19.62° (c = 1 in CHCl 3)

1H-NMR (400 MHz, CDCl3): δ = 8.83 (s, 1H), 8.11 (s, 1H), 4.33 (d, J = 6.8 Hz, 1H), 3.96 (dd,

J = 10.2, 8.1 Hz, 1H), 3.88 (dd, J = 10.2, 5.9 Hz, 1H), 2.59 (ddd, J = 13.3, 6.6, 2.1 Hz, 1H),

2.26 – 2.15 (m, 2H), 1.78 (t, J = 5.0 Hz, 1H), 1.73 (ddd, J = 9.3, 4.8, 1.0 Hz, 1H), 1.46 (s,

18H), 1.36 (ddd, J = 9.1, 5.0, 1.9 Hz, 1H), 0.94 (s, 9H), 0.89 (s, 9H), 0.12 (s, 3H), 0.11 (s,

3H), 0.06 (s, 3H), 0.05 (s, 3H).

13C-NMR (100 MHz, CDCl3): δ = 154.1, 151.9, 150.7, 150.2, 145.7, 129.4, 83.7, 74.3, 64.7,

53.1, 42.9, 41.5, 27.8, 26.4, 26.1, 25.8, 18.5, 17.7, 16.8, -4.7, -4.8, -5.3, -5.4.

IR (film): ν 2928, 2856, 2359, 2338, 1734, 1460, 1368, 1253, 1101, 837, 778.

HR-MS (ESI): calcd for C34H59N5NaO6Si2 [M+H]+: 712.3896, found: 712.3917.

Experimental Section

210

169

Di-tert-butyl (9-((1S,3S,4R,5S)-3-(( tert-butyldimethylsilyl)oxy)-4-(hydroxymethyl)-

bicyclo[3.1.0]hexan-1-yl)-9H-purin-6-yl)carbamate

To a solution of 168 (41 mg, 0.059 mmol) in CH2Cl2/MeOH1:1 (0.8 ml) CSA (14 mg, 0.059

mmol) was added at 0°C and the reaction mixture was stirred for 4 h at 0°C and for additional

2 h at 10°C. Additional 2 mg of CSA were added and the reaction mixture was stirred for

additional 3 h at 0°C. The reaction was quenched wi th sat. aq. NaHCO3-sol. until the pH

reached a value of 8 and the solvents were evaporated to give the crude product as white

solid (approx.70 mg). Purification by flash column chromatograph (Hex/EtOAc 2:1) afforded

30 mg (0.052 mmol, 88%) of the product as a colorless oil.

Rf = 0.23 (Hex/EtOAc 2:1)

[a] 24D = -13.88° (c = 1 in CHCl 3)

1H-NMR (400 MHz, CDCl3): δ = 8.83 (s, 1H), 8.17 (s, 1H), 5.65 (d, J = 10.6 Hz, 1H), 4.47 (dd,

J = 6.5, 1.3 Hz, 1H), 4.08 (d, J = 12.3 Hz, 1H), 3.86 (td, J = 11.9, 3.3 Hz, 1H), 2.65 (ddd, J =

13.0, 6.6, 2.3 Hz, 1H), 2.20 (dd, J = 2.3, 0.7 Hz, 1H), 2.15 (d, J = 13.1 Hz, 1H), 1.81 (ddd, J =

9.5, 4.8, 1.2 Hz, 1H), 1.75 (t, J = 5.0 Hz, 1H), 1.48 (s, 18H), 1.31 (ddd, J = 9.5, 5.0, 2.4 Hz,

1H), 0.90 (s, 9H), 0.06 (s, 3H), 0.05 (s, 3H).

13C-NMR (400 MHz, CDCl3): δ = 151.6, 146.2, 84.3, 76.3, 65.5, 53.1, 44.3, 43.8, 27.8, 26.8,

25.9, 17.1, -4.8, 4.9.

IR (film): ν 2963, 2928, 2856, 2363, 2321, 1760, 1599, 1368, 1259, 1090, 1024, 799, 672,

461.

HR-MS (ESI): calcd forC28H46N5O6Si [M-H]+: 576.3212, found: 576.3213.

Experimental Section

211

170

((1S,2R,3S,5S)-5-(6-((di- tert-butoxycarbonyl)amino)-9H-purin-9-yl)-3-(( tert-

butyldimethylsilyl)- oxy)bicyclo[3.1.0]hexan-2-yl)m ethyl 4-methylbenzenesulfonate

169 (30 mg) was dissolved in CHCl3 (0.4 ml). Triethylamine (10 ul), DMAP (0.5 mg) and TsCl

(13 mg, 0.066 mmol) were added and the reaction mixture was stirred for 3 d at RT (after 1 d

again 13 mg of TsCl and 18 ul of Et3N were added). The reaction mixture was directly put on

the column and purified by flash column chromatography (Hex/EtOAc 5:2) to afford 18 mg

(0.025 mmol, 75%) of the product as a colorless oil.

Rf = 0.37 (Hex/EtOAc 2:1)

[αααα]24D = -12.22° (c = 0.87 in CHCl 3)

1H-NMR (400 MHz, CDCl3): δ = 8.74 (s, 1H), 8.07 (s, 1H), 7.88 – 7.82 (m, 2H), 7.36 (dd, J =

8.5, 0.6 Hz, 2H), 4.47 (dd, J = 10.1, 8.7 Hz, 1H), 4.33 (dd, J = 10.1, 6.1 Hz, 1H), 4.20 (dd, J =

6.4, 1.0 Hz, 1H), 2.53 (ddd, J = 13.6, 6.6, 2.2 Hz, 1H), 2.45 (s, 3H), 2.37 (dd, J = 8.6, 6.1 Hz,

1H), 2.13 (d, J = 13.6 Hz, 1H), 1.79 (t, J = 5.3 Hz, 1H), 1.66 (ddd, J = 9.8, 4.9, 1.2 Hz, 1H),

1.48 (s, 18H), 1.40 (ddd, J = 9.7, 5.6, 2.2 Hz, 1H), 0.87 (s, 9H), 0.04 (s, 3H), 0.03 (s, 3H).

13C-NMR (100 MHz, CDCl3): δ = 153.9, 151.9, 150.7, 150.2, 145.3, 145.0, 132.9, 129.9,

129.3, 128.0, 83.9, 73.8, 70.7, 49.9, 42.8, 42.0, 40.9, 27.9, 25.7, 21.7, 17.9, 16.6, 14.2, -4.8,

-4.9.

IR (film): ν 2927, 2856, 2362, 2337, 1790, 1730, 1598, 1459, 1367, 1255, 1175, 1139, 1101,

1025, 957, 939, 815, 785, 696, 659, 572, 553, 530.

HR-MS (ESI): calcd for C35H52N5O8SSi [M+H]+: 730.3300, found: 730.3286.

286

(S)-tert-butyl (2-oxotetrahydrothiophen-3-yl)carbam ate

(L)-homocysteine thiolactone hydrochloride (70 mg) was slowly added to a mixture of

NaHCO3 (191 mg) in 1 ml of dioxane/H2O (1:1). The mixture was stirred for 30 min and then

Experimental Section

212

Boc2O (152 mg) was added and the reaction mixture was stirred overnight. The reaction

mixture was diluted with EtOAc, the phases were separated and the water phase was

extracted with EtOAc, the combined organic phases were washed with brine, dried and

evaporated to afford the crude product (approx. 150 mg). The crude product was purified by

flash column chromatography (EtOAc/Hex 1:4) to give 83 mg of product (white solid, 84%).

Rf = 0.3 (CHCl3/MeOH 10:1)

1H-NMR (400 MHz, CDCl3): δ = 4.96 (s, 1H), 4.36 – 4.17 (m, 1H), 3.31 (td, J = 11.7, 5.1 Hz,

1H), 3.23 (ddd, J = 11.3, 7.0, 1.2 Hz, 1H), 2.96 – 2.75 (m, 1H), 1.97 (qd, J = 12.5, 7.2 Hz,

1H), 1.45 (s, 9H).

MS (ESI): calcd for C9H15NNaO3S [M-Na]+: 240.0670, found: 240.10 (I = 100%).

287

(S)-methyl 4-((((1S,2S,3S,5S)-5-(6-(bis(tert-butoxy carbonyl)amino)-9H-purin-9-yl)-3-

((tert-butyldimethylsilyl)oxy)bicyclo[3.1.0]hexan-2 -yl)methyl)thio)-2-((tert-

butoxycarbonyl)amino)butanoate

170 (6 mg) was co-evaporated with DMF to remove atmospheric water. 286 (3 mg) was co-

evaporated with toluene. NaOMe (1.5 mg) was added to a solution of 286 in MeOH (0.2 ml)

and stirred for 1h, then 170 was added and the reaction mixture was refluxed (75°C) for 8 h.

The reaction mixture was directly put on the column and purified by flash chromatography

(Hex/EtOAc 2:1) to yield 4 mg (60%) of the product.

Note: An impurity probably derived from the reagent could not be removed, leading to

additional peaks at 5.02, 4.32, 3.50, 2.66, 2.16, 1.92, and 1.38.

Rf = 0.3 (Hex/EtOAc 2:1)

1H-NMR (400 MHz, CDCl3): δ = 8.77 (s, 1H), 8.05 (s,1H), 4.23 (d, J = 6.0 Hz, 1H), 3.74 (s,

3H), 3.53 – 3.49 (m, 1H), 3.40-3.36 (m, 1H), 2.93 – 2.88 (m, 1H), 2.62 – 2.59 (m, 1H), 2.58-

2.55 (m, 1H), 2.21 – 2.13 (m, 2H), 2.12 – 2.07 (m, 1H), 1.96-1.89 (m, 2H), 1.86-1.82 (m, 1H),

1.78 – 1.73 (m, 1H), 1.47 (s, 9H), 1.41 (s, 9H), 1.38 (s, >9H), 1.34-1.26 (m, 1H), 0.84 (s, 9H),

0.02 (s, 3H), 0.01 (S, 3H).

13C-NMR: (100 MHz, MeOD): δ = 151.9, 147.1, 84.1, 76.3, 51.4, 50.2, 49.9, 43.2, 40.3, 34.9,

31.3, 28.4, 28.1, 27.3, 26.9, 26.5, 24.9, 17.1, 16.6, -5.6, -6.0.

Experimental Section

213

MS (ESI): calcd for C38H63N6NaO9SSi [M-Na]+: 829.40, found: 829.40 (I = 100%).

173

(S)-4-((((1S,2S,3S,5S)-5-(6-(bis(tert-butoxycarbony l)amino)-9H-purin-9-yl)-3-((tert-

butyldi methylsilyl)oxy)bicyclo[3.1.0]hexan-2-yl)me thyl)thio)-2-((tert-

butoxycarbonyl)amino)butanoic acid

Methyl ester 287 (8 mg) was dissolved in THF (0.5 ml) and cooled to 0°C, then KOH (20 ul, 1

M aq. sol.) and water (50 ul) were added. The reaction mixture was stirred for 14 h. As TLC

and MS indicated the end of the reaction, the reaction mixture was cooled to 0°C and

quenched by adding HCl (20 ul, 1M aq.sol.), concentrated under reduced pressure and

purified by FC (CHCl3/MeOH/H2O 8:1:0.2%) to yield the product as a colorless solid (5.5 mg,

70%).

Note: The 1H-NMR spectrum displayed a splitting of TBS and aromatic peaks in the ratio of

2:3; and still contained some minor impurity derived from the reagent of the previous step.

The impurity was removed in the next step.

Rf = 0.3 (CHCl3/MeOH 10:1)

1H-NMR (500 MHz, MeOD): δ = 8.88 (s, 0.6H), 8.62 (s, 0.4H), 8.57 (s, 0.6H), 8.33 (s, 0.4H),

4.62 (s, 1H), 4.39 (dd, J = 20.3, 6.2 Hz, 1H), 4.17 (s, 1H), 3.72 – 3.66 (m, 1H), 3.06 (dd, J =

13.4, 7.0 Hz, 1H), 3.01 – 2.91 (m, 1H), 2.76 – 2.62 (m, 3H), 2.32 – 2.11 (m, 3H), 2.08 – 1.93

(m, 2H), 1.83 (dt, J = 21.5, 4.9 Hz, 1H), 1.59 (s, 5H), 1.50 (dd, J = 13.5, 7.1 Hz, 1H), 1.47 –

1.42 (m, 11H), 1.40 (s, 11H), 0.93 (s, 6H), 0.93 (s, 3H), 0.13 (s, 1H), 0.12 (s, 2H), 0.11 (s,

1H), 0.10 (s, 2H).

13C-NMR (400 MHz, MeOD): δ = 180.5, 151.6, 150.0, 131.8, 128.8, 83.2, 72.4, 60.7, 37.8,

36.3, 30.8, 28.9, 28.2, 27.8, 27.3, 25.6, 23.1, 22.4, 17.5, 13.8, 10.7, -4.6, -5.0.

HR-MS (ESI): calc. for C37H61N6O9SSi [M+H]+ : 793.3985, found: 793.3985.

Experimental Section

214

287a

(S)-methyl 4-((((1S,2S,3S,5S)-5-(6-amino-9H-purin-9- yl)-3-((tert-

butyldimethylsilyl)oxy)bicyclo[3.1.0]hexan-2-yl)met hyl)thio)-2-((tert-

butoxycarbonyl)amino)butanoate

NaOMe (1.5 mg) was added to a solution of 286 (3 mg) in MeOH and stirred for 1 h, then

170 was added and the reaction mixture was refluxed (75°C) for 18 h. 0.2 ml of aq. NaHCO 3-

sol. was added and the mixture was concentrated under reduced pressure. Purification by

flash column chromatography (EtOAc/Hex 5:1 � EtOAc/MeOH 20:1) gave 3 mg (60%) of the

title compound.

Rf = 0.48 (EtOAc/MeOH 20:1)

1H-NMR (400 MHz, CDCl3): δ = 8.77 (s, 1H), 8.05 (s,1H), 4.23 (d, J = 6.0 Hz, 1H), 3.74 (s,

3H), 3.53 – 3.49 (m, 1H), 3.40-3.36 (m, 1H), 2.93 – 2.88 (m, 1H), 2.62 – 2.59 (m, 1H), 2.58-

2.55 (m, 1H), 2.21 – 2.13 (m, 2H), 2.12 – 2.07 (m, 1H), 1.96-1.89 (m, 2H), 1.86-1.82 (m, 1H),

1.78 – 1.73 (m, 1H), 1.41 (s, 9H), 1.34-1.26 (m, 1H), 0.84 (s, 9H), 0.02 (s, 3H), 0.01 (S, 3H).

MS (ESI): calcd for C28H47N6O5SSi [M-H]+: 607.31, found: 607.40 (I = 100%).

173a

(S)-4-((((1S,2S,3S,5S)-5-(6-amino-9H-purin-9-yl)-3 -((tert-

butyldimethylsilyl)oxy)bicyclo[3.1.0]hexan-2-yl)met hyl)thio)-2-((tert-

butoxycarbonyl)amino)butanoic acid

287a (7 mg) was dissolved in THF (0.4 ml) and cooled to 0°C, then KOH-sol. (23 ul, 1M aq.

sol.) and water (50 ul) were added and the reaction mixture was stirred for 14 h. The reaction

mixture was cooled to 0°C and quenched by adding HC l (23 ul, 1M aq. sol.). The solvents

were evaporated and the crude product was purified by flash column chromatography

Experimental Section

215

(CHCl3/MeOH/H2O/acetic acid 8:1:0.1%:0.1%) to afford 3.5 mg (51%) of the title compound

as a white solid.

Rf = 0.2 (CHCl3/MeOH 10:1)

1H-NMR (500 MHz, MeOD): δ = 8.21 (s, 1H), 8.14 (s, 1H), 4.62 (s, 1H), 4.63-4.51 (m, 2H),

4.35 (d, J = 6.4 Hz, 1H), 4.09 (s, 1H), 3.03-2.96 (m, 1H), 2.93-2.86 (m, 1H), 2.68-2.59 (m,

3H), 2.26 – 2.21 (m, 1H), 2.19-2.12 (m, 2H), 1.78 (t, J = 5.1 Hz, 1H), 1.63-1.51 (m, 2H), 1.43

(s, 9H), 1.38-1.35 (m, 1H), 1.28 (s, 9H), 0.92 (s, 12H), 0.11 (s, 3H), 0.09 (s, 3H).

MS (ESI): calc. for C27H45N6O5SSi [M-H]+ : 593.29, found: 593.20 (I = 100%).

173b

(S)-2-amino-4-((((1S,2S,3S,5S)-5-(6-amino-9H-purin- 9-yl)-3-((tert-

butyldimethylsilyl)oxy)bicyclo[3.1.0]hexan-2-yl)met hyl)thio)butanoic acid

TBS-protected acid 173a (3.5 mg) was dissolved in EtOAc (0.2 ml), HCl (360 ul, 1M- sol. in

EtOAc) was added and the reaction mixture was stirred for 3 d at 40°C. A white precipitate

formed. The solvent was evaporated and the crude product was dried under high vacuum

conditions to give 4 mg (>100%) of a white solid.

Rf = 0.45 (acetic acid/BuOH/H2O 1:3:1, ninhydrin)

MS (ESI): calc. for C22H37N6O3SSi [M+H]+ : 493.24, found: 493.10 (I = 100%).

Note: Compound 7 also formed but the deprotection reaction did not go to completion with

the reagent used. TLC analysis indicated a 1:1 mixture of 173b and 7.

7

(S)-2-amino-4-((((1S,2S,3S,5S)-5-(6-amino-9H-purin- 9-yl)-3-hydroxybicyclo[3.1.0]hexan-

2-yl)methyl)thio)butanoic acid

Fully protected acid 173 (5 mg) was dissolved in CH2Cl2 (0.5 ml), TFA (200 ul) was added

and RM stirred for 24 h at RT. After this time, TLC and MS indicated that all Boc-groups had

Experimental Section

216

been removed but that the TBS group was still on. H2O (20 ul) was added and the reaction

mixture was stirred at RT for additional 18 h. TLC and MS indicated end of reaction. The

solvents and the TFA were evaporated under high vacuum conditions. The residue was

dissolved in water and extracted with EtOAc. The water phase was lyophylized to yield 7 mg

(>100%) of the crude product as a TFA salt in the form of a colorless solid.

HPLC analysis indicated the presence of two compounds with retention times 6.3 min and 8

min; they were separated by semi-preparative HPLC using an Agilent Zorbax SB-Phenyl

column (5 µm, 9.4 x 150 mm) using CH3CN with 0.025 % TFA and water with 0.03% TFA as

eluents. Subsequent analytical HPLC (Agilent Zorbax SB-Phenyl column, 3.5 µm, 4.6 x 150

mm) displayed the same two peaks (after 6.3 min and 8 min). Since the NMR data were

identical, we believe that compound 7 exists in a pH-dependent equilibrium of two forms.

Alternatively, 7 was prepared from partially deprotected acid 173b. 173b (4 mg) was

dissolved in CH2CH2 (0.5 ml), TFA (50 ul) was added and the reaction mixture was stirred for

12 h at RT. Additional 50 ul of TFA and 10 ul of H2O were added and the reaction mixture

was stirred for additional 8 h. The solvents were evaporated and the crude product was dried

under high vacuum conditions to yield 5 mg of a slightly green solid that was dissolved in

water (0.5 ml) and extracted with EtOAc (0.3 ml). The organic phase was removed and the

water phase was lyophilizised to afford 4.2 mg (>100%) of the crude product as a TFA salt in

the form of a colorless solid. This batch was also subjected to purification by semi-

preparative HPLC using the same conditions as above. Both batches combined yielded 2.8

mg of 7 after purification.

Rf = 0.3 (acetic acid/BuOH/H2O 1:3:1, ninhydrin)

1H NMR (500 MHz, D2O): δ = 8.44 (s, 1H), 8.40 (s, 1H), 4.41 (dd, J = 6.9, 1.0 Hz, 1H), 3.96

(dt, J = 6.7, 3.4 Hz, 1H), 3.04 (dd, J = 13.1, 7.3 Hz, 1H), 2.95 (dd, J = 13.1, 7.8 Hz, 1H), 2.81

(t, J = 7.6 Hz, 2H), 2.65 (ddd, J = 14.4, 7.0, 2.1 Hz, 1H), 2.39 (d, J = 14.2 Hz, 1H), 2.29 (t, J =

7.4 Hz, 2H), 2.25 – 2.14 (m, 2H), 1.67 – 1.61 (m, 1H), 1.54 (ddd, J = 9.7, 5.9, 2.1 Hz, 1H).

13C-NMR (500 MHz, D2O): δ = 173.8, 150.4, 149.5, 145.3, 144.6, 118.6, 75.9, 53.8, 48.8,

42.9, 40.2, 34.3, 30.5, 28.1, 26.9, 17.9.

HR-MS (ESI): calcd for C16H23N6O3S [M-H]+: 379.1547, found: 379.1554.

Biological Evaluation of S-Adenosylhomocysteine Ana logue 7:

SAH Analogue 7 has been tested for activity on dengue N-7 and O-2’ methyltransferases by

Dr. Sebastian Sonntag. Protocols as described in 173 and 172 were used, with the following

modifications in RNA substrate and buffer conditions:

Experimental Section

217

Buffer conditions: the O-2’ assay buffer was 50 mM Tris (pH 7.5), 10 mM KCl, 2mM MgCl2,

0.05% CHAPS; the N-7 assay buffer was 50 mM Tris (pH 7.5), 75 mM NaCl, 0.01% TritonX-

100.

RNA substrates: For the N-7 assay, the biotinylated substrate was 110 nucleotides in length,

and used at a final concentration of 240 nM. For the O-2’ assay, the biotinylated RNA was an

8mer, and was used at a final concentration of 40 nM.

Curiculum Vitae

218

6 Curriculum Vitae

PERSONAL DETAILS

Name Anna Schlegel

Date of Birth 9.11.1977

Place of Birth Arbon, Switzerland

Nationality Swiss

EXPERIENCE

05.2005 – present PhD student in the group of Prof. Dr. Karl-Heinz Al tmann,

Institut of Pharmaceutical Sciences, Departement of Chemistry and Applied Biosiences (D-CHAB), ETH Zurich

• „Synthesis of Chemical Libraries based on a Bicyclo[3.1.0]hexane Scaffold and Studies on the Synthesis of Bicyclo[3.1.0]hexane- based S-Adenosylhomocysteine and Pentostatin Analogues“

02.2010 – 11.2010 Teacher of Mathematics and Physics

at “Katholisches Gymnasium Zürich“

• teaching of physics and mathematics

01.2005 - 05.2005 Associate Researcher, Flisom AG, Technopark Zurich

• development of thin film solar cells

06.2004 - 10.2004 Trainee, section research and development, medicinal chemistry,

F. Hoffmann - La Roche AG

• up-scaling and optimisation of organic syntheses

07.2002 - 09.2002 Trainee at University of Manchester

• design and construction of a detector of molecular negative ions

10.2001 – 06.2003 Junior Assistant at the Department of Mathematics, ETH Zurich

• teaching of Analysis I und II

Curiculum Vitae

219

EDUCATION

01.2004 – 04.2004 Diploma thesis in the group of Prof. Dr. Francois D iederich,

Institute of Organic Chemistry, Departement of Chem istry and Applied Biosiences (D-CHAB), ETH Zurich

„Long-arm Cavitands via Sonogashira Couplings: Synthesis and NMR Studies”

1998 - 2004 Master of Science ETH

in Interdisciplinary Sciences, ETH Zurich

with majors

organic chemistry, physical chemistry

quantum electronics, particle physics

1992 - 1998 Grammar School in Romanshorn

PUBLICATIONS

Vladimir A.Azov, Anna Schlegel, Francois Diederich

Functionalized calix[4]resorcinarene cavitands. Ver satile platforms for the modular

construction of extended molecular switches

Bulletin of the Chemical Society of Japan 2006, 79(12), 1926-1940

Vladimir A.Azov, Anna Schlegel, Francois Diederich

Geometrically precisely defined multinanometer expa nsion/contraction motions in a

resorcin[4]arene cavitand based molecular switch

Angewandte Chemie Int. Ed. 2005, 44(29), 4635-4638

Bibliography

220

7 Bibliography

[1] K. H. Bleicher, H.-J. Bohm, K. Muller, A. I. Alanine, Nat Rev Drug Discov 2003, 2, 369-378.

[2] G. M. Keseru, G. M. Makara, Drug Discovery Today 2006, 11, 741-748.

[3] D. C. Rees, M. Congreve, C. W. Murray, R. Carr, Nat Rev Drug Discov 2004, 3, 660-672.

[4] S. Ghosh, A. Nie, J. An, Z. Huang, Current Opinion in Chemical Biology 2006, 10, 194-202.

[5] M. R. Reddy, L. Parrill Abby, in Rational Drug Design, Vol. 719, American Chemical Society,

1999, pp. 1-11.

[6] R. Macarron, M. N. Banks, D. Bojanic, D. J. Burns, D. A. Cirovic, T. Garyantes, D. V. S. Green, R.

P. Hertzberg, W. P. Janzen, J. W. Paslay, U. Schopfer, G. S. Sittampalam, Nat Rev Drug Discov

2011, 10, 188-195.

[7] O. Roche, P. Schneider, J. Zuegge, W. Guba, M. Kansy, A. Alanine, K. Bleicher, F. Danel, E.-M.

Gutknecht, M. Rogers-Evans, W. Neidhart, H. Stalder, M. Dillon, E. Sjögren, N. Fotouhi, P.

Gillespie, R. Goodnow, W. Harris, P. Jones, M. Taniguchi, S. Tsujii, W. von der Saal, G.

Zimmermann, G. Schneider, Journal of Medicinal Chemistry 2001, 45, 137-142.

[8] T. W. J. Cooper, I. B. Campbell, S. J. F. Macdonald, Angewandte Chemie International Edition

2010, 49, 8082-8091.

[9] aK. M. Hoeld, N. S. Sirisoma, J. E. Casida, Chem. Res. Toxicol. 2001, 14, 589-595; bT. Deiml,

2003.

[10] P.-L. Fang, Y.-L. Cao, H. Yan, L.-L. Pan, S.-C. Liu, N.-B. Gong, Y. Lü, C.-X. Chen, H.-M. Zhong, Y.

Guo, H.-Y. Liu, Journal of Natural Products 2011, 74, 1408-1413.

[11] S.-P. Yang, Z.-B. Gao, Y. Wu, G.-Y. Hu, J.-M. Yue, Tetrahedron 2008, 64, 2027-2034.

[12] J. Kawabata, Y. Fukushi, S. Tahara, J. Mizutani, Phytochemistry 1990, 29, 2332-2334.

[13] X. S. Peng, Y. S. Lu, Org Lett 2011, 13, 2940-2943.

[14] A. F. Cameron, K. K. Cheung, G. Ferguson, Robertso.Jm, J Chem Soc B 1969, 559-&.

[15] W. Haas, H. Sterk, M. Mittelbach, J. Nat. Prod. 2002, 65, 1434-1440.

[16] W. Haas, H. Sterk, M. Mittelbach, Journal of Natural Products 2002, 65, 1434-1440.

[17] aD. A. D. Eric V. Anslyn, p. 101; bK. S. Pitzer, W. E. Donath, Journal of the American Chemical

Society 1959, 81, 3213-3218.

[18] R. L. Cook, T. Malloy, Journal of the American Chemical Society 1974, 96, 1703-1707.

[19] V. S. Mastryukov, E. L. Osina, L. V. Vilkov, R. L. Hilderbrandt, Journal of the American Chemical

Society 1977, 99, 6855-6861.

[20] M. F. Grostic, D. J. Duchamp, C. G. Chidester, The Journal of Organic Chemistry 1971, 36,

2929-2932.

[21] P. K. Freeman, M. F. Grostic, F. A. Raymond, The Journal of Organic Chemistry 1965, 30, 771-

777.

[22] S. Winstein, E. C. Friedrich, R. Baker, Y.-I. Lin, Tetrahedron 1966, 22, 621-645.

[23] R. J. Abraham, C. M. Holden, P. Loftus, D. Whittaker, Organic Magnetic Resonance 1974, 6,

184-189.

[24] K. Siam, J. D. Ewbank, L. Schäfer, C. V. Alsenoy, Journal of Molecular Structure: THEOCHEM

1987, 150, 121-128.

[25] P. Aped, N. L. Allinger, Journal of the American Chemical Society 1992, 114, 1-16.

[26] R. Okazaki, J. Niwa, S. Kato, B Chem Soc Jpn 1988, 61, 1619-1624.

[27] aL. L. Koh, H. H. Huang, K. Y. Sim, Acta Crystallographica Section C 1992, 48, 1641-1645; bR.

W. W. Hooft, J. Kroon, Acta Crystallographica Section C 1995, 51, 721-723.

[28] aV. G. Puranik, Acta Crystallogr., Sect. C: Cryst. Struct. Commun. 1988, C44, 163-164; bD. G.

Morris, P. Murray-Rust, J. Murray-Rust, J. Chem. Soc., Perkin Trans. 2 1977, 1577-1580.

[29] C. Altona, Sundaral.M, Journal of the American Chemical Society 1972, 94, 8205-&.

Bibliography

221

[30] V. E. Marquez, H. R. Moon, M. A. Siddiqui, G. Sun, I. V. Filippov, N. A. Landsman, Y. C. Lee, K.

M. Adams, J. J. Barchi, J. R. Deschamps, M. C. Nicklaus, J. A. Kelley, Tetrahedron 2010, 66,

6707-6717.

[31] J. Plavec, W. Tong, J. Chattopadhyaya, Journal of the American Chemical Society 1993, 115,

9734-9746.

[32] V. E. Marquez, M. A. Siddiqui, A. Ezzitouni, P. Russ, J. Y. Wang, R. W. Wagner, M. D.

Matteucci, Journal of Medicinal Chemistry 1996, 39, 3739-3747.

[33] aV. E. Marquez, A. Ezzitouni, P. Russ, M. A. Siddiqui, H. Ford, R. J. Feldman, H. Mitsuya, C.

George, J. J. Barchi, Journal of the American Chemical Society 1998, 120, 2780-2789; bV. E.

Marquez, P. Russ, R. Alonso, M. A. Siddiqui, K. J. Shin, C. George, M. C. Nicklaus, F. Dai, H.

Ford, Nucleosides Nucleotides & Nucleic Acids 1999, 18, 521-530.

[34] V. E. Marquez, A. Ezzitouni, M. A. Siddiqui, P. Russ, H. Ikeda, C. George, Nucleosides

Nucleotides 1997, 16, 1431-1434.

[35] V. E. Marquez, Abstr Pap Am Chem S 2008, 236.

[36] E. De Clercq, Current Opinion in Pharmacology 2010, 10, 507-515.

[37] P. A. Furman, J. A. Fyfe, M. H. St Clair, K. Weinhold, J. L. Rideout, G. A. Freeman, S. N.

Lehrman, D. P. Bolognesi, S. Broder, H. Mitsuya, Proceedings of the National Academy of

Sciences 1986, 83, 8333-8337.

[38] G. R. Painter, A. E. Aulabaugh, C. W. Andrews, Biochemical and Biophysical Research

Communications 1993, 191, 1166-1171.

[39] Y. Choi, C. George, M. J. Comin, J. J. Barchi, H. S. Kim, K. A. Jacobson, J. Balzarini, H. Mitsuya,

P. L. Boyer, S. H. Hughes, V. E. Marquez, Journal of Medicinal Chemistry 2003, 46, 3292-3299.

[40] aJ. Balzarini, P. Herdewijn, E. Declercq, J Biol Chem 1989, 264, 6127-6133; bW. Y. Gao, R.

Agbaria, J. S. Driscoll, H. Mitsuya, J Biol Chem 1994, 269, 12633-12638.

[41] V. E. Marquez, S. H. Hughes, S. Sei, R. Agbaria, Antiviral Research 2006, 71, 268-275.

[42] M. J. Comin, B. C. Vu, P. L. Boyer, C. Liao, S. H. Hughes, V. E. Marquez, Chemmedchem 2008,

3, 1129-1134.

[43] V. E. Marquez, Y. Choi, M. J. Comin, P. Russ, C. George, M. Huleihel, T. Ben-Kasus, R. Agbaria,

Journal of the American Chemical Society 2005, 127, 15145-15150.

[44] aK. H. Altmann, R. Kesselring, E. Francotte, G. Rihs, Tetrahedron Lett 1994, 35, 2331-2334;

bV. E. Marquez, T. Ben-Kasus, J. J. Barchi, Jr., K. M. Green, M. C. Nicklaus, R. Agbaria, Journal

of the American Chemical Society 2004, 126, 543-549.

[45] W. Saenger, Springer, 1984, pp. 51-104.

[46] V. E. Marquez, Y. Choi, M. J. Comin, P. Russ, C. George, M. Huleihel, T. Ben-Kasus, R. Agbaria,

Journal of the American Chemical Society 2005, 127, 15145-15150.

[47] M. J. Comin, R. Agbaria, T. Ben-Kasus, M. Huleihel, C. Liao, G. Sun, M. C. Nicklaus, J. R.

Deschamps, D. A. Parrish, V. E. Marquez, Journal of the American Chemical Society 2007, 129,

6216-6222.

[48] aK. M. Reilly, D. F. Kisor, Onco Targets Ther 2009, 2, 219-228; bM. S. Czuczman, P. Porcu, J.

Johnson, D. Niedzwiecki, M. Kelly, E. D. Hsi, J. R. Cook, G. Canellos, B. D. Cheson, Leuk

Lymphoma 2007, 48, 97-103; cM. S. Hershfield, Eur J Immunol 2005, 35, 25-30.

[49] G. Hasko, B. N. Cronstein, Trends Immunol 2004, 25, 33-39.

[50] R. Franco, R. Pacheco, J. M. Gatell, T. Gallart, C. Lluis, Crit Rev Immunol 2007, 27, 495-509.

[51] M. Asakura, H. Asanuma, J. Kim, Y. Liao, K. Nakamaru, M. Fujita, K. Komamura, T. Isomura, H.

Furukawa, H. Tomoike, M. Kitakaze, Hypertens Res 2007, 30, 781-787.

[52] R. H. Adamson, D. W. Zaharevitz, D. G. Johns, Pharmacology 1977, 15, 84-89.

[53] R. P. Agarwal, T. Spector, R. E. Parks, Jr., Biochem Pharmacol 1977, 26, 359-367.

[54] aT. Robak, E. Lech-Maranda, A. Korycka, E. Robak, Curr Med Chem 2006, 13, 3165-3189; bP.

P. Major, R. P. Agarwal, D. W. Kufe, Blood 1981, 58, 91-96.

[55] R. S. Hosmane, Research Signpost, 2002, pp. 133-151.

[56] Z. Wang, F. A. Quiocho, Biochemistry 1998, 37, 8314-8324.

Bibliography

222

[57] E. T. Larson, W. Deng, B. E. Krumm, A. Napuli, N. Mueller, W. C. Van Voorhis, F. S. Buckner, E.

Fan, A. Lauricella, G. DeTitta, J. Luft, F. Zucker, W. G. J. Hol, C. L. M. J. Verlinde, E. A. Merritt,

Journal of Molecular Biology 2008, 381, 975-988.

[58] L.-M. Ting, W. Shi, A. Lewandowicz, V. Singh, A. Mwakingwe, M. R. Birck, E. A. T. Ringia, G.

Bench, D. C. Madrid, P. C. Tyler, G. B. Evans, R. H. Furneaux, V. L. Schramm, K. Kim, J Biol

Chem 2005, 280, 9547-9554.

[59] K. Kim, L. M. Ting, W. X. Shi, A. Lewandowicz, V. Singh, A. Mwakingwe, M. R. Birck, E. A. T.

Ringia, G. Bench, D. C. Madrid, P. C. Tyler, G. B. Evans, R. H. Furneaux, V. L. Schramm, J Biol

Chem 2005, 280, 9547-9554.

[60] P. C. Tyler, E. A. Taylor, R. F. G. Fröhlich, V. L. Schramm, Journal of the American Chemical

Society 2007, 129, 6872-6879.

[61] T. Cacciamani, A. Vita, G. Cristalli, S. Vincenzetti, P. Natalini, S. Ruggieri, A. Amici, G. Magni,

Arch Biochem Biophys 1991, 290, 285-292.

[62] M. Lemaire, L. F. Momparler, M. L. Bernstein, V. E. Marquez, R. L. Momparler, Anticancer

Drugs 2005, 16, 301-308.

[63] O. R. Ludek, G. K. Schroeder, C. Liao, P. L. Russ, R. Wolfenden, V. E. Marquez, Journal of

Organic Chemistry 2009, 74, 6212-6223.

[64] aS. Xiang, S. A. Short, R. Wolfenden, C. W. Carter, Biochemistry 1995, 34, 4516-4523; bA.-H.

Teh, M. Kimura, M. Yamamoto, N. Tanaka, I. Yamaguchi, T. Kumasaka, Biochemistry 2006, 45,

7825-7833; cS. J. Chung, J. C. Fromme, G. L. Verdine, Journal of Medicinal Chemistry 2005, 48,

658-660.

[65] F. Lyko, R. Brown, Journal of the National Cancer Institute 2005, 97, 1498-1506.

[66] P. A. Jones, S. B. Baylin, Nat Rev Genet 2002, 3, 415-428.

[67] S. P. Lim, L. S. Sonntag, C. Noble, S. H. Nilar, R. H. Ng, G. Zou, P. Monaghan, K. Y. Chung, H.

Dong, B. Liu, C. Bodenreider, G. Lee, M. Ding, W. L. Chan, G. Wang, Y. L. Jian, A. T. Chao, J.

Lescar, Z. Yin, T. R. Vedananda, T. H. Keller, P.-Y. Shi, J. Biol. Chem. 2011, 286, 6233-6240.

[68] R. J. Roberts, X. Cheng, Annu Rev Biochem 1998, 67, 181-198.

[69] P. Wang, A. S. Brank, N. K. Banavali, M. C. Nicklaus, V. E. Marquez, J. K. Christman, A. D.

MacKerell, Journal of the American Chemical Society 2000, 122, 12422-12434.

[70] S. A. Poulsen, R. J. Quinn, Bioorg Med Chem 1998, 6, 619-641.

[71] C. E. Muller, Curr Med Chem 2000, 7, 1269-1288.

[72] S. Tchilibon, B. V. Joshi, S. K. Kim, H. T. Duong, Z. G. Gao, K. A. Jacobson, Journal of Medicinal

Chemistry 2005, 48, 1745-1758.

[73] K. A. Jacobson, X. D. Ji, A. H. Li, N. Melman, M. A. Siddiqui, K. J. Shin, V. E. Marquez, R. G. Ravi,

Journal of Medicinal Chemistry 2000, 43, 2196-2203.

[74] C. S. Carr, R. J. Hill, H. Masamune, S. P. Kennedy, D. R. Knight, W. R. Tracey, D. M. Yellon,

Cardiovascular Research 1997, 36, 52-59.

[75] K. A. Jacobson, Z. G. Gao, S. Tchilibon, H. T. Duong, B. V. Joshi, D. Sonin, B. T. Liang, Journal of

Medicinal Chemistry 2005, 48, 8103-8107.

[76] M. V. Sitkovsky, A. Ohta, Trends Immunol 2005, 26, 299-304.

[77] K. A. Jacobson, J.-M. Boeynaems, Drug Discovery Today 2010, 15, 570-578.

[78] H. S. Kim, M. Ohno, B. Xu, H. O. Kim, Y. Choi, X. D. Ji, S. Maddileti, V. E. Marquez, T. K. Harden,

K. A. Jacobson, Journal of Medicinal Chemistry 2003, 46, 4974-4987.

[79] D. M. Bourdon, S. K. Mahanty, K. A. Jacobson, J. L. Boyer, T. K. Harden, J Thromb Haemost

2006, 4, 861-868.

[80] K. A. Jacobson, S. Costanzi, A. A. Ivanov, S. Tchilibon, P. Besada, Z. G. Gao, S. Maddileti, T. K.

Harden, Biochem Pharmacol 2006, 71, 540-549.

[81] H. Ko, I. Fricks, A. A. Ivanov, T. K. Harden, K. A. Jacobson, Journal of Medicinal Chemistry

2007, 50, 2030-2039.

[82] K. A. Jacobson, S. Costanzi, B. V. Joshi, P. Besada, D. H. Shin, H. Ko, A. A. Ivanov, L.

Mamedova, Agonists and Antagonists for P2 Receptors, John Wiley & Sons, Ltd, 2008.

Bibliography

223

[83] S. Costanzi, B. V. Joshi, S. Maddileti, L. Mamedova, M. J. Gonzalez-Moa, V. E. Marquez, T. K.

Harden, K. A. Jacobson, Journal of Medicinal Chemistry 2005, 48, 8108-8111.

[84] Y. Liu, F.-J. Nan, Tetrahedron Lett 2010, 51, 1374-1376.

[85] Y.-S. Lu, X.-S. Peng, Org Lett 2011, 13, 2940-2943.

[86] J. Li, T. L. Lowary, Org Lett 2008, 10, 881-884.

[87] aS. Berg, D. Kaur, M. Jackson, P. J. Brennan, Glycobiology 2007, 17, 35-56R; bM. Belanova, P.

Dianiskova, P. J. Brennan, G. C. Completo, N. L. Rose, T. L. Lowary, K. Mikusova, J Bacteriol

2008, 190, 1141-1145.

[88] A. Srikrishna, P. Praveen Kumar, Tetrahedron 2000, 56, 8189-8195.

[89] O. R. Ludek, V. E. Marquez, Synthesis 2007, 2007, 3451,3460.

[90] V. E. Marquez, M. I. Lim, C. K. H. Tseng, A. Markovac, M. A. Priest, M. S. Khan, B. Kaskar, The

Journal of Organic Chemistry 1988, 53, 5709-5714.

[91] J. A. Staroscik, B. Rickborn, J. Org. Chem. 1972, 37, 738-740.

[92] L. S. Jeong, V. E. Marquez, C. S. Yuan, R. T. Borchardt, Heterocycles 1995, 41, 2651-2656.

[93] M. A. Siddiqui, H. Ford, C. George, V. E. Marquez, Nucleosides & Nucleotides 1996, 15, 235-

250.

[94] A. Ezzitouni, P. Russ, V. E. Marquez, The Journal of Organic Chemistry 1997, 62, 4870-4873.

[95] aK. Biggadike, A. D. Borthwick, D. Evans, A. M. Exall, B. E. Kirk, S. M. Roberts, L. Stephenson,

P. Youds, Journal of the Chemical Society, Perkin Transactions 1 1988, 549-554; bO. R. Ludek,

C. Meier, Nucleosides Nucleotides Nucleic Acids 2003, 22, 683-685.

[96] N. Kondo, H. Fueno, H. Fujimoto, M. Makino, H. Nakaoka, I. Aoki, S. Uemura, Journal of

Organic Chemistry 1994, 59, 5254-5263.

[97] Y. Choi, H. R. Moon, Y. Yoshimura, V. E. Marquez, Nucleosides, Nucleotides and Nucleic Acids

2003, 22, 547 - 557.

[98] N. Cohen, Organic Synthesis 1985, 63.

[99] B. V. Joshi, A. Melman, R. L. Mackman, K. A. Jacobson, Nucleosides Nucleotides Nucleic Acids

2008, 27, 279-291.

[100] L. A. Paquette, S. Bailey, The Journal of Organic Chemistry 1995, 60, 7849-7856.

[101] Y.-X. Yang, Z. Li, H.-J. Feng, G.-R. Chen, Y.-C. Li, Tetrahedron Lett 2010, 51, 3848-3851.

[102] S. Takano, A. Kurotaki, M. Takahashi, K. Ogasawara, Synthesis 1986, 1986, 403,406.

[103] K.-H. Altmann, R. Imwinkelried, R. Kesselring, G. Rihs, Tetrahedron Lett 1994, 35, 7625-7628.

[104] A. Ezzitouni, V. E. Marquez, Journal of the Chemical Society, Perkin Transactions 1 1997,

1073-1078.

[105] H. Saneyoshi, J. R. Deschamps, V. E. Marquez, J. Org. Chem. 2010, 75, 7659-7669.

[106] S.-H. Kim, M. J. Sung, K. C. Jin, Org. Synth. 2003, 80, No pp. given.

[107] T. H. Keller, A. Pichota, Z. Yin, Current Opinion in Chemical Biology 2006, 10, 357-361.

[108] P. Das, C. P. Spears, A. H. Shahinian, S. K. Dasgupta, N. G. Kundu, Bioorganic & Medicinal

Chemistry Letters 1996, 6, 2477-2480.

[109] aZ. Jin, Natural Product Reports 2009, 26, 382-445; bM. V. N. de Souza, Journal of Sulfur

Chemistry 2005, 26, 429-449.

[110] H.-J. Gais, H. J. Lindner, T. Lied, K. L. Lukas, W. A. Ball, B. Rosenstock, H. Sliwa, Liebigs Annalen

der Chemie 1986, 1986, 1179-1212.

[111] T. Van Truong, H. Rapoport, The Journal of Organic Chemistry 1993, 58, 6090-6096.

[112] R. T. Borchardt, J. A. Huber, Y. S. Wu, Journal of Medicinal Chemistry 1976, 19, 1094-1099.

[113] R. T. Borchardt, Journal of Medicinal Chemistry 1980, 23, 347-357.

[114] M. Milani, E. Mastrangelo, M. Bollati, B. Selisko, E. Decroly, M. Bouvet, B. Canard, M.

Bolognesi, Antiviral Research 2009, 83, 28-34.

[115] R. T. Borchardt, Y. S. Wu, Journal of Medicinal Chemistry 1975, 18, 300-304.

[116] A. Ezzitouni, V. E. Marquez, Journal of the Chemical Society-Perkin Transactions 1 1997, 1073-

1078.

Bibliography

224

[117] aH. J. Gais, H. J. Lindner, T. Lied, K. L. Lukas, W. A. Ball, B. Rosenstock, H. Sliwa, Liebigs

Annalen Der Chemie 1986, 1179-1212; bH. J. Gais, K. L. Lukas, W. A. Ball, S. Braun, H. J.

Lindner, Liebigs Annalen Der Chemie 1986, 687-716.

[118] T. P. Mawhinney, M. A. Madson, The Journal of Organic Chemistry 1982, 47, 3336-3339.

[119] S. Dong, L. A. Paquette, The Journal of Organic Chemistry 2006, 71, 1647-1652.

[120] aY. Mori, K. Yaegashi, H. Furukawa, The Journal of Organic Chemistry 1998, 63, 6200-6209;

bA. Fürstner, C. Mathes, C. W. Lehmann, Chemistry – A European Journal 2001, 7, 5299-5317.

[121] T. M. Willson, P. Kocienski, K. Jarowicki, K. Isaac, P. M. Hitchcock, A. Faller, S. F. Campbell,

Tetrahedron 1990, 46, 1767-1782.

[122] K.-H. Altmann, R. Kesselring, U. Pieles, Tetrahedron 1996, 52, 12699-12722.

[123] Y. Li, Synlett 2010, 2010, 837,838.

[124] aM. Takebayashi, Kagakushi Kenkyu (Narashino, Jpn.) 1989, 16, 149-153; bA. Bertho, Journal

für Praktische Chemie 1928, 120, 89-118; cT. Curtius, Journal für Praktische Chemie 1916, 94,

273-299.

[125] J. H. Saunders, R. J. Slocombe, Chemical Reviews 1948, 43, 203-218.

[126] T. Shioiri, K. Ninomiya, S. Yamada, Journal of the American Chemical Society 1972, 94, 6203-

6205.

[127] S. Linke, G. T. Tisue, W. Lwowski, Journal of the American Chemical Society 1967, 89, 6308-

6310.

[128] A. Speicher, T. Klaus, T. Eicher, Journal für Praktische Chemie/Chemiker-Zeitung 1998, 340,

581-583.

[129] T. Holletz, D. Cech, Synthesis 1994, 1994, 789,791.

[130] E. J. Thomas, J. W. F. Whitehead, Journal of the Chemical Society, Perkin Transactions 1 1989,

507-518.

[131] aP. B. Alper, H. Liu, A. K. Chatterjee, K. T. Nguyen, D. C. Tully, C. Tumanut, J. Li, J. L. Harris, T.

Tuntland, J. Chang, P. Gordon, T. Hollenbeck, D. S. Karanewsky, Bioorganic & Medicinal

Chemistry Letters 2006, 16, 1486-1490; bD. C. Tully, H. Liu, A. K. Chatterjee, P. B. Alper, R.

Epple, J. A. Williams, M. J. Roberts, D. H. Woodmansee, B. T. Masick, C. Tumanut, J. Li, G.

Spraggon, M. Hornsby, J. Chang, T. Tuntland, T. Hollenbeck, P. Gordon, J. L. Harris, D. S.

Karanewsky, Bioorganic & Medicinal Chemistry Letters 2006, 16, 5112-5117.

[132] A. Melman, M. Zhong, V. E. Marquez, K. A. Jacobson, The Journal of Organic Chemistry 2008,

73, 8085-8088.

[133] G. Shaw, R. N. Warrener, Journal of the Chemical Society (Resumed) 1958, 157-161.

[134] B. N. Yoo, H. O. Kim, H. R. Moon, S. K. Seol, S. K. Jang, K. M. Lee, L. S. Jeong, Bioorganic &

Medicinal Chemistry Letters 2006, 16, 4190-4194.

[135] D. J. P. Pinto, M. J. Orwat, S. Koch, K. A. Rossi, R. S. Alexander, A. Smallwood, P. C. Wong, A.

R. Rendina, J. M. Luettgen, R. M. Knabb, K. He, B. Xin, R. R. Wexler, P. Y. S. Lam, Journal of

Medicinal Chemistry 2007, 50, 5339-5356.

[136] P. A. Jacobi, H. G. Selnick, Journal of Organic Chemistry 1990, 55, 202-209.

[137] P. R. Krishna, V. V. R. Reddy, R. Srinivas, Tetrahedron 2007, 63, 9871-9880.

[138] G. Stupka, L. Gremaud, A. F. Williams, Helvetica Chimica Acta 2005, 88, 487-495.

[139] D. L. Evans, D. K. Minster, U. Jordis, S. M. Hecht, A. L. Mazzu, A. I. Meyers, The Journal of

Organic Chemistry 1979, 44, 497-501.

[140] S. Kreimerman, I. Ryu, S. Minakata, M. Komatsu, Comptes Rendus de l'Académie des Sciences

- Series IIC - Chemistry 2001, 4, 497-503.

[141] S. Crosignani, A. C. Young, B. Linclau, Tetrahedron Lett 2004, 45, 9611-9615.

[142] N. A. Braun, M. Ousmer, J. D. Bray, D. Bouchu, K. Peters, E.-M. Peters, M. A. Ciufolini, The

Journal of Organic Chemistry 2000, 65, 4397-4408.

[143] T. Aoyama, N. Sonoda, M. Yamauchi, K. Toriyama, M. Anzai, A. Ando, T. Shioiri, Synlett 1998,

1998, 35,36.

[144] C. Kashima, H. Arao, Synthesis 1989, 1989, 873,874.

Bibliography

225

[145] C. Wu, E. R. Decker, N. Blok, H. Bui, T. J. You, J. Wang, A. R. Bourgoyne, V. Knowles, K. L.

Berens, G. W. Holland, T. A. Brock, R. A. F. Dixon, Journal of Medicinal Chemistry 2004, 47,

1969-1986.

[146] Y. Kodama, M. Ori, T. Nishio, Helvetica Chimica Acta 2005, 88, 187-193.

[147] A. I. Meyers, D. L. Temple, Jr., J. Amer. Chem. Soc. 1970, 92, 6644-6646.

[148] D. Rai, M. Johar, T. Manning, B. Agrawal, D. Y. Kunimoto, R. Kumar, Journal of Medicinal

Chemistry 2005, 48, 7012-7017.

[149] C. F. Matta, A. A. Arabi, D. F. Weaver, European Journal of Medicinal Chemistry 2010, 45,

1868-1872.

[150] H. Mizufune, M. Nakamura, H. Mitsudera, Tetrahedron 2006, 62, 8539-8549.

[151] J. I. Concepcion, C. G. Francisco, R. Freire, R. Hernandez, J. A. Salazar, E. Suarez, J. Org. Chem.

1986, 51, 402-404.

[152] S. Kirkpatrick, C. D. Gelatt, M. P. Vecchi, Science 1983, 220, 671-680.

[153] V. Granville, M. Krivanek, J. P. Rasson, Pattern Analysis and Machine Intelligence, IEEE

Transactions on 1994, 16, 652-656.

[154] N. Metropolis, A. W. Rosenbluth, M. N. Rosenbluth, A. H. Teller, E. Teller, Equation of State

Calculations by Fast Computing Machines, Vol. 21, AIP, 1953.

[155] C. A. G. Haasnoot, L. F. A. A. M. De, C. Altona, Tetrahedron 1980, 36, 2783-2792.

[156] aH. Rapoport, Y. W. Chen, R. M. Mohareb, J. H. Ahn, T. B. Sim, J. Z. Ho, Chem Pharm Bull

2003, 51, 1153-1156; bT. Vantruong, H. Rapoport, Journal of Organic Chemistry 1993, 58,

6090-6096.

[157] P. Serafinowski, E. Dorland, K. R. Harrap, J. Balzarini, C. E. De, J. Med. Chem. 1992, 35, 4576-

4583.

[158] K. A. Houck, W. P. Bocchinfuso, M. S. Dowless, K. M. Borchert, Drug Discovery Ser. 2006, 5,

357-369,, 351 plate.

[159] M. Whitney, E. Rockenstein, G. Cantin, T. Knapp, G. Zlokarnik, P. Sanders, K. Durick, F. F.

Craig, P. A. Negulescu, Nat Biotech 1998, 16, 1329-1333.

[160] A. Citri, Y. Yarden, Nat Rev Mol Cell Biol 2006, 7, 505-516.

[161] A. B. Tsoupras, M. Chini, N. Tsogas, A. Lioni, G. Tsekes, C. A. Demopoulos, M. C. Lazanas, J.

Inflammation (London, U. K.) 2011, 8, 17.

[162] S. Ulisse, E. Baldini, S. Sorrenti, M. D'Armiento, Curr. Cancer Drug Targets 2009, 9, 32-71.

[163] Q. Xiao, S. Tang, X. Fan, Xiandai Zhongliu Yixue 2008, 16, 674-676.

[164] V. Hook, G. Hook, M. Kindy, Biol. Chem. 2010, 391, 861-872.

[165] S. Ludewig, M. Kossner, M. Schiller, K. Baumann, T. Schirmeister, Curr. Top. Med. Chem.

(Sharjah, United Arab Emirates) 2010, 10, 368-382.

[166] C. Sala, N. Dhar, R. C. Hartkoorn, M. Zhang, Y. H. Ha, P. Schneider, S. T. Cole, Antimicrob.

Agents Chemother. 2010, 54, 4150-4158.

[167] A. Martin, M. Camacho, F. Portaels, J. C. Palomino, Antimicrob. Agents Chemother. 2003, 47,

3616-3619.

[168] D. Rai, M. Johar, T. Manning, B. Agrawal, D. Y. Kunimoto, R. Kumar, J. Med. Chem. 2005, 48,

7012-7017.

[169] D. Rai, M. Johar, N. C. Srivastav, T. Manning, B. Agrawal, D. Y. Kunimoto, R. Kumar, J Med

Chem 2007, 50, 4766-4774.

[170] A. Chicca, F. Pellati, B. Adinolfi, A. Matthias, I. Massarelli, S. Benvenuti, E. Martinotti, A. M.

Bianucci, K. Bone, R. Lehmann, P. Nieri, Br J Pharmacol 2008, 153, 879-885.

[171] N. A. Englert, B. C. Spink, D. C. Spink, J. Steroid Biochem. Mol. Biol. 2011, 123, 140-150.

[172] S. P. Lim, L. S. Sonntag, C. Noble, S. H. Nilar, R. H. Ng, G. Zou, P. Monaghan, K. Y. Chung, H. P.

Dong, B. P. Liu, C. Bodenreider, G. Lee, M. Ding, W. L. Chan, G. Wang, Y. L. Jian, A. T. Chao, J.

Lescar, Z. Yin, T. R. Vedananda, T. H. Keller, P. Y. Shi, J Biol Chem 2011, 286, 6233-6240.

[173] S. P. Lim, D. Wen, T. L. Yap, C. K. Yan, J. Lescar, S. G. Vasudevan, Antiviral Res. 2008, 80, 360-

369.

[174] S. P. Lim, K. Y. Chung, H. P. Dong, A. T. Chao, P. Y. Shi, J. Lescar, Virology 2010, 402, 52-60.

Bibliography

226