study of the lewis acid catalyzed povarov reaction for the ... · 1 abstract during last years,...

78
Study of the Lewis Acid catalyzed Povarov reaction for the synthesis of polycyclic tetrahydroquinoline derivatives Riccardo Vallesi Thesis to obtain the Master of Science Degree in Chemistry Supervisors: Dr. Cristina Cimarelli Dr. Alexandra Antunes Examination Committee Chairman: Profª. Isabel M. Marrucho Supervisor: Dra. Alexandra Antunes Member of committee: Prof. João Paulo Cabral Telo December 2017

Upload: vucong

Post on 08-Feb-2019

218 views

Category:

Documents


0 download

TRANSCRIPT

Study of the Lewis Acid catalyzed Povarov reaction

for the synthesis of polycyclic tetrahydroquinoline derivatives

Riccardo Vallesi

Thesis to obtain the Master of Science

Degree in

Chemistry

Supervisors: Dr. Cristina Cimarelli

Dr. Alexandra Antunes

Examination Committee

Chairman: Profª. Isabel M. Marrucho

Supervisor: Dra. Alexandra Antunes

Member of committee: Prof. João Paulo Cabral Telo

December 2017

1

Abstract

During last years, tetrahydroquinolines have improved their synthetic importance due to their

biological activity from a pharmaceutical and medicinal point of view. A great number of substituted

tetrahydroquinolines has been discovered in nature, especially in plants. Their biological activity and

industrial applications stimulate researchers all over the world and, as a result, a huge number of

synthetic approaches have been developed. One of the most used and well-known strategies involves

pericyclic reactions and, among all this type of reactions, aza-Diels-Alder approach is a valid

alternative; Povarov reactions can be included in this area. Lewis catalyzed Povarov processes have

awakened attention especially when Lanthanide salts are used. Among them, Cerium(III) has attracted

researcher’s interest as its salt CeCl3∙7H2O because it is an ecofriendly and cheap reagent, which can

be easily employed also in air and non-anhydrous conditions. The aims of this work are: (i) study of

some aspects of the mechanism of a Povarov reaction of 3,4-dihydro-2H-pyran (DHP), aniline and

benzaldehyde; (ii) study of catalyst and temperature effect of the Povarov reaction of indolines and N-

Vinyl-2-pyrrolidone; (iii) synthetic applications of Povarov reaction to these substrates. This last point

afforded to the synthesis of unprecedented products.

Keywords

Povarov reaction, Lewis acids, Tetrahyroquinolines derivatives, Metal iodides, Cerium

trichloride heptahydrated.

2

Resumo

Nos últimos anos, as tetrahidroquinolinas melhoraram a sua importância sintética devido à

sua atividade biológica do ponto de vista farmacêutico e medicinal. Um grande número de

tetrahidroquinolinas substituídas foi descoberto na natureza, especialmente em plantas. Sua atividade

biológica e aplicações industriais estimulam pesquisadores em todo o mundo e, como resultado, um

grande número de abordagens sintéticas foram desenvolvidas. Uma das estratégias mais utilizadas e

bem conhecidas envolve reações pericíclicas e, entre todo esse tipo de reações, a abordagem aza-

Diels-Alder é uma alternativa válida; As reações de Povarov podem ser incluídas nesta área. Os

processos de Povarov catalisados por Lewis despertaram atenção especialmente quando os sais de

Lantanídeos são usados. Entre eles, o Cerium (III) atraiu o interesse do pesquisador como seu sal

CeCl3∙7H2O porque é um reagente ecofriendly e barato, que pode ser facilmente empregado também

em condições de ar e não anidras. Os objetivos deste trabalho são: (i) estudar alguns aspectos do

mecanismo de uma reação de Povarov de 3,4-dihidro-2H-pirano (DHP), anilina e benzaldeído; (ii)

estudo do efeito de catalisador e temperatura da reação de Povarov de indolinas e N-vinil-2-

pirrolidona; (iii) aplicações sintéticas da reação de Povarov a esses substratos. Este último ponto

conferido à síntese de produtos sem precedents.

Palavras-Chave

Reação de Povarov, ácidos de Lewis, Derivados de Tetrahroquinolinas, iodetos de metais, Tricloreto

de cerio heptaidratado.

3

Table of contents

Abstract 1

Keywords 1

Resumo 2

Palavras-Chave 2

Index of Figures 5

Index of Schemes 6

Index of Tables 9

Index of Equations 10

Abbreviation List 11

1. Pericyclic reactions 13

1.1 Sigmatropic reactions 14

[3,3]-Rearrangement 14

Claisen rearrangement 15

Cope rearrangement 15

[2,3]-Rearrangements 15

1.2 Electrocyclic reactions 17

1.3 Cycloadditions 17

Diels-Alder reaction 17

Imino Diels-Alder reactions (Povarov reactions) 17

1.3.1 Applications of Povarov reactions 19

Brønsted acids and other catalysts 19

Lewis acids as catalysts 21

2. CeCl3, CeCl3∙7H2O and CeCl3∙7H2O/NaI 25

2.1 Organocerium compounds 25

2.2 Application of CeCl3∙7H2O in reductions 28

2.3 CeCl3∙7H2O /NaI 30

4

Applications of CeCl3∙7H2O /NaI 31

3. Tetrahydroquinolines: applications and synthesis 34

3.1 Application of tetrahydroquinolines 35

Chemotherapeutic targets 35

Pharmacodynamic targets 36

Application in Chemistry 38

3.2 Synthesis of tetrahydroquinolines 39

Intramolecular oxidative-cyclization/Lactamization (Formation of N-C2 bond) 39

Conjugate addition-cyclization sequence (Formation of C2-C3 bond) 40

Intramolecular cyclization of ene-C=N functionality (formation of C3-C4 bond) 42

Intramolecular Friedel-Crafts related reactions (formation of C4-C4a bond) 42

Photochemical reactions (formation of C8a-N bond) 44

Diels-Alder approach (Formation of N-C2 and C3-C4 bonds) 44

Brønsted acid-catalyzed reactions (Formation of three or more bonds) 45

4. Experimental section 46

Aim of the work 46

Results and discussion 47

Materials and methods for the synthesis of compounds 52

Characterization of compounds 55

5. Conclusions 63

6. References 64

7. Annexes 71

5

Index of Figures

Figure 1. Intermediate of the Brønsted acid catalyzed Povarov reaction between N-phenyl-C-

methoxycarbonyl imine and methylenecycloprpopane.

Figure 2. Benzastatins C 140 and D 141.

Figure 3. Penigequinolone A 142(S) and B 142(R) and Peniprequinolone 143.

Figure 4. (+)-Sceletium A-4 144, (+)-Tortuosamine 145 and (+)-N-Formyltortuosamine 146.

Figure 5. Molecules showing efficacy against HIV virus.

Figure 6. Interesting antibacterial agents.

Figure 7. Molecules used in the treatment of Malaria (Plasmonium falciparum’s farnesyltransferase).

Figure 8. Molecules having interesting activity on ion channels.

Figure 9. Molecules with important role on membranes and neurotransmitter receptors.

Figure 10. Selective Estrogen Receptors Modulator (SERM).

Figure 11. Selective Androgen Receptor Modulator (SARM).

Figure 12. Tetrahydroquinolines used as electron donors in dye-sensitized solar cells (DSSC).

Figure 13. Chiral ligand for Rhodium-catalyzed hydrogenation of amino acrylates.

6

Index of Schemes

Scheme 1. Formation of lactones.

Scheme 2. Chlorination of methane.

Scheme 3. Claisen rearrangement.

Scheme 4. Cope rearrangement.

Scheme 5. Oxy-Cope reaarangement.

Scheme 6. [2,3]-Rearrangement.

Scheme 7. Synthesis of allylic sulfoxides by [2,3]-rearrangement.

Scheme 8. Stereospecific conversion of 3,4-dimethylcyclobutene to two different isomeric dienes.

Scheme 9. Reverse process that affords to syn-3,4-dimethylcyclobutene.

Scheme 10. Synthesis of Periplanone B.

Scheme 11. Classification of Diels-Alder reactions.

Scheme 12. General Povarov reaction.

Scheme 13. Original Povarov reaction.

Scheme 14. The two possible Povarov reaction mechanism.

Scheme 15. Brønsted acid catalyzed Povarov reaction between N-phenyl-C-methoxycarbonyl imine

and methylenecycloprpopane.

Scheme 16. Three component Povarov reaction catalyzed by a chiral BINOL type phpsphoric acid

derivative.

Scheme 17. Povarov reaction catalyzed by an achiral Brønsted acid and a chiral urea derivative.

Scheme 18. Povarov reaction catalyzed by Titanium(IV) oxide and UV light staring from ethanol and

3-nitrotoluene.

Scheme 19. Synthesis of 4-aryl-3-methyltetrahydroquinoline from N-benzylanilines and formaldehyde.

Scheme 20. Synthesis of 2-sypiro-tetrahydroquinoline from isoeugenol and 3-aryliminosatins.

Scheme 21. Synthesis of 2-aryl-1,2,3,4-tetrahydroquinolines using N-vinyl-2-pirrolindone as

dienophile.

Scheme 22. Sc(OTf)3 catalyzed Povarov reaction.

Scheme 23.Synthesis of the first G protein-coupled estrogen receptor using Povarov chemistry.

7

Scheme 24. Povarov reaction using catalytic amount of ytterbium complex.

Scheme 25. Synthesis of trans-(2S,5S)-(1,1-diphenylmethyl)pyrrolidine.

Scheme 26. Synthesis of VEGF-R2 kinase inhibitor.

Scheme 27. Examples of the activity of Cerium(III) trichloride on C=C bonds.

Scheme 28. Luche’s mechanism with NaBH4 and MeOH.

Scheme 29. Synthesis of 1,9-Deoxypreaxinellamine.

Scheme 30. Synthesis of WRC-0571.

Scheme 31. Synthesis of biologically active 3-mercapto-2(1H)-pyridinones.

Scheme 32. Use of CeCl3∙7H2O/NaI in the Knoevagel condensation of ethyl-tert-butylmalonate with

aromatic or heteroaromatic aldehydes.

Scheme 33. Synthesis of furan derivatives by cyclization reaction of homobenzylic alcohols.

Scheme 34. Synthesis of S-(-)-Pulegone.

Scheme 35. Synthesis of the N-protected nine component of Griseoviridin.

Scheme 36. Different strategies for tetrahydroquinolines synthesis.

Scheme 37. Oxidative cyclization of aromatic aminoalcohols to tetrahydroquinoline derivatives.

Scheme 38. Plausible mechanism of oxidative cyclization of aromatic aminoalcohols to

tetrahydroquinoline derivatives.

Scheme 39. Conjugate addition-cyclization sequence with electron-deficient alkenes catalyzed by

Rhodium.

Scheme 40. Tandem conjugate addition-cyclization sequence of lithium (R)-N-benzyl-N-(α-

methylbenzyl)amide to aromatic imines having an electron-deficient alkene in ortho position.

Scheme 41. Typical Radical Addition Cyclization Elimination(RACE) reaction.

Scheme 42. Synthesis of 1,2,3,4-tetrahydroquinolines from ω-vinylimines.

Scheme 43. Synthesis of biquinoline derivatives from homoallylamines from primary amines and

quinolonecarboxyaldehyde.

Scheme 44. Acid-catalyzed reaction of 1-allyl-1-N-arylaminocyclohexanones.

Scheme 45. Synthesis of tetrahydroquinolines starting from primary amines sulfonamides having an

aryl group in ϒ-position under photochemical conditions.

Scheme 46. Corey’s Diels-Alder approach to tetrahydroquinolines synthesis.

8

Scheme 47. Brønsted acid catalyzed Povarov reaction with a range of different imines and

methylenecyclopropanes.

Scheme 48. Alternative one pot synthesis of isoindolo[2,1-a]quinolone derivative based on a Povarov

cyclocondensation domino approach.

Scheme 49. Povarov reaction between3-aminoacetophenone, benzaldehyde, cyclopentadiene and

TFA:

Scheme 50. Synthesis of spiro-tetrahydroquinolines from arylamines and keto sugars.

Scheme 51. Povarov reaction between aniline, benzaldehyde and 3,4-dihydro-2H-pyran.

Scheme 52. Povarov reaction between indolines, benzaldehyde and N-Vinyl-2-pirrolidinone.

Scheme 53. Povarov reaction with different indolines.

Scheme 54. Results of the trial for the stability of the diastereoisomers.

Scheme 55. Synthesis of indoline.

Scheme0 56. Povarov reaction with different temperatures and metal iodide sources.

Scheme 57. Synthesis of substituted indolines.

Scheme 58. Povarov reaction starting with different indolines.

9

Index of Tables

Table 1. Syn : anti ratio of Povarov reaction at -10°C in solvent/solventless conditions.

Table 2. Syn : anti ratio and yield of Povarov reaction at different temperatures and catalyst.

Table 3. Synthesis of the substituted indolines.

Table 4. Result of the Povarov reaction with substituted indolines.

10

Index of Equations

Equation 1. Preparation of organocerium compounds by reaction of Grignard with organolithium

reagents.

Equation 2. Exchange equation of CeCl3 and NaI.

Equation 3. Exchange equation of the 1:1 combination of CeCl3 and NaI.

11

Abbreviation List

DHP 3,4-dihydro-2H-pyran

HAD Hetero Diels Alder

DFT Density Functional Theory

MCP Methylenecyclopropane

BINOL 1,1’-Bi-2-naphthol

dr Diastereomeric ratio

ee Enantiomeric ratio

UV Ultra Violet light

DBU 1,5-Diazabiciclo(5.4.0)undec-5-ene

DTBP 2,6-di-tert-butylpyrindine

Ce Cerium

NaBH4 Sodium BoroHydride

XPS X-Ray Photoelectron Spectroscopy

MCR Multi Component Reaction

ETBM Ethyl-Tert-ButylMalonate

HIV Human Immunodeficiency Virus

RT Reverse Transcriptase

DNA Deoxyribonucleic Acid

tRNA Transfer RiboNucleic Acid

SERM Selective Estrogen Receptor Modulator

SARM Selective Androgen Receptor Modulator

DSSC Dye-Sensitizied Solar Cells

RACE Radical Addition Cyclization Elimination

GC-MS Gas Chromatography-Mass Spectroscopy

TLC Thin Layer Chromatography

MeCN Acetonitrile

12

r.t. Room Temperature

NaBH3CN Sodium Cyano BoroHydride

N2 Molecular Nitrogen

CH3COOH Acetic Acid

XI Metal Iodide

LiI Lithium Iodide

KI Potassium Iodide

CuI Copper Iodide

NaI Sodium Iodide

NMR Nuclear Magnetic Resonance

CH2Cl2 DichloroMethane

Hex Hexane

EtOAc Etil Acetate

MgSO4 Magnesium Sulphate

HCl Chloride Acid

NaHCO3 Sodium Bicarbonate

Na2SO4 Sodium Sulphate

NaOH Sodium Hydroxide

Et2O DiEthyl Ether

Mp Melting point

13

1. Pericyclic Reactions

Organic reactions may take place in different ways. Breaking and forming of bonds may involve

polar (ionic) intermediates (heterolytic pathway) or radicals (homolytic pathway). In the ionic processes,

electrons “flow” from an electron-rich atom to an electron-poor one: formation of lactones is a typical

example in which, the formation of the desired product involves five steps and four cationic

intermediates: it’s acid-catalyzed and, because it is an ionic reaction, electrons move towards the

positive charge, as reported in Scheme 1.

Scheme 1

Radical reactions have a different pathway: radicals are formed from the homolytic scission of a

covalent bond generally caused by heating or irradiation and they are very reactive. Typically, a

radical reaction has three steps:

1. Initiation, where there is formation of the radicals;

2. Propagation, where the reaction happens;

3. Termination, where the reaction stops and the free radicals are not present anymore.

Classical example of a radical reaction is the chlorination of methane (Scheme 2):

Scheme 2

14

There is another kind of organic processes that differ from the first two: pericyclic reactions in which

electrons do not flow linearly but they move roundly without anionic or cationic intermediates and for this

reason, these reactions are called “pericyclic”.1 There are three main classes of pericyclic reactions:

Sigmatropic rearrangements;

Electrocyclic reactions;

Cycloadditions.

1.1 Sigmatropic Reactions

A sigmatropic reaction is a pericyclic reaction wherein the net result is that a σ-bond is changed

to another σ-bond in an intramolecular process: as a consequence, a substituent moves from one part

of a π-bonded system to another part with simultaneous rearrangement of the π system itself.

[3,3]-Rearrangement

-Claisen rearrangement: This sigmatropic process is called [3,3] because the new-formed σ-bond

has a [3,3] relationship with the previous one. The most famous is the Claisen rearrangement of allyl

vinyl ethers, under heating conditions, which give a γ,δ-unsaturated carbonyl compound, that is more

difficult to obtain respect to the α,β ones. The process occurs under a six-member cyclic chair-like

transition state, which allows predicting the stereochemistry of the final alkene (Scheme 3).2

Scheme 3

-Cope rearrangement: The Cope Rearrangement is the thermal isomerization of a 1,5-diene to a

thermodynamically more stable isomer (Scheme 4):

Scheme 4

An interesting variant is the Oxy-Cope rearrangement (Scheme 5), in which a hydroxy group in a

suitable position affords to an enolether that then tautomerizes to the corresponding carbonyl

compound. The tautomerization is the driving force for this process. The use of a strong base promotes

and speeds up the reaction.3

15

Scheme 5

[2,3]-Rearrangements

This type of rearrangement has a five-membered transition state. The substrate is a benzyl allyl ether

that isomerizes to a final product with a new C-C σ bond (Scheme 6):

Scheme 6

Heteroatoms like sulfur (S) or selenium (Se) can change their oxidation state by two, providing two bond-

forming electrons. This makes the reaction particularly useful for the synthesis of allylic sulfoxides. The

starting alcohol reacts with PhSCl and gives a not stable sulfenate ester; pyridine promotes the [2,3]

rearrangement involving O and S. The product is an allylic sulfoxide (Scheme 7):4

Scheme 7

1.2 Electrocyclic reactions

The main feature of these pericyclic reactions is the formation or breaking of a ring. In this

process, only one σ-bond is broken (or formed) across the end of conjugated π-system.5 One of the

most known example is the stereospecific conversion of 3,4-dimethylcyclobutene to two different

isomeric dienes (Scheme 8):

16

Scheme 8

Syn 3,4-dimethylcyclobutene gives only E,Z-2,4-hexadiene while the trans one gives the E,E diene

product.6 The reverse process affords to the original diastereomer (Scheme 9):

Scheme 9

The methyl substituents move as new σ-bonds are formed. Schreiber et al., in 1984, introduced

electrocylic reactions in the final steps of the synthesis of Periplanone B, as described in Scheme 10:7

Scheme 10

In the initial step, an Oxy-Cope rearrangement affords to the cyclobutene structure that isomerizes, as

an electrocyclic ring opening, to a 12-membered cyclic carbonyl compound (mixture of Z/E isomers).

The transformation to the trans one is favored by irradiation.

17

1.3 Cycloadditions

According to the IUPAC Gold Book,8 in a cycloaddition reaction two or more molecules reacts

together and a cyclic product with a reduction of bond multiplicity is formed. The most famous

cycloaddition reaction is the “Diels-Alder” reaction, discovered in 1928 by Otto Diels and Kurt Alder, that

were awarded the Chemistry Nobel prize in 1950.9

Diels-Alder reaction

During the years, Diels-Alder’s reaction has been used in a wide range of synthetic applications

and it has become an important part in processes of synthesis of polycarbo- and polyheterocyclic

derivatives. This reaction is an excellent alternative for the construction of six-membered rings with a

great control of enantio-and diastereoselectivity. This family of reactions can be distinguished in Carbo-

Diels Alder and Hetero-Diels Alder. The most important of the latter one is imino(aza)-Diels Alder

reactions (Scheme 11):

Scheme 11

Imino Diels-Alder reactions (Povarov reactions)

An inverse electron-demand Diels–Alder reaction is a particular type of Diels–Alder

cycloaddition between an electron-rich dienophile and an electron-poor diene. In the early 1960s, the

Russian chemist Povarov developed an alternative acid-catalyzed inverse electron demand [4+2]

18

cycloaddition between electron-rich dienophiles and N-arylimines, in order to synthetize 1,2,3,4-

tetrahydroquinolines (Scheme 12).10

Scheme 12

This reaction is generally classified as aza- or imino-Diels Alder and allows very versatile applications

of a wide range of catalysts and a huge number of substrates. Furthermore, it can be also performed in

a three-component way, using a dienophile and N-arylimines generated in situ, that are often unstable

and difficult to isolate. In his first pioneering work, Povarov made the reaction of ethyl vinyl ether or

sulfide and N-arylaldimine under BF3-OEt2 to obtain 2,4 substituted tetrahydroquinolines, lately

converted into the corresponding quinolone (Scheme 13):11,12

Scheme 13

Based on this work, the same author and his research group discussed and proposed two possible

mechanisms that are still debated until now (Scheme 14):13

19

Scheme 14

The first is stepwise and goes through the ionic intermediate A with a final intramolecular electrophilic

substitution of a carbenium ion; the second one passes through the asynchronous transition state B.

Researchers believe that the first one is more probable.14

1.3.1 Applications of Povarov reactions

Brønsted acids and other catalysts

Brønsted acid are widely applied in Povarov chemistry. Recently, Rios-Gutierrez and coworkers

made a DFT study of the Brønsted acid Povarov reaction molecular mechanism between N-phenyl-C-

methoxycarbonyl imine and methylenecyclopropane (MCP) (Scheme 15):15

Scheme 15

The study demonstrates that the electrophilic character of the starting imine seems not to be sufficiently

electrophilic for the nucleophilic attack at MCP and that Brønsted acid catalyzed Povarov reaction are

domino processes. The electrophilicity of the imine is enhanced by protonation of the nitrogen and the

reaction starts with a nucleophilic attack on MCP that affords to a cationic intermediate C (Figure 1):

20

Figure 1

That intermediate C undergoes to an intramolecular Friedel-Crafts reaction to give the desired product.

However, the rate-determining step is the initial electrophilic attack. The advantages of using Brønsted

acids are that on one hand, they change the entire molecular mechanism and on the other, they favor

the intramolecular Friedel-Craft step.

Brønsted acids catalyzed Povarov chemistry is applied in other interesting examples. Shi’s research

group studied a three-component Povarov reaction where the catalyst is a chiral BINOL type phosphoric

acid derivative (Scheme 16), obtaining the final tetrahydroquinoline in high diastereomeric ratio (up to

99:1) and enantiomeric excess (97%):16

Scheme 16

Hao and coworkers developed an alternative method using an innovative combined two-catalyst system

which is composed of an achiral strong Brønsted acid and a chiral urea derivative (Scheme 17). The

advantage is the simultaneous and stereocontrolled creation of almost three stereogenic centers from

achiral substrates: 17

21

Scheme 17.

Lewis acids as catalysts

Lewis acids have been applied in Povarov chemistry since first Povarov’s publication. In last

years, CAN-catalyzed reactions have gained attention as an alternative and efficient method to

synthetize substituted tetrahydroquinolines. A wide range of anilines and vinyl ethers has been tested:

the method can be applied due to its convience.18 Similarly, a heterogeneous solution of 3-nitrotoluene,

ethanol and titanium(IV) oxide upon UV irradiation formed 4-ethoxytetrahydroquinoline (Scheme 18):19

Scheme 18

Interestingly, the reaction mechanism includes the formation of ethyl vinyl ether, aniline and N-

ethyldene-p-methylaniline.

Vladimir Kouznetzov, one of the most important researchers in the field of heterocycles synthesis,

studied a process for the synthesis of 4-aryl-3-methyltetrahydroquinoline starting from substituted N-

benzylanilines and formaldehyde in the presence of BF3∙OEt2. In this process iminium ions are used as

dienophiles (Scheme 19):20

22

Scheme 19

2-spiro-tetrahydroquinoline can be synthetized in moderate yield from the BF3∙OEt2 catalyzed reaction

between isoeugenol and 3-aryliminoisatins (Scheme 20):21

Scheme 20

N-Vinyl-2-pyrrolidone is an important and interesting dienophile in Povarov chemistry. A recent

application on the synthesis of 2-aryl-1,2,3,4-tetrahydroquinolines was reported by Astudillo (Scheme

21): 22

Scheme 21

Also Sc(OTf)3 was used as Lewis acid catalyst in reactions for the synthesis of tetrahydroquinolines

using as dienophiles unsaturated lactams with endo- and exocyclic C-C bonds (Scheme 22):

23

Scheme 22

The overall diastereoselectivity was very low (from dr:50:50 to dr:85:15) but in the case of 2-furyl

substituent a single diastomer was obtained.23 A similar Sc(OTf)3 approach was used for the synthesis

of the first G protein-coupled estrogen receptor and its analogues (Scheme 23):

Scheme 23

A single crystal X-Ray analysis confirmed the structure of the tetrahydroquinoline derivative and

additional work on this type of compounds using Povarov chemistry is present in literature.24-26

The enantioselective Povarov reaction with chiral Lewis acids is an alternative approach, although up

to date it is still an unexplored area of research, because most chiral Lewis acids cannot work in a basic

environment being the catalytic cycle blocked.27,28 Kobayashi and Ishitani developed an efficient method

using a catalytic amount of a chiral ytterbium complex (Scheme 24):

24

Scheme 24

The catalyst was synthetized starting from DBU, (R)-(+)-BINOL, Yb(OTf)3 and 2,6-di-tert-butylpyrindine

(DTBP). The products have high diastereoselectivity (from dr: 94:6 to dr: 98:2).29

25

2. CeCl3, CeCl3∙7H2O and CeCl3∙7H2O/NaI

More ecofriendly chemical processes are one of the most desired aspirations for organic

chemists. Over the last years, a great number of attempts has been done in order to study and stimulate

the utilization of Lewis acid promoters in common organic reactions, because of their high resistance to

air and water, low cost and toxicity, stability towards oxygen and easy availability, that makes them really

environment friendly catalysts.30,31 In particular, Lanthanides possesses all of this properties and the

corresponding salts can be used as ecofriendly Lewis acid promoters. Recently, their importance has

been improved in a huge part of organic transformations. Among all the “rare earths”, Cerium is the most

abundant: even Tin, Zinc and Cobalt are present on Earth in lower amount respect to it.

Cerium has two stable oxidation states: Ce4+ is used as one-electron oxidant for a wide range of

processes.32 The electronic configuration of Ce atom is [Xe] 4f15d16s2, where the 4f shell remains

inactive and it is shielded by the 5s2 and 5p6 orbitals. No s-donor-p-acceptor bonding mode is possible

because Cerium(III) is a tripositively charged ion.33 According to Pearson’s principle,34 Ce3+ is a hard

cation and, as result, has a deep affinity to hard bases. The oxophilicity of Cerium(III) cation is due to

the high Lewis acidity combined with ionic bond contributions,35 Lewis acidity decreases as ionic radius

increases, so CeCl3 has to be seen as a mild Lewis acidic promoter.36 In organic reactions, several

Cerium salts have been used but the most common are the ones with halides, nitrate and triflate. The

cost and the use of triflates promoters is not practicable at industrial scale: in addition, there are some

concerns about its application in experimental procedures. These considerations shift the attention to

CeCl3, which has large applications both in hydrated and anhydrous forms.

2.1 Organocerium compounds

Firstly employed by Inamoto,37 organocerium derivatives are used to be added to electrophilic

compounds, which have acidic hydrogens or a high reduction potential.38 They must be prepared freshly

and display interesting properties such as high nucleophilicity and low basicity. They can be prepared

by reaction of Grignard with organolithium reagents (Equation 1):

RLi + CeCl3 ⟶ RCeCl2 + LiCl

RMgX + CeCl3 ⟶ RMgX ∙ CeCl3

Equation 1

This strategy can be applied in different situations: organocerium addition is a valid alternative in a series

of carbon-carbon bond forming reaction.39 Using this type of promoter, alcohols can be obtained from

carbonyl compounds even from starting materials capable of enolization.40 The superiority of

organocerium can be seen in the synthesis of trans-(2S,5S)-(1,1-diphenylmethyl)pyrrolidine as shown

in Scheme 25:

26

Scheme 25

When the starting pyrrolidine reacts with PhMgBr/CeCl3, the resulting alcohol is obtained in high yield

(92%): if the Grignard reagent is used alone, the complete degradation of the starting ester is

observed.41

Other important pharmacological targets are carbon-nitrogen and carbon-carbon multiple bonds:42 in

the first case, the use of organocerium derivatives is a real alternative since the double addition to

tertiary carbamines and nitriles has been proved to be efficient.43 An example is the synthesis of VEGF-

R2 kinase inhibitor which has a significant oral activity against tumor growth in murine models. This

procedure is easily reproducible and scaled by calculating the optimal and necessary CeCl3 quantity to

be milled (Scheme 26):44

27

Scheme 26

Some methods have been studied in order to overcome the inertness of C=C bonds towards nucleophilic

attack and they are still studied because of the very harsh reaction conditions.45 However, some

example have been published and they are reported in Scheme 27. The procedure is efficient and the

products are obtained without chromatographic purification in a very good yield (respectively 97%, 86%

and 48%).46

Scheme 27

28

2.2 Application of CeCl3∙7H2O in reductions

Reduction in organic synthesis is the removal of oxygen from and/or the addition of hydrogen to

a certain functional group of a molecule. The ideal goal is the selectivity: the complete reduction of

insaturations in a molecule is achieved without any particular trouble, while reducing one particular part

respect to another is a useful but a hard challenge. As a rule of thumb, the more active is the catalyst,

the less selective is his action: a very low active catalyst, mild conditions and a good operation rate are

the ideal guidelines to obtain high selectivity in this kind of reactions. In order to achieve it, additives as

Brønsted or Lewis acids, play an important role,47 in particular, Lewis acids shows a significant

improvement on selectivity.48 CeCl3∙7H2O is one of the most effective among all the lanthanide salts and

it has been employed in the reduction of carbonyl groups.

The utility of CeCl3 combined with NaBH4 was firstly demonstrated by Luche (Scheme 28):49

Scheme 28

CeCl3∙7H2O is crucial in the conversion of NaBH4 to [BH4-n(OMe)n]-. The carbonyl group is activated by

hydrogen bonding with methanol and the borohydride attacks the carbonyl carbon to give the

corresponding alcohol. This strategy maintains the stereochemistry of the starting material unaltered.50

Luche’s work on the combination of CeCl3 with metal hydride is a valid alternative to classical reductions

and it has been used for the synthesis of complex biologically active molecules. Baran’s research group

applied it in an easy pathway for the synthesis of 1,9-deoxypreaxinellamine, a marine-derived natural

product, in which Cerium(III) chloride allows the successful reduction of the carbonyl group because of

the high covalent character of cerium-oxygen bond that makes the reduction irreversible and kinetically

favored (Scheme 29): 51

29

Scheme 29

In the pharmaceutical industry, adenosine is a very important nucleoside: during the years, a great

number of adenosine receptors ligands have been made.52 Among all of them, WRC-0571, an

antagonist of adenosine’s A1 receptor, is probably the one with the best selectivity and potency.53 Jin’s

research group studied a new alternative synthesis of WRC-0571, using CeCl3∙7H2O (Scheme 30):54

30

Scheme 30

In the previous syntheses of WRC-0571, the use of only NaBH4 did not give satisfying results:55 Using

Cerium trichloride heptahydrated the reaction is almost quantitative and with a high degree of

stereoselectivity.

2.3 CeCl3∙7H2O/NaI

As discussed before, CeCl3∙7H2O is interesting as a Lewis acid for its mildness and stability in

water.56 Crystallization water can influence the Lewis acid reactions which have to be made under

anhydrous conditions.57 However, catalytic activity of the trivalent Lanthanide salts is due to their large

ionic radii and of the formation of an equilibrium between the Lewis acid and water.58 In order to find an

environmentally friendly promoter, Bartoli’s research group combined CeCl3∙7H2O with NaI. Sodium

iodide improves the activity of CeCl3∙7H2O: other Cerium halides (CeBr3, CeI3) show a decreased activity

compared to Cerium(III) chloride. The exchange equation (Equation 2)

CeCl3 + nNaI(n = 1 − 3) ⟶ CeCl3−nIn + nNaCl

Equation 2

is not a possible explanation to the NaI acceleration effect because CeBr3 and CeI3, even if they are

more soluble, show lower results compared to CeCl3∙7H2O.59 Without any doubt, the 1:1 combination

give CeCl2I∙7H2O species (Equation 3):

31

CeCl3 ∙ 7H2O + NaI ⟶ CeCl2I ∙ 7H2O + NaCl

Equation 3

Even if this complex hasn’t been characterized, it could be a better Lewis acid than its only hydrated

precursor.60 The formation of the complex can be explained using Pearson’s principle: the hard CeCl3

can probably interacts with the iodide ion, forms this species and, as a result, the nucleophilicity of the

donor improves the electrophilic property of the Lanthanide metal.61,62

An X-ray photoelectron spectroscopy (XPS) analysis has been made in order to understand how NaI

interacts with CeCl3∙7H2O. Bartoli and co-workers found that the reaction takes place in heterogeneous

phase on the Cerium salt surface and not in the solution phase.63 A chlorinated bridge oligomeric

structure of Cerium(III) trichloride is formed and broken by an I- species. The result of this process is a

more powerful Lewis acid promoter.64 An other important aspect is crystallization water: CeCl3∙7H2O

/NaI promoted procedures work better in presence of water and the overall activity is even amplified.

This is demonstrated by using anhydrous CeCl3 instead of CeCl3∙7H2O: catalytic activity of the CeCl3-

NaI is absent. However, adding one or more equivalent of water to the dry mixture generates more

active Lewis acid species and the reaction goes with good results.65 During the last years, this catalytic

system has been applied in numerous organic reactions and processes in order to demonstrate its

efficiency and economy.

Applications of CeCl3∙7H2O/NaI

During last years, industrial companies have been demanding to organic chemists the

improvement or the development of new synthetic pathways for compounds. In this area or research,

MCR (Multi Component Reactions) have increase their importance as an efficient procedure in terms of

economical and ecological impact.66,67 Moreover, MCR catalyzed by Lewis acids have increased their

synthetic interest.

Recently, biologically active 3-mercapto-2(1H)-pyridinones with anti-bacterial and anti-fungal activity

have been synthetized by a CeCl3∙7H2O/NaI three-component diastereoselective reaction (Scheme

31):68-70

32

Scheme 31

CeCl3∙7H2O/NaI catalytic combination can also be useful in Michael additions: this reaction has

interesting applications in organic and bioorganic chemistry.71,72 However, side reactions such as

condensation and polymerization are the big drawback of processes.73 Bartoli and coworkers tried

CeCl3∙7H2O/NaI in the Knoevagel condensation of ethyl-tert-butylmalonate (ETBM) with aromatic or

heteroaromatic aldehydes (Scheme 32):74

Scheme 32

This kind of strategy overcomes two of the major obstacles in Knoevagel reaction: firstly, the inability to

stop the coupling of the aldehydes at the mono addition stage; secondly, the spontaneous

decarboxylation that happens during the reaction of malonic acid monoester.75,76 However, this

procedure does not work for aliphatic aldehydes because of the retro-aldol reaction that brings back to

the starting aldehyde.77 Another interesting application is the cyclization reaction of homobenzylic

alcohols to furan derivatives that was studied by Yeh’s research group (Scheme 33):78

Scheme 33

33

This example demonstrates that CeCl3∙7H2O/NaI is an alternative reagent because it is safer and it can

be used in mild conditions although yields are not very high.79

One of the advantage of CeCl3∙7H2O/NaI is its role of promoting selectivity in processes: Bartoli’s

research group applied this protocol to the synthesis of S-(-)-Pulegone (Scheme 34)

Scheme 34

and of a N-protected nine-membered component of Griseoviridin (Scheme 35):79,80

Scheme 35

34

3. Tetrahydroquinolines: applications and synthesis

In the agrochemical and pharmaceutical industry, one of the most important and interesting

family of compounds is constituted by nitrogen containing heterocycles that constitutes about the 60%

of all drugs. The tetrahydroquinoline moiety (a benzene ring fused to a piperidine one) is found

commonly in biologically active substances and it is involved in important therapeutic strategies. A very

broad range of 1,2,3,4-tetrahydroquinoline-based natural products has been discovered during the last

three decades:81 Yoo’s research group found in Streptomyces sp. a couple of tetrahydroquinolines

alkaloids derivatives, benzastatins C and D that shows inhibitor activity against lipid peroxidation and

glutamate toxicity (Figure 2).82

Figure 2

Koshino’s group research activity is focused on the discover of 4-(p-methoxyphenyl)-3,4-

tetrahydroquinolin-2(1H)-one natural derivatives: two active pollen-growth from the mycellas mats of

Penicillium Sp. n°410 were isolated.83 The same research group found in Penicillium simplicissimum

Peniprequinolone, an alkaloid derivative with nematicidal properties (Figure 3):84

Figure 3

Later, the same compound was after isolated in the fungus Penicillium janczwskii, obtained from

Prumnopitys andina.85 In nature, different hydrogen patterns different from the common 1,2,3,4-

35

tetrahydroquinolinic one are not present in huge quantity: the compounds shown in Figure 4 are the

most important and they are found in Sceletium plants:86

Figure 4

3.1 Application of tetrahydroquinolines

Chemotherapeutic targets

In recent years, HIV has become a big issue in the world. Some molecules containing the

tetrahydroquinoline skeleton have shown interactions with retroviral targets for anti-HIV therapy (Figure

5):

Figure 5

These compounds, from an experimental outcome, are non-nucleoside, RT allosteric inhibitors and

antagonists of CXCR4, a G-protein coupled chemokine receptor that it is thought to be HIV strains

coreceptor.87-90

Recently, researchers are trying to develop compounds that can be interesting antibacterial agents.

Several tetrahydroquinolines are found to be active against DNA gyrase and methionyl tRNA synthetase

(Figure 6):91,92

36

Figure 6

The center compound can be an alternative in infections treatment because of the so-called “Gram

positive bacterial resitance”.93

Imidazole ring is an important tool in pharmaceutical chemistry and its derivatives showed potential

antimalarial activity: they display a potent cytotoxic effect against Plasmonium falciparum’s

farnesyltransferase (Figure 7):94-98

Figure 7

Pharmacodynamic targets

Tetrahydroquinolines having a guanidine functionality in N-1 position show interesting activity

on ion channels (Figure 8):

37

Figure 8

The first group of compounds possesses neuronal Na+ channel antagonist activity: the second one is

Na+/H+ exchanger inhibitor, important in the treatment of ischemia-reperfusion after a myocardial

infarction, and the third one derives from Povarov chemistry and it is an agonist of BKCa (large-

conductance calcium-activated potassium channel).99-101 It was also demonstrated the important role of

tetrahydroquinolines on membranes and neurotransmitter receptors (Figure 9):

Figure 9

In this sense, the first tetrahydroquinoline derivative 158 is a positive allosteric modulator of the α7

nicotinic acetylcholine receptor; Sumanirole is dopaminergic D2 receptors agonist and 160 is an

antagonist of the serotonin 5-HT3 receptor.102-104 This class of compounds is also acting at steroid

hormone receptors: they can be selective estrogen receptor modulator (SERMs), where the active part

is 6-hydroxy-2-phenyl-1,2,3,4-tetrahydroquinoline system (Figure 10):105

38

Figure 10

Another type of tetrahydroquinoline is found to have a potent anabolic activity and to be a SARM

(Selective Androgen Receptor Modulator) (Figure 11):106

Figure 11

Applications in Chemistry

In recent years, tetrahydroquinolines derivatives with a cyano-acrylic moiety as electron acceptor have

been used as electron donors in dye-sensitized solar cells (DSSC) (Figure 12): 107

Figure 12

Asymmetric synthesis, nowadays, is one of the most interesting and studied topic in organic chemistry.

In this view, also tetrahydroquinoline derivatives have showed to be suitable chiral ligands for Rh-

39

catalyzed hydrogenation acrylates of amino acrylates having a product with a 99% of enantiomeric

excess and for asymmetric Friedel-Crafts indoles reaction are shown in Figure 13:108,109

Figure 13

3.2 Synthesis of tetrahydroquinolines

During the years, researchers have developed different strategies for tetrahydroquinolines

synthesis as shown in Scheme 36:

Scheme 36

In the next sub-sections, some strategies will be discussed.

Intramolecular oxidative-cyclization/Lactamization (Formation

of N-C2 bond)

Fujika’s research group worked on the oxidative cyclization of aromatic aminoalcohols to

tetrahydroquinoline derivatives catalyzed by iridium catalyst (Scheme 37):

40

Scheme 37

In the presence of 5%mol of catalyst (Ir[Cp*IrCl2]2/K2CO3 is the best), a wide range of 3-(2-aminophenyl)-

propanols with different substituents have been tested with good results.110 A plausible reaction

mechanism has been proposed (Scheme 38):111

Scheme 38

The catalytic oxidation of the starting aminoalcohol yields an iridium-hydride species and an aldehyde

D: intermediate E is formed by intramolecular cyclization and then affords to imine C that reacts with the

iridium-hydride species and gives the amido iridium intermediate G. The catalytic cycle finishes with the

exchange of the iodine complex between G and a molecule of starting material: the product is the

desired tetrahydroquinoline.112

Conjugate addition-cyclization sequence (Formation of C2-C3

bond)

The conjugate addition-cyclization sequence involves imine–substituted electron-deficient

alkenes. A rhodium-catalyzed domino reaction of this type was studied by Youn (Scheme 39):113

41

Scheme 39

Arylboronic acids and rhodium catalyst helps the yield of the reaction and favor the formation of

diastereomeric mixture of the tetrahydroquinoline product. Another procedure involves tandem

conjugate addition-cyclization of lithium (R)-N-benzyl-N-(α-methylbenzyl)amide to aromatic imines

having an electron-deficent alkene in ortho position (Scheme 40):

Scheme 40

The product is a 2-aryl-4-aminotetrahydroquinoline-3-carboxylic acid with an excellent diastereo- and

enantioselectivity.114 The conjugate addition of lithium amide, gives the rigid enolate intermediate H. The

second step is the cyclization whose anti selectivity is due to the boat-like intermediate I. In order to

obtain the carboxylic acid, a catalytic reduction catalyzed by Pd(OH)2/C needs to be performed.

42

Intramolecular cyclization of ene-C=N functionality (formation

of C3-C4 bond)

An oxime ether having a 2-pyrrolidone functionality can react and give a diastereomeric mixture

(Scheme 41):115

Scheme 41

This example shows a typical RACE (Radical Addition Cyclization Elimination) strategy, first developed

for the synthesis of martinelline.116 Okamoto’s research group synthetized 1,2,3,4- tetrahydroquinoline

derivatives starting from ω-vinylimines: in this way, it is allowed the introduction of other different

substituents from halogen to hydroxy groups (Scheme 42):117

Scheme 42

Intramolecular Friedel-Crafts related reactions (formation of C4-

C4a bond)

Kouznetsov’s research group found a new alternative way to afford biquinoline derivatives from

homoallylamines prepared by reaction of primary amines and quinolonecarboxyaldehydes (Scheme

43):

43

Scheme 43

In the presence of sulphuric acid, the reaction is very successful.118 The acid catalyzed cyclization

reaction of 1-allyl-1-N-arylaminocyclohexanones is interesting and gives an unexpected spiro-product

(Scheme 44):

Scheme 44

The product on the right is obtained from an intramolecular ipso substitution-alkylation sequence. The

isomeric tetrahydroquinolines on the left are in a 65:35 ratio. The attack of the carbocation (generated

44

in acid conditions) followed by 1,2-shift of the ethyl substituent could be the reason for the presence of

the other product.119

Photochemical reactions (formation of C8a-N bond)

It has been reported the synthesis of tetrahydroquinolines starting from primary amines

sulfonamides having an aryl group in ϒ-position under photochemical conditions (Scheme 45):

Scheme 45

Obviously, the nature of nitrogen substituents has a great influence on the reactivity of the starting

materials. The mechanism proposed involves the formation of a N-iodo intermediate, the homolytic

cleavage of N-I bond, the formation of a sulfonamidyl radical and a conclusive oxidative aromatization.120

Diels-Alder approach (Formation of N-C2 and C3-C4 bonds)

Starting from C2-C3 fragment and an o-substituted aniline, a tetrahydroquinoline derivative can

be synthetized using cyclization of o-azaxylenes with suitable dienophiles.121 Corey and Steihagen

studied a good method that consists in a formal [4+2] cycloaddition of electron-rich alkenes and o-

azaxylenes (Scheme 46):122

Scheme 46

45

Brønsted acid-catalyzed reactions (Formation of three or more

bonds)

Brønsted acids are capable of catalyzing Povarov reaction for the synthesis of

tetrahydroquinoline derivatives. The treatment of different imines and a wide range of

methyleneciclopropanes in the presence of montmorillonite K-10 or triflic acid gives very satisfactory

yields of the desired product (Scheme 47):123

Scheme 47

Khadem and co-workers studied an alternative one-pot synthesis of isoindolo[2,1-a]quinoline derivative

based on a Povarov-cyclocondensation domino approach (Scheme 48):124

Scheme 48

The advantage of this reaction is the fact that only one diastereomer is formed. It can be seen also in

the reaction between 3-aminoacetophenone, benzaldehyde, and cyclopentadiene with TFA (1

equivalent) (Scheme 49):

Scheme 49

The adjacent carbonyl group is responsible for the stabilization of the carbocationic intermediate

generated during the reaction and this fact explains the diasteroselectivity of this reaction. Important

spiro-tetrahydroquinolines can be synthetized starting from arylamines and keto sugars in presence of

p-toluensulfonic acid (Scheme 50):

46

Scheme 50

The reaction starts with the formation of the imine S which is in equilibrium with the enamine form T.

The next Povarov-like [4+2] cycloaddition in the presence of acid catalyst gives the tetrahydroquinoline

product in good yield: this compound showed immunobiological and cytotoxic activity.125

4. Experimental section

Aim of the work

In the first part of my work, I studied some mechanistic aspects of Povarov reaction of aniline,

benzaldehyde and 3,4-dihydro-2H-pyran (Scheme 51):

Scheme 51

47

The aim of this study was to understand the effect of the amount of solvent (acetonitrile) on the

stereoselectivity of the products.

In the second part of my thesis work, I focused my attention on the optimization of the Povarov reaction

between indolines, benzaldehyde and N-vinyl-pyrrolidone (Scheme 52):

Scheme 52

When the optimization study was finished, I attempted to synthetize tetrahydroquinoline derivatives from

substituted indolines (Scheme 53):

Scheme 53

The obtained tetrahydroquinolines can be applied in useful applications.

Result and discussion

The pilot reaction was carried out in solvent/solventless conditions using aniline, benzaldehyde and 3,4-

dihydro-2H-pyran (DHP), exploring CeCl3·7H2O/NaI as promoting system. The reaction was monitored

by GC-MS and TLC until the starting reagents were consumed or a constant composition of the mixture

was reached, and the final tetrahydroquinolines were obtained as racemates in syn/anti diastereomer

mixture. Based on the previous work of the research group, the reaction in CeCl3∙7H2O/NaI in dry

acetonitrile at 50°C for one hour led to an excellent syn:anti ratio of 15:85 and 82% yield while in

solventless conditions and lowering the temperature at -10°C it afforded to a 73% yield with an

interesting inverted diastereoselectivity (syn:anti 77:23). Generally the reactions performed in solvent

show a strong preference for the anti diastereomer together with slightly lower yields with respect to the

corresponding ones in solventless conditions. The aim of this part of my thesis is to understand if the

different selectivity is due to a different selectivity of the reaction in solvent or solventless conditions and

48

at the same time on a different stability of the two diastereomers in the reaction mixture. Experiments

were made to understand if the selectivity depends on a kinetic or thermodynamic control of the reaction

system in the different conditions. Generally, at high concentration and low temperatures the kinetic

control of the reaction is favored while at high dilution conditions and higher temperatures the

thermodynamic control is allowed. Our strategy was to monitor the diastereomeric outcome of the

reaction with different amounts of solvent: the reactions were performed at -10°C and the results are

shown in Table 1;

Time(h)

Condition 1 2 4 Overnight

Solventless 54:46 81:19 58:42 60:40

1 eq. CH3CN 76:24 71:29 75:25 51:49

5 eq. CH3CN 38:62 48:52 54:46 32:68

5mL. CH3CN 14:86 - 35:65 17:83

Table 1. Syn : anti ratio of Povarov reaction at -10°C in solvent/solventless conditions

As an experimental proof, the solventless reaction gives as a major product the syn-diastereomer. When

the reaction is performed with 1 equivalent of CH3CN at -10°C, a quite constant dr around a 75:25

syn:anti ratio was found during the reaction. The amount of CH3CN was raised to 5 equivalents at -10°C

and the result was that the anti diastereomer starts to be formed and reached a 54:46 syn:anti ratio after

4 hours. At higher dilution, in 5 mL of CH3CN at -10°C, the reaction is strongly anti selective, starting

from a 14:86 to reach after 4 hours a 35:65 syn:anti ratio. After four hours at – 10°C all the reaction

mixtures were left to reach r.t. and stirred overnight, and all were found enriched in the anti diastereomer,

except when 1 equivalent of solvent was used that showed a final 51:49 syn:anti ratio. The last

experiment was performed in 10 mL of CH3CN at r.t. and resulted in a final 17:83 syn:anti ratio after

overnight stirring. These observations confirm that at high concentrations and at low temperatures,

conditions that favor kinetic control, the syn product is favored and low concentration, long reaction

times and high dilutions favor the formation of the anti diastereomer. Another reason for the different

selectivity in solvent/solventless conditions may be the different stability of the two diastereomers in the

reaction mixture. This issue was addressed dissolving in CH3CN a mixture of the two diastereomers

(syn:anti = 41:59), adding the catalyst and left stirring overnight at room temperature (Scheme 54):

Scheme 54

49

The diastereomers were isolated in mixture and the yield and the dr were measured, showing that the

initial dr ratio passed to 32:68 demonstrating that the mixture was enriched in the anti diastereomer.

Significative is the fact that only the 82% of the initial amount of the mixture was recovered, that in more

detail revealed a 5% loss of the anti product and a 37% loss of the syn. These results confirm that the

selectivity of this reaction depends both on kinetic or thermodynamic control in the different reaction

conditions and on a different stability of the two diastereomers in the reaction mixture.

In the second part of my thesis work, I studied the effect of different additives of the reaction in Scheme

52 at different temperatures (0°C-r. t.-40°C). The first step is the synthesis of the starting indoline by

reduction of indole with sodium cyanoborohydride in acetic acid at room temperature (Scheme 55):

Scheme 55

The procedure is efficient and the yields are high. The indoline was synthetized, purified by filtration on

basic alumina (Al2O3) then used in the successive Povarov reaction (Scheme 56):

Scheme 56

These reactions are performed at three different temperatures and with the promoting system

CeCl3∙7H2O/different iodide sources (LiI, KI, CuI and NaI) to evaluate their effect on this Povarov

reaction. The results are shown in Table 2:

50

T NaI CuI KI LiI

dr(syn:anti) Yield dr(syn:anti) Yield dr(syn:anti) Yield dr(syn:anti) Yield

0°C 92.8 60 % 94:6 51 % 91:9 27 % 92:8 18 %

rt 91:9 48 % 95:5 42 % 93:7 48 % 93:7 45 %

40°C 90:10 51 % 95:5 51 % 91:9 45 % 93:7 42 %

Table 2. Syn : anti ratio and yeld of Povarov reaction at different temperatures and catalyst

When the reaction was judged to be complete, the solution was diluted with dichloromethane, filtered

with celite and purified with flash chromatography; the dr vas evaluated by NMR analysis on the

chromatographic fraction of the diasteroisomers isolated in mixture. As shown in the Table 2, except for

a few cases, yields are quite similar and the syn adduct is favored over the anti one. The reason for this

behavior is the electron-donating dienophile, N-vinyl-2-pyrrolidone. According to literature,126,127 we

thought that the syn adduct in high quantity respect to the anti one: this hypothesis was confirmed during

these trials. The optimal reaction conditions derived from these trials are the CeCl3∙7H2O/NaI as catalyst

and 0°C as operating conditions that we apply to the synthesis of a series of substituted indolines that

were prepared by reduction of the corresponding indole with NaCNBH3 in acetic acid at room

temperature under inert atmosphere (Scheme 57), according to literature.128 The results are shown in

Table 3.

Scheme 57

Indoline Yield (%)

214b 92

214c 76

214d 81

214e 89

214f 84

Table 3. Synthesis of the substituted indolines.

Scheme 58 shows the successive Povarov reaction and the results are shown on Table 4:

51

Scheme 58

Indoline Yield (%) dr (syn:anti)

213b 72 80:20

213c 53 90:10

213d 64 95:5

213e 50 92:8

213f 65 95:5

Table 4. Results of the Povarov reaction with substituted indolines.

The reaction mixture was diluted with CH2Cl2, filtered with celite and the product were purified by flash

chromatography (Hex : EtOAc). Unexpectedly, yields are quite high compared to the ones with

unsubstituted indoline and as expected, dr are also quite good in these reactions.

52

Materials and methods for the synthesis of compounds

All reagents and solvents were purchased from commercial suppliers and used without further

purification, unless mentioned otherwise. All reactions were monitored by thin-layer chromatography

using EMD/Merck silica gel 60 pre-coated plates (0.25 mm), and the compounds were visualized either

by using UV light (254 nm), and vanillin stains as developing agent. Purification of the reaction products

was carried out by column flash-chromatography using silica gel (0.040–0.063 mesh). 1H and 13C NMR

spectra were recorded on a Varian Mercury 400 (400 MHz, 100 MHz or 377 MHz respectively). Chemical

shifts are given in ppm with reference to residual H in deuterated solvents as the internal standard.

Coupling constants J are reported in Hz. Diastereomeric ratio was determined by integration of 1H-NMR

signals (H4 and H6) of the mixture of diasteromers. The compounds are described as ”M” for the major

isomer and ”m” for the minor one. Mass spectra were obtained using a gas chromatograph Agilent 6850

equipped with a HP5MS column (0.25 mm diameter) and a mass-selective detector Agilent 5973N, or

using a LC-MS Hewlett Packard series 1100 MSD.

53

Method for the synthesis of 5-phenyl-3,4,4a,5,6,10b-hexahydro-2H-

pyrano[3,2-c]quinoline (211a-b):

In a round bottom flask, aniline (1 mmol) and benzaldehyde (1 mmol) were stirred together in different

amounts of CH3CN according to Table 1 in the presence of anhydrous MgSO4 (200 mg). The mixture

was stirred until imine was completely formed. Then, CeCl3·7H2O (112 mg, 0.3 mmol) and NaI (45 mg,

0.3 mmol) were added, followed by 2,3 dihydropyran (1 mmol) and the mixture was stirred at -10°C until

consumption of starting materials (TLC, hexane/EtOAc, vanillin stain). The mixture was diluted with

CH2Cl2 and washed with HCl 0.5 M. The aqueous layer was extracted with CH2Cl2, then treated with

satured solution of NaHCO3 until basic pH was reached and extracted again with CH2Cl2. The organic

layers were unified, washed with brine and dried over anhydrous Na2SO4. The crude was purified by

flash chromatography over silica (eluent hexane/EtOAc) to afford the pure tetrahydroquinoline products.

The reaction was also performed in solventless conditions.

Method for the synthesis of indolines (212a-f):

In inert atmosphere, to a stirred solution of the starting indole (2-5 mmol) in acetic acid (4-10 mL) was

added NaBH3CN (3-7.5 mmol, 1.5 eqiv.). The reaction was allowed to stir at room temperature and

reaction progress was monitored by TLC. After the reaction was judged to be complete, the reaction

mixture was diluted with H2O, brought to pH~9 with a 2M NaOH solution and extracted with Et2O or

EtOAc. The combined organic layers were washed with H2O, brine, dried over Na2SO3, filtered and

concentrated unfer reduced pressure. The crude product was filtered on basic alumina (Al2O3) or purified

by flash column cromatography (Hex:EtOAc) to yield the corresponding indoline.

54

Method for the synthesis of 1-(3-phenyl-1,2,3,5,6,7-

hexahydropyrido[3,2,1-ij]quinolin-1-yl)pyrrolidin-2-one and its

derivatives (213a-f):

In a round bottom flask, indoline (substituted or not) (1 mmol) and benzaldehyde (1 mmol) were stirred

together in 5 ml of CH3CN in the presence of anhydrous MgSO4 (200 mg). The mixture was stirred until

imine was completely formed. Then, CeCl3·7H2O (112 mg, 0.3 mmol) and MI (X=Na, Li, Cu, K, 0.3

mmol) were added, followed by N-vinyl-2-pyrrolidinone (1 mmol) and the mixture was stirred at 0, 40°C

or room temperature until consumption of starting materials (TLC, hexane/EtOAc, vanillin stain). The

mixture was diluted with CH2Cl2, filtered on a gooch and Celite and evaporate to dryness. The crude

was purified by flash chromatography over silica (eluent hexane/EtOAc) to afford the pure

tetrahydroquinoline derivative products.

55

Characterization of compounds

5-phenyl-3,4,4a,5,6,10b-hexahydro-2H-pyrano[3,2-c]quinoline (211a)

White solid. Mp: 130 °C. 1H NMR (400 MHz, CDCl3) δH (ppm) 7.44-7.28 (m, 6H),

7.10 (t, J= 7.4 Hz, 1H), 6.80 (t, J = 7.4 Hz, 1H), 6.61 (d ,J= 7.9 Hz, 1H), 5.33 (d,

J= 5.4 Hz, 1H), 4.69 (d, J= 2.1 Hz, 1H), 3.88 (m, 1H), 3.58 (dd, J1 = 11.3 Hz, = 3.8

Hz, 1H), 3.43 (td, J= 11.0, 2.3 Hz, 1H), 2.17-2.16 (m, 1H), 1.58-1.42(m, 3H), 1.31-

1.25 (m, 1H); 13C NMR (400 MHz, CDCl3) δC (ppm) 145.1, 141.1, 128.3, 128.1, 127.6, 127.5, 126.8,

119.8, 118.2, 114.4, 72.7, 60.6, 59.3, 38.9, 25.4, 18.0. MW: 265.350 g/mol. MS m/z: 265 (M)+; 234; 220;

194; 129; 117; 91; 77. Data are in accordance with literature.129

5-phenyl-3,4,4a,5,6,10b-hexahydro-2H-pyrano[3,2-c]quinoline (211b)

Viscous oil. 1H NMR (400 MHz, CDCl3) δH (ppm) 7.44-7.29 (m, 5H), 7.24-7.21 (m,

1H), 7.10 (dt, J= 8.0, 1.4 Hz, 1H), 6.70 (t, J= 7.4 Hz, 1H), 6.51 (d, J= 8.0 Hz, 1H),

4.71 (d, J= 10.8 Hz, 1H), 4.38 (d, J= 2.6 Hz, 1H), 4.13-4.08 (m, 2H), 3.71 (td, J=

11.5, 2.5 Hz, 1H), 2.12-2.05 (m, 1H), 1.93-1.77 (m, 1H), 1.71-1.59 (m, 1H), 1.49-

1.44 (m, 1H), 1.36-1.26(m, 1H); 13C NMR (400 MHz, CDCl3) δC (ppm) 144.8, 142.4,

130.9, 129.4, 128.7, 127.9, 127.9, 120.6, 117.4, 114.2, 74.5, 68.6, 54.8, 38.9, 24.2, 22.1. MW: 265.15

g/mol. MS m/z: 265 (M)+, 234; 220; 194; 129; 117; 91; 77. Data are in accordance with literature.129

Indoline 212a

Pale yellow oil. Yields from 74 to 90%. 1H NMR (400 MHz, CDCl3) δH 7.03 (d, J= 7.2 Hz,

1H), 6.93 (t, J=7.6 Hz, 1H), 6.62 (t, J=7.4 Hz, 1H), 6.55 (d, J=7.7 Hz, 1H), 3.44 (t, J= 8.3

Hz, 3H), 2.93 (t, J=8.3 Hz, 3H). 13C NMR (400 MHz, CDCl3) δC 151.64, 129.37, 127.25,

124.68, 118.71, 109.51, 47.37, 29.89; MW: 119.166 g/mol. Data are in accordance with literature.130

56

(±)-3-Methylindoline 212b

Pale red/brown oil. Yield: 92 %. 1H NMR (400 MHz, CDCl3) δH 7.14-6.98 (m, 2H), 6.75 (t,

J=7.3 Hz, 1H), 6.66 (d, J= 7.8 Hz, 1H), 3.71 (t, J= 8.7 Hz, 1H), 3.38 (q, J= 7.7 Hz, 1H), 3.12

(t, J=8.6 Hz, 2H), 1.33 (d, J=6.7 Hz, 3H); 13C NMR (400 MHz, CDCl3) δC 151.2, 134.5,

127.4, 123.5, 118.9, 109.7, 55.5, 36.7, 18.7; MW: 133,193 g/mol. Data are in accordance with

literature.131

(±)-2-Phenylindoline 212c

Yellow oil. Yield: 76 %. 1H NMR (400 MHz, CDCl3) δH 7.51-7.41 (m, 2H), 7.40-7.33

(m, 2H), 7.32-7.26 (m, 1H), 7.16-7.03 (m, 2H), 6,76 (td, J=7.4,1.0 Hz, 1H), 6.69 (d,

J= 7.7 Hz, 1H), 4.97 (t, J=9.0 Hz, 1H), 4.03 (s, 1H), 3.46 (dd, J=15.6, 9.2 Hz, 1H),

3.01 (ddt, J= 15.6, 8.7, 1.1 Hz, 1H); ). 13C NMR (400 MHz, CDCl3) δC 150.5, 144.3, 129.1, 128.7, 128.4,

127.7, 127.6, 126.5, 125.3, 124.7, 119.3, 109.4, 63.6, 39.6; MW: 195,264 g/mol. Data in accordance

with literature.132

5-Bromoindoline 212d

Yellow oil. Yield: 81%. 1H NMR (400 MHz, CDCl3) δH 7.19 (dt, J=2.2, 1.2 Hz, 1H), 7.12-

7.07 (m, 1H), 6.49 (d, J=8.3 Hz, 1 H), 3.73-3.61 (m, 1H), 3.56 (t, 8.4 Hz, 2H), 3.01 (t,

J=8.3,1.0, 2H); 13C NMR (400 MHz, CDCl3) δC 150.7, 131.9, 129.9, 127.7, 110.6, 110.2, 47.7, 29.8;

MW: 198,062 g/mol. Data in accordance with literature.132

5-Methoxyindoline 212e

Pale yellow oil. Yield: 89%. 1H NMR (400 MHz, CDCl3) H 6.72 (dd, J= 2.4, 1.2 Hz,

1H), 6.59 (ddt, J= 8.5, 2.6, 0.9 Hz, 1H), 6.54 (d, J= 8.4 Hz, 1H), 3.97 (tq, J= 8.1, 6.2

Hz, 1H), 3.74 (s, 3H), 3.38 (s, 1H), 3.11 (m, 1H), 2.62 (ddt, J= 15.5, 7.9, 1.0 Hz, 1H), 1.29 (d, J= 6.2 Hz,

3H);13C NMR (400 MHz, CDCl3) C 153.4, 145.2, 131.11, 112.0, 111.4, 110.1, 55.8, 47.7, 30.4; MW:

149,192 g/mol. Data are in accordance with literature.133

57

(±)-5-Methoxy-2-methylindoline 212f

. Yellow oil. Yield: 84%. 1H NMR (400 MHz, CDCl3) δH 6.71–6.70 (m, 1H), 6.59–

6.51 (m, 2H), 4.00–3.90 (m, 1H), 3.73 (s, 1H), 3.40 (m, 1H), 3.10 (dd, J = 15.6,

8.4 Hz, 1H), 2.60 (dd, J = 15.4, 7.6Hz, 1H), 1.27 (d, J = 6.0 Hz, 3H); 13C NMR (400 MHz, CDCl3) δC

153.4, 144.7, 130.6, 112.0,111.6, 109.8, 55.9, 55.6, 38.2, 22.1. MW: 163,219 g/mol. Data are in

accordance in literature.134

1-(4-phenyl-2,4,5,6-tetrahydro-1H-pyrrolo[3,2,1-ij]quinolin-6-yl)pyrrolidin-2-one 213a (M)

White foam. Yield from 18 to 60%. FT-IR (neat, cm-1) 2959, 2928, 2860,

1717,1267; 1H NMR (400 MHz, CDCl3) δH 7.47–7.40 (m, 2H), 7.37 ( t, J = 7.5

Hz, 2H), 7.34–7.27 (m, 1H), 7.03 (d, J= 7.0 Hz, 1H), 6.83–6.66 (m, 2H), 5.69

(dd, J= 11.3, 6.5 Hz, 1H), 4.02 (dd, J=11.0, 2.5 Hz, 1H), 3.42–3.21 (m, 3H),

3.04–2.92 (m, 1H), 2.90–2.81 (m, 1H), 2.79–2.71 (m, 1H), 2.56–2.39 (m, 2H), 2.27-2.11 (m, 2H), 2.08–

1.94 (m, 2H); 13C NMR (400 MHz, CDCl3) δC 175.8, 148.8, 141.6, 129.9, 128.7, 127.7, 126.9, 124.3,

123.6, 119.3 , 117.3, 116.2, 63.0, 56.7, 53.4, 47.8, 42.4, 36.8, 31.4, 28.8, 18.2; Anal Calcd for

C21H22N2O; MS m/z: 318.2 (M+), 232 (100), 204, 185, 156, 135, 115, 91, 65, 41; MW: 318.418 g/mol; C

79.21% H 6.96% N 8.80% Found C 79.25% H 6.94% N 8.78%. Data are in accordance with literature.135

1-(4-phenyl-2,4,5,6-tetrahydro-1H-pyrrolo[3,2,1-ij]quinolin-6-yl)pyrrolidin-2-one 213a (m)

White foam. Yield from 18 to 60%. FT-IR (neat, cm-1) 2959, 2928, 2860,

1717,1267; 1H NMR (400 MHz, CDCl3) δH 7.47–7.40 (m, 2H), 7.37 ( t, J = 7.5

Hz, 2H), 7.34–7.27 (m, 1H), 7.03 (d, J= 7.0 Hz, 1H), 6.83–6.66 (m, 2H), 5.42

(dd, J= 6.4, 2.3 Hz, 1H), 3.82 (dd, J=11.1, 3.1 Hz, 1H), 3.42–3.21 (m, 3H), 3.04–

2.92 (m, 1H), 2.90–2.81 (m, 1H), 2.79–2.71 (m, 1H), 2.56–2.39 (m, 2H), 2.27-2.11 (m, 2H), 2.08–1.94

(m, 2H); 13C NMR (400 MHz, CDCl3) δC 175.8, 148.8, 141.6, 129.9, 128.7, 127.7, 126.9, 124.3, 123.6,

119.3 , 117.3, 116.2, 63.0, 56.7, 53.4, 47.8, 42.4, 36.8, 31.4, 28.8, 18.2; Anal Calcd for C21H22N2O; MS

m/z: 318.2 (M+), 232 (100), 204, 185, 156, 135, 115, 91, 65, 41; MW: 318.418 g/mol; C 79.21% H 6.96%

N 8.80% Found C 79.25% H 6.94% N 8.78%. Data are in accordance with literature.134

58

1-(3-methyl-4-phenyl-2,4,5,6-tetrahydro-1H-pyrrolo[3,2,1-ij]quinolin-6-yl)pyrrolidin-2-one 213b

(M)

Light red foam. Yield: 72%. FT-IR (neat, cm-1) 2959, 2928, 2860, 1717,1267; 1H

NMR (400 MHz, CDCl3) δH 7.47 (d, J=7.26 Hz, 2H), 7.40 (t, J= 7.5,7.5 Hz, 2H),

7.3-7.27 (m, 1H), 7.02 (d, J=7.1 Hz, 1H), 6.73 (t, J=8.9 Hz, 1H), 6.67 (t, J=7.4 Hz,

1H), 5.70 (dd, J=11.5, 5.9 Hz, 1H), 5.31 (s, 1H), 4.47 (dd, J=11.0,2.6 Hz, 1H),

3.84-3.74 (m, 1H), 3.35-3.19 (m, 2H), 2.60-2.42 (m, 3H), 2.25-2.09 (m, 2H), 2.09-

1,95 (m, 2H), 0.90 (dd, J=6.5, 3.8 Hz, 3H); 13C NMR (400 MHz, CDCl3) δC 175.3, 148.8, 128.7, 128.5,

128.24, 128.1, 127.0, 124,1, 123.9, 118.1, 116.8, 77.3, 77.1, 76.9, 57.3, 56.8, 53.45, 36.5, 43.7, 42.4,

37.2, 31.5. 18.3, 15.9; Anal Calcd for C22H24N2O; MS m/z: 332.2 (M+), 246 (100), 213, 196, 170, 142,

115, 91, 69, 41; MW: 332.170 g/mol; C 79.48% H 7.28% N 8.43% Found: C 79.51% H 7.31% N 9.37%.

1-(3-methyl-4-phenyl-2,4,5,6-tetrahydro-1H-pyrrolo[3,2,1-ij]quinolin-6-yl)pyrrolidin-2-one 213b

(m)

Light red foam.. Yield: 72%. FT-IR (neat, cm-1) 2959, 2928, 2860, 1717,1267; 1H

NMR (400 MHz, CDCl3) δH 7.47 (d, J=7.26 Hz, 2H), 7.40 (t, J= 7.5,7.5 Hz, 2H),

7.3-7.27 (m, 1H), 7.02 (d, J=7.1 Hz, 1H), 6.73 (t, J=8.9 Hz, 1H), 6.67 (t, J=7.4 Hz,

1H), 5.42 (dd, J= 6.4, 2.3 Hz, 1H), 5.31 (s, 1H), 3.80 (dd, J=11.1, 3.1 Hz, 1H),

3.84-3.74 (m, 1H), 3.35-3.19 (m, 2H), 2.60-2.42 (m, 3H), 2.25-2.09 (m, 2H), 2.09-

1,95 (m, 2H), 0.90 (dd, J=6.5, 3.8 Hz, 3H); 13C NMR (400 MHz, CDCl3) δC 175.3, 148.8, 128.7, 128.5,

128.24, 128.1, 127.0, 124,1, 123.9, 118.1, 116.8, 77.3, 77.1, 76.9, 57.3, 56.8, 53.45, 36.5, 43.7, 42.4,

37.2, 31.5. 18.3, 15.9; Anal Calcd for C22H24N2O; MS m/z: 332.2 (M+), 246 (100), 213, 196, 170, 142,

115, 91, 69, 41; MW: 332.170 g/mol; C 79.48% H 7.28% N 8.43% Found: C 79.51% H 7.31% N 9.37%.

59

1-(2,4-diphenyl-2,4,5,6-tetrahydro-1H-pyrrolo[3,2,1-ij]quinolin-6-yl)pyrrolidin-2-one 213c (M)

Light green oil. Yield: 53%. FT-IR (neat, cm-1) 2959, 2928, 2860, 1717,1267; 1H

NMR (400 MHz, CDCl3) δH 7.37-6.88 (m, 10 H), 6.79-6.65 (m, 3H), 5.66 (dd, J=

11.8, 5.4 Hz, 1H), 4.54 (t, J= 10.1 Hz, 1H), 4.15-4.08 (m, 1H), 3.69-3.58 (m,

1H), 3.43-3.18 (m, 2H), 3.01 (t, J=11.8 Hz, 1H), 2.60-2.40 (m, 2H), 2.31-2.22

(m, 1H), 2.12-1.92 (m, 2H), 1.28 (t, J=7.1 Hz, 1H); 13C NMR (400 MHz, CDCl3)

δC 175.9, 149.7,145.6, 141.0, 140.7, 128.5, 128.3, 127.8, 127.6, 127.3, 126.68, 124.1, 123.7, 118.3,

116.0, 65.4, 57.7, 47.5, 42.5, 37.6, 36.8, 31.4, 18.3; Anal Calcd for C27H26N2O; MS m/z: 394.2 (M+), 309

(100), 232, 205, 178, 154, 115, 91, 65, 41; MW: 394.205 g/mol; C 88.20% H 6.64% N 7.10%; Found C

82.23 % H 6.66% N 7.05%.

1-(2,4-diphenyl-2,4,5,6-tetrahydro-1H-pyrrolo[3,2,1-ij]quinolin-6-yl)pyrrolidin-2-one 213c (m)

Light green oil. Yield: 53%. FT-IR (neat, cm-1) 2959, 2928, 2860, 1717,1267; 1H

NMR (400 MHz, CDCl3) δH 7.37-6.88 (m, 10 H), 6.79-6.65 (m, 3H), 5.42 (dd, J=

6.4, 2.3 Hz, 1H), 4.54 (t, J= 10.1 Hz, 1H), 3.79 (dd, J=11.1, 3.1 Hz, 1H), 3.69-

3.58 (m, 1H), 3.43-3.18 (m, 2H), 3.01 (t, J=11.8 Hz, 1H), 2.60-2.40 (m, 2H),

2.31-2.22 (m, 1H), 2.12-1.92 (m, 2H), 1.28 (t, J=7.1 Hz, 1H); 13C NMR (400

MHz, CDCl3) δC 175.9, 149.7,145.6, 141.0, 140.7, 128.5, 128.3, 127.8, 127.6, 127.3, 126.68, 124.1,

123.7, 118.3, 116.0, 65.4, 57.7, 47.5, 42.5, 37.6, 36.8, 31.4, 18.3; Anal Calcd for C27H26N2O; MS m/z:

394.2 (M+), 309 (100), 232, 205, 178, 154, 115, 91, 65, 41; MW: 394.205 g/mol; C 88.20% H 6.64% N

7.10%; Found C 82.23 % H 6.66% N 7.05%.

60

1-(8-bromo-4-phenyl-2,4,5,6-tetrahydro-1H-pyrrolo[3,2,1-ij]quinolin-6-yl)pyrrolidin-2-one 213d

(M)

White foam. Yield: 50%. FT-IR (neat, cm-1) 2959, 2928, 2860, 1717,1267; 1H

NMR (400 MHz, CDCl3) δH 7.47-7.24 (m, 5H), 7.17-7.04 (m, 1H), 6.87 (s, 1H),

5.67 (dd, J= 11.4, 6.4 Hz, 1H), 4.01 (dd, J= 11.1, 2.5 Hz, 1H), 3.31 (dt, J= 14.0,

7.7 Hz, 4H), 3.08-2.69 (m, 2H), 2.61-2.37 (m, 2H), 2.26-1.91 (m, 4H); 13C NMR

(400 MHz, CDCl3) δC 175.9, 150.4, 141.2, 132.2, 129.9, 128.8, 127.9, 126.9,

126.8, 126.7, 118.9, 111.0, 77.4, 77.1, 76.8, 62.8, 53.0, 47.5, 42.3, 36.5, 31.3, 28.6, 18.2; Anal cald for

C21H21BrN2O; MS m/z: 396.1 (M+), 312 (100), 234, 204, 178, 154, 115, 91, 63, 41; MW: 397,122 g/mol;

C 63.48% H 5.33% Br 20.11% N 7.05%; Found C 63.50% H 5.31% Br 20.13% N 7.03%.

1-(8-bromo-4-phenyl-2,4,5,6-tetrahydro-1H-pyrrolo[3,2,1-ij]quinolin-6-yl)pyrrolidin-2-one 213d

(m)

White foam. Yield: 50%. FT-IR (neat, cm-1) 2959, 2928, 2860, 1717,1267; 1H

NMR (400 MHz, CDCl3) δH 7.47-7.24 (m, 5H), 7.17-7.04 (m, 1H), 6.87 (s, 1H),

5.42 (dd, J= 6.4, 2.3 Hz, 1H), 3.78 (dd, J=11.1, 3.1 Hz, 1H), 3.31 (dt, J= 14.0, 7.7

Hz, 4H), 3.08-2.69 (m, 2H), 2.61-2.37 (m, 2H), 2.26-1.91 (m, 4H); 13C NMR (400

MHz, CDCl3) δC 175.9, 150.4, 141.2, 132.2, 129.9, 128.8, 127.9, 126.9, 126.8,

126.7, 118.9, 111.0, 77.4, 77.1, 76.8, 62.8, 53.0, 47.5, 42.3, 36.5, 31.3, 28.6, 18.2; Anal cald for

C21H21BrN2O; MS m/z: 396.1 (M+), 312 (100), 234, 204, 178, 154, 115, 91, 63, 41; MW: 397,122 g/mol;

C 63.48% H 5.33% Br 20.11% N 7.05%; Found C 63.50% H 5.31% Br 20.13% N 7.03%.

1-(8-methoxy-4-phenyl-2,4,5,6-tetrahydro-1H-pyrrolo[3,2,1-ij]quinolin-6-yl)pyrrolidin-2-one 213e

(M)

Yellow oil. Yield: 64%. FT-IR (neat, cm-1) 2959, 2928, 2860, 1717,1267; 1H NMR

(400 MHz, CDCl3) δH 7.59-7.19 (m, 5H), 6.72 (s, 1H), 6.36 (s, 1H), 5.69 (dd, J=

10.4, 7.4, 1H), 3.91 (t, J= 10.4 Hz, 1H), 3.75 (s, 3H), 3.34 (dt, J= 13.5, 7.6 Hz,

3H), 2.97 (d, J= 4.5 Hz, 1H), 2.87-2.77 (m, 1H), 2.71 (s, 1H), 2.57-2.39 (m, 2H),

2.26-2.12 (m, 2H), 2.08-1.97 (m, 2H); 13C NMR (400 MHz, CDCl3) δC 175.8, 150.4, 148.8, 141.2, 132.2,

129.9, 128.7, 127.7, 127.2, 127.0, 111.3, 109.1, 56.2, 54.1, 47.9, 42.9, 36.7, 31.4, 29.1, 18.2; Anal Calcd

for C22H24N2O2; MS m/z: 348.2 (M+), 262 (100), 232, 204, 186, 160, 143, 115, 91, 69, 41; MW: 348,123

g/mol; C 75.83% H 6.94% N 8.04%; Found C 75.84% H 6.93% N 8.02%.

61

1-(8-methoxy-4-phenyl-2,4,5,6-tetrahydro-1H-pyrrolo[3,2,1-ij]quinolin-6-yl)pyrrolidin-2-one 213e

(m)

Yellow oil. Yield: 64%. FT-IR (neat, cm-1) 2959, 2928, 2860, 1717,1267; 1H NMR

(400 MHz, CDCl3) δH 7.59-7.19 (m, 5H), 6.72 (s, 1H), 6.36 (s, 1H), 5.42 (dd, J=

6.4, 2.3 Hz, 1H), 3.85 (dd, J=11.1, 3.1 Hz, 1H), 3.75 (s, 3H), 3.34 (dt, J= 13.5,

7.6 Hz, 3H), 2.97 (d, J= 4.5 Hz, 1H), 2.87-2.77 (m, 1H), 2.71 (s, 1H), 2.57-2.39

(m, 2H), 2.26-2.12 (m, 2H), 2.08-1.97 (m, 2H); 13C NMR (400 MHz, CDCl3) δC 175.8, 150.4, 148.8,

141.2, 132.2, 129.9, 128.7, 127.7, 127.2, 127.0, 111.3, 109.1, 56.2, 54.1, 47.9, 42.9, 36.7, 31.4, 29.1,

18.2; Anal Calcd for C22H24N2O2; MS m/z: 348.2 (M+), 262 (100), 232, 204, 186, 160, 143, 115, 91, 69,

41; MW: 348,123 g/mol; C 75.83% H 6.94% N 8.04%; Found C 75.84% H 6.93% N 8.02%.

1-(8-methoxy-2-methyl-4-phenyl-2,4,5,6-tetrahydro-1H-pyrrolo[3,2,1-ij]quinolin-6-yl)pyrrolidin-2-

one 213f (M)

Brown/red foam. Yield: 65%. FT-IR (neat, cm-1) 2959, 2928, 2860, 1717,1267;

1H NMR (400 MHz, CDCl3) δH 7.57-7.26 (m, 5H), 6.70 (s, 1H), 6.32 (s, 1H), 5.68

(dd, J=10.4, 6.8 Hz, 1H), 5.32 (s, 1H), 4.37 (d, J=12.2 Hz, 1H), 3.78 (d, J= 30.1

Hz, 3H), 3.35-3.11 (m,2H), 2.61-2.37 (m, 2H), 2.18 (d, J= 7.0 Hz, 2H), 2.14 (dd,

J= 37.3, 16.6 Hz, 2H), 2.08-1.95 (m, 2H), 1.09-0.67 (m,3H); 13C NMR (400 MHz,

CDCl3) δC 175.9, 129.9, 128.7, 128.5, 128.3, 127.7, 127.0, 111.8, 108.9, 57.8, 57.1, 56.2, 53.4, 47.9,

42.5, 37.3, 36.7, 31.4, 18.4, 18.3, 15.4; Anal. calcd for C23H26N2O2; MS m/z: 362.2 (100, M+), 276, 249,

218, 200, 172, 147, 115, 91, 69, 41; MW: 362,154 g/mol; C 76.21% H 7.23% N 7.73%; Found C 76.22%

H 7.25% N 7.70%.

62

1-(8-methoxy-2-methyl-4-phenyl-2,4,5,6-tetrahydro-1H-pyrrolo[3,2,1-ij]quinolin-6-yl)pyrrolidin-2-

one 213f (m)

Brown/red foam. Yield: 65%. FT-IR (neat, cm-1) 2959, 2928, 2860, 1717,1267;

1H NMR (400 MHz, CDCl3) δH 7.57-7.26 (m, 5H), 6.70 (s, 1H), 6.32 (s, 1H), 5.42

(dd, J= 6.4, 2.3 Hz, 1H), 5.32 (s, 1H), 4.03 (dd, J=11.1, 3.1 Hz, 1H), 3.78 (d, J=

30.1 Hz, 3H), 3.35-3.11 (m,2H), 2.61-2.37 (m, 2H), 2.18 (d, J= 7.0 Hz, 2H), 2.14

(dd, J= 37.3, 16.6 Hz, 2H), 2.08-1.95 (m, 2H), 1.09-0.67 (m,3H); 13C NMR (400

MHz, CDCl3) δC 175.9, 129.9, 128.7, 128.5, 128.3, 127.7, 127.0, 111.8, 108.9, 57.8, 57.1, 56.2, 53.4,

47.9, 42.5, 37.3, 36.7, 31.4, 18.4, 18.3, 15.4; Anal. calcd for C23H26N2O2; MS m/z: 362.2 (100, M+), 276,

249, 218, 200, 172, 147, 115, 91, 69, 41; MW: 362,154 g/mol; C 76.21% H 7.23% N 7.73%; Found C

76.22% H 7.25% N 7.70%.

63

5. Conclusions

The main purpose of this experimental thesis was to study some Lewis acid catalyzed Povarov

reactions for the synthesis of important and biologically active polycyclic tetrahydroquinolines

derivative. The results are quite interesting: on one hand, the choice of dienophile (DHP or N-vinyl-

2.pyrrolidone) had a great influence on the reaction outcome, on the other reaction conditions, such as

temperature and the presence/absence of solvent, played an important role. In the first part of the

work, a confirmation of the previous research group’s work has been obtained. In the second one,

among all the metal iodide tested, NaI was the one with better results in combination with CeCl3∙7H2O:

this is another experimental proof of the optimal activity of this catalytic couple. In the third and last

part of the work, the synthesis of new polycyclic tetrahydroquinoline derivatives starting from

substituted indolines gave good results. Each compound has been analyzed and characterized. For

the future research work, parallel to the synthesis of new compounds of this type, mew methods can

be studied in order to obtain the desired diastereomer.

64

6. References

Pericylic Reactions

1 Tietze, L. F.; Brasche, G.; Gericke, K. Domino Reactions in Organic Synthesis, 2006, Wiley-VCH;

2 Claisen, L. Chem. Ber., 1912, 45, 3157;

3 Reid, J. P.; McAdam, C. A.; Johnston, A. J. S.; Grayson, M. N.; Goodman, J. M.; Cook, M. J. J. Org.

Chem., 2015, 80, 1472-1498;

4 Nishibayashi, Y.; Uemura, S. Top. Curr. Chem., 2000, 208, 201-235;

5 Clayden, J.; Greeves, N.; Warren, S. Organic Chemistry, Second Edition, 2012, Oxford University

Press;

6 Ernst, R,; Winter, K. Tetrahedron Letters, 1965, 6, 1207-1212;

7 Schreiber, S. L.; Santini, C., J. Am. Chem. Soc., 1984, 106, 4038;

8 IUPAC, Compendium of Chemical Terminology, Gold Book, 2014;

9 Diels, O.; Alder, K. Justus Liebigs Ann. Chem., 1928, 460, 98–122;

10 Povarov, L. S., Russ. Chem. Rev., 1967, 36, 656;

11 Povarov, L. S.; Mikhailov, B.M.; Izv. Akad. Nauk, S.S.R. Ser. Khim., 1963, 953–956;

12 Povarov, L. S.; Grigos, V. I.; Mikhailov, B. M.; Izv. Akad. Nauk, S.S.R. Ser. Khim. 1963, 2039–2041;

13 Buonora, P.; Olsen, J. C.; Oh, T. Tetrahedron, 2001, 57, 6099–6138;

14 (a) Beifuss, U.; Ledderhose, S.; Ondrus, V. Arkivoc, 2005, 147–173; (b) Alves, M. J.; Azoia, N. G.;

Fortes, A. G. Tetrahedron, 2007, 63, 727–734; (c) Mayr, H.; Ofial, A. R.; Sauer, J.; Schmied, B. Eur. J.

Org. Chem., 2000, 2013–2020;

15 Ríos-Gutierrez, M.; Layeb, H.; Domingo, L. R. Tetrahedron, 2015, 71, 9339-9345;

16 Shi, F.; Xing, G.J.; Tao, Z.L.; Luo, S.W.; Tu, S.J.; Gong, L.Z. J. Org. Chem., 2012, 77, 6970-6979;

17 Hao, X.; Zhang, H.; Jacobsen, E. N. Nat Protoc., 2014, 9, 1860–1866;

18 Sridharan, V.; Avendano, C.; Menendez, J. C. Tetrahedron, 2007, 63, 673–681;

19 Park, K. H.; Joo, H. S.; Ahn, K. I.; Jun, K. Tetrahedron Lett., 1995, 36, 5943–5946;

20 Bohorquez, A. R. R.; Kouznetsov, V. V. Synlett, 2010, 6, 970;

21 Kouznetsov, V. V.; Forero, J. S. B.; Torres, D. F. A. Tetrahedron Lett., 2008, 49, 5855;

22 Astudillo, L. S.; Gutierrez, M.; Gaete, H.; Kouznetsov, V. V.; Melendez, C. M.; Palenzuela, J. A.;

Vallejos, G. Lett. Org. Chem., 2009, 6, 208;

65

23 Vicente-García, E.; Catti, F.; Ramon, R.; Lavilla, R. Org. Lett., 2010, 4, 860.

24 Dennis, M. K.; Burai, R.; Ramesh, C.; Petrie, W. K.; Alcon, S. N.; Nayak, T. K.; Bologa, C. G.; Leitao,

A.; Brailoiu, E.; Deliu, E.; Dun, N. J.; Sklar, L. A.; Hathaway, H. J.; Arterburn, J. B.; Oprea, T. I.; Prossnitz,

E. R.; Nat. Chem. Biol., 2009, 5, 421;

25 Burai, R.; Ramesh, C.; Shorty, M.; Curpan, R.; Bologa, C.; Sklar, L. A.; Oprea, T.; Prossnitz, E. R.;

Arterburn, J. B. Org. Biomol. Chem., 2010, 8, 2252;

26 Ramesh, C.; Nayak, T. K.; Burai, R.; Dennis, M. K.; Hathaway, H. J.; Sklar, L. A.; Prossnitz, E. R.;

Arterburn, J. B. J. Med. Chem., 2010, 53, 1004;

27 Kobayashi, S.; Jørgensen, K. A. Cycloaddition Reactions in Organic Synthesis, 2002, Wiley-VCH:

Weinheim;

28 Povarov, L. S.; Mikhailov, B. M. Ser. Khim. 1963, 953–956;

29 Ishitani, H.; Kobayashi, S. Tetrahedron Lett., 1996, 37, 7357–7360;

CeCl3, CeCl3∙7H2O and CeCl3∙7H2O/NaI

30 Yamamoto, H. Lewis acid reagents: A Pratical Approach, 1999, Oxford Press: New York;

31 Santelli, M.; Pons, J-M. Lewis Acids and Selectivity in Organic Synthesis, 1995, CRC Press: Boca

Ranton;

32 (a) Nair, V.; Deepthi, A. Chem. Rev., 2007, 107, 1862. (b) Mehdi, H.; Bodor, A.; Lantos, D.; Horva´th,

I. T.; De Vos, D. E.; Binnemans, K. J. Org. Chem., 2007, 72, 517;

33 Anwander, R.; Herrmann, W. A. Top. Curr. Chem., 1996, 179, 1;

34 Pearson, R. G. J. Chem. Educ., 1987, 64, 561;

35 Murad, E.; Hildenbrand, D. L. J. Chem. Phys., 1980, 73, 4005;

36 Tsuruta, H.; Yamaguchi, K.; Imamoto, T. Chem. Commun., 1999, 1703;

37 Imamoto, T.; Kusumoto, T.; Yokoyama, N. J. Chem. Soc. Chem. Commun., 1982, 1042;

38 Molander, G. A. In Chemistry of the Metal-Carbon Bond, Hartley, 1989, John Wiley: New York,;

39 Bartoli, G.; Marcantoni, E.; Sambri, L. Synlett, 2003, 5, 2101;

40 Imamoto, T.; Kusumoto, T.; Tawarayama, Y.; Sugiura, Y.; Mita, T.; Hatamaka, Y.; Yokoyama M. J.

Org. Chem., 1984, 49, 3904;

41 Shi, M.; Satoh, Y.; Masaki, Y. J. Chem. Soc., Perkin Trans, 1998, 1, 2547;

42 Nakanishi, K.; Goto, T.; Ito, S.; Natori, S.; Nozoe, S. Natural Product Chemistry; 1983, University

Press: Oxford;

66

43 (a) Dahlenburg, L.; Treffert, H.; Dannhauser, J.; Heinemann, F. W. Inorg. Chem. Acta, 2007, 360,

1474. (b) Ciganek, E. J. Org. Chem,. 1992, 57, 4521;

44 Reuman, M.; Beish, S.; Davis, J.; Batchelor, M. J.; Hutchings, M. C.; Moffat, D. F. C.; Connolly, P. J.;

Russell, R. K. J. Org. Chem., 2008, 73, 1121;

45 Moret, E.; Schlosser, M. Tetrahedron Lett., 1985, 26, 4423;

46 (a) Bartoli, G.; Dalpozzo, R.; De Nino, A.; Procopio, A.; Sambri, L.; Tagarelli, A. Tetrahedron Lett.

2001, 42, 8833. (b) Bartoli, G.; Bellucci, M. C.; Bosco, M.; Dalpozzo, R.; De Nino, A.; Sambri, L.;

Tagarelli, A. Eur. J. Org. Chem. 2000, 99. (c) Bartoli, G.; Bellucci, M. C.; Bosco, M.; Dalpozzo, R.; De

Nino, A.; Sambri, L.; Tagarelli, A. J. Org. Chem. 1998, 63, 9559;

47 Wang, C.; Xi, Z. Chem. Soc. Rev,. 2007, 36, 1395;

48 Johnstone, R. A. W.; Wilby, A. H.; Entwistle, I. D. Chem. Rev., 1985, 85, 129;

49 Luche, J.-L. J. Am. Chem. Soc., 1978, 100, 2226;

50 Iacazio, G. Chem. Phys. Lipids, 2003, 125, 115;

51 Yamaguchi, J.; Seiple, I. B.; Young, I. S.; O’Malley, D. P.; Mane, M.; Baran, P. S. Angew. Chem. Int.

Ed., 2008, 47, 3578;

52 Williams, M. Adenosine and Adenosine Receptors, 1990, Humana Press: Clifton;

53 Martin, P. L.; Wysocki, R. J.; Barrett, R. J. Jr.; May, J. M.; Linden, J. J, Pharmacol. Exp. Ther., 1996,

276, 490;

54 Jin, C.; Burgess, J. P.; Rehder, K. S.; Brine, G. A. Synthesis, 2007, 219;

55 Peck, J. V.; Wysock, R. J.; Uwaydah, I. M.; Cusak, N. J. U.S. Patent No. 5670501, 1997; Chem. Abstr.

1996, 125, 58207;

56 Khodaei, M. M.; Khosropour, A. R.; Kookhazadeh, M. Russ. J. Org. Chem., 2005, 41, 1445;

57 Schinzer, D. Selective in Leiws Acid Promoted Reactions; 1989, Kluwer Academic Publisher:

Dordrect;

58 Iimura, S.; Manabe, K.; Kobayashi, S. Tetrahedron, 2004, 60, 7673;

59 (a) Fukuzawa, S.; Tsurute, T.; Fujinami, T.; Sakai, S.; J. Chem. Soc., Perkin Trans., 1987, 1473. (b)

Fukuzawa, S.; Fujinami, T.; Sakai, S. J. Chem. Soc., Chem. Commun., 1985, 777;

60 Myers, E.; Butts, C. P.; Aggarwal, V. K. Chem. Commun. 2006, 4434;

61 Cappa, A.; Marcantoni, E.; Torregiani, E.; Bartoli, G.; Bellucci, M. C.; Bosco, M.; Sambri, L. J. Org.

Chem., 1999, 64, 5696.

62 a) Denmark, S. E.; Wynn, T.; Bentner, G. L. J. Am. Chem. Soc. 2002, 124, 13405-13407. (b) Denmark,

S. E.; Wynn, T. J. Am. Chem. Soc. 2001, 123, 6199-6200;

67

63 Bartoli, G.; Fernández-Bolaños, J. G.; Di Antonio, G.; Foglia, G.; Giuli, S.; Gunnella, R.; Mancinelli,

M.; Marcantoni, E.; Paoletti, M. J. Org. Chem., 2007, 72, 6029–6036;

64 Evans, W. J.; Feldman, J. D.; Ziller, J. W. J. Am. Chem. Soc., 1996, 118, 4581;

65 Narayau, S.; Muldoon, J.; Finn, M. G.; Fokin, V. V.; Kolb, H. C.; Sharpless, K. B. Angew. Chem., Int.

Ed., 2005, 44, 3275;

66 (a) Schreiber, S. L. Science, 2000, 287, 1964. (b) Posner, G. H. Chem. Rev. 1986, 86, 831;

67 (a) Dax, S. L.; Nally, J.-J. M. C.; Youngman, M. A. Curr. Med.Chem., 1999, 6, 255 (b) Gallop, M. A.;

Banett, R. W.; Dower, W. J.; Fodor, S. P. A. J. Med. Chem,. 1994, 37, 1385;

68 Yadav, L. D. S.; Kapoor, R. Tetrahedron Lett., 2008, 49, 4840;

69 Teshima, Y.; Shiya, K.; Shimazu, A.; Furihata, K.; Chul, H. S.; Hayakawa, Y.; Nagi, K.; Seto, H.J.

Antibiot., 1991, 44, 685;

70 Cox, R. J.; O’Hagan, D. J. Chem. Soc., Perkin Trans. 1, 1991, 2537;

71 Duval, D.; Geribaldi, S. In The Chemistry of Enones Part 1, 1989, Interscience: New York;

72 Talalay, P.; De long, M. J.; Prochaska, H. J. Proc. Natl. Acad. Sci. U.S.A., 1988, 85, 8261;

73 Christoffers, J. J. Chem. Soc., Perkin Trans. 1, 1997, 3141;

74 Bartoli, G.; Beleggia, R.; Giuli, S.; Giuliani, A.; Marcantoni, E.; Massaccesi, M.; Paoletti, M.

Tetrahedron Lett,. 2006, 47, 6501;

75 Tietze, L. F.; Beifuss, U. Comprehensive Organic Synthesis, 1991, Pergamon Press: Oxford;

76 Tanaka, M.; Oota, O.; Miramatsu, H.; Fujiwara, K. Bull. Chem. Soc. Jpn., 1988, 61, 2473;

77 Bartoli, G.; Bosco, M.; Carlone, A.; Dalpozzo, R.; Galzerano, P.; Melchiorre, P.; Sambri, L.

Tetrahedron Lett., 2008, 49, 2555;

78 Yeh, M.-C. P.; Yeh, W.-J.; Tu, L.-H. Wu J.-R., Tetrahedron, 2006, 62, 7466;

79 Marcantoni, E.; Massaccesi, M.; Petrini, M.; Bartoli, G.; Bellucci, M. C.; Bosco, M.; Sambri, L. J. Org.

Chem., 2000, 65, 4553;

80 Bartoli, G.; Bosco, M.; Dalpozzo, R.; Giuliani, A.; Marcantoni, E.; Mecozzi, T.; Sambri, L.; Torregiani,

E. J. Org. Chem., 2002, 67, 9111;

Tetrahydroquinolines: applications and synthesis

81 Katritzky, A. R.; Rachwal, S.; Rachwal, B. Tetrahedron, 1996, 52, 15031;

82 (a) Kim, W.-G.; Kim, J.-P.; Kim, C.-J.; Lee, K.-H.; Yoo, I.-D.; J. Antibiot., 1996, 49, 20. (b) Kim, W.-G.;

Kim, J.-P.; Yoo, I.-D. J. Antibiot., 1996, 49, 26;

68

83 Kimura, Y.; Kusano, M.; Koshino, H.; Uzawa, J.; Fujioka, S.; Tani, K. Tetrahedron Lett., 1996, 37,

4961.

84 Kusano, M.; Koshino, H.; Uzawa, J.; Fujioka, S.; Kawano, T.; Kimura, Y. Biosci., Biotechnol.,

Biochem., 2000, 64, 2559;

85 Schmeda-Hirschmann, G.; Hormazabal, E.; Astudillo, L.; Rodriguez, J.; Theoduloz, C. World J.

Microbiol. Biotechnol., 2005, 21, 27;

86 Yamada, O.; Ogasawara, K. Tetrahedron Lett., 1998, 39, 7747;

87 Gudmundsson, K. S.; Sebahar, P. R.; Richardson, L. D.; Miller, J. F.; Turner, E. M.; Catalano, J. G.;

Spaltenstein, A.; Lawrence, W.; Thomson, M.; Jenkinson, S. Bioorg. Med. Chem. Lett., 2009, 19, 5048;

88 Catalano, J. G.; Gudmundsson, K. S.; Svolto, A.; Boggs, S. D.; Miller, J. F.; Spaltenstein, A.; Thomson,

M.; Wheelan, P.; Minick, D. J.; Phelps, D. P.; Jenkinson, S. Bioorg. Med. Chem. Lett., 2010, 20, 2186;

89 Miller, J. F.; Turner, E. M.; Gudmundsson, K. S.; Jenkinson, S.; Spaltenstein, A.; Thomson, M.;

Wheelan, P. Bioorg. Med. Chem. Lett., 2010, 20, 2125;

90 Skerlj, R. T.; Bridger, G. J.; Kaller, A.; McEachern, E. J.; Crawford, J. B.; Zhou, Y.; Atsma, B.; Langille,

J.; Nan, S.; Veale, D.; Wilson, T.; Hartwig, C.; Hatse, S.; Princen, K.; De Clercq, E.; Schols, D. J. Med.

Chem., 2010, 53, 3376;

91 Ramesh, E.; Manian, R. D. R. S.; Raghunathan, R.; Sainath, S.; Raghunathan, M.; Bioorg. Med.

Chem., 2009, 17, 660;

92 Jarvest, R. L.; Berge, J. M.; Berry, V.; Boyd, H. F.; Brown, M. J.; Elder, J. S.; Forrest, A. K.; Fosberry,

A. P.; Gentry, D. R.; Hibbs, M. J.; Jaworski, D. D.; O’Hanlon, P. J.; Pope, A. J.; Rittenhouse, S.;

Sheppard, R. J.; Slater-Radosti, C.; Worby, A. J. Med. Chem., 2002, 45, 1959;.

93 Jarvest, R. L.; Armstrong, S. A.; Berge, J. M.; Brown, P.; Elder, J. S.; Brown, M. J.; Copley, R. C. B.;

Forrest, A. K.; Hamprecht, D. W.; O’Hanlon, P. J.; Rittenhouse, S.; Witty, D. R. Bioorg. Med. Chem.

Lett., 2004, 14, 3937;

94 Bendale, P.; Olepu, S.; Suryadevara, P. K.; Bulbule, V.; Rivas, K.; Nallan, L.; Smart, B.; Yokoyama,

K.; Ankala, S.; Pendyala, P. R.; Floyd, D.; Lombardo, L. J.; Williams, D. K.; Buckner, F. S.; Chakrabarthi,

D.; Verlinde, C. L. M. J.; Voorhis, W. C. V.; Gelb, M. H. J. Med. Chem., 2007, 50, 4585;

95 Eastman, R. T.; White, J.; Hucke, O.; Yokoyama, K.; Verlinde, C. L. M. J.; Hast, M. A.; Beese, L. S.;

Gelb, M. H.; Rathod, P. K; Voorhis, W. C. V. Mol. Biochem. Parasitol., 2007, 152, 66;

96 Gupta, M. K.; Prabhakar Y. S. Eur. J. Med. Chem., 2008, 43, 2751;.

97 Kaur, K.; Jain, M.; Reddy, R. P.; Jain, R. Eur. J. Med. Chem., 2010, 45, 3245;

98 Pagliero, R. J.; Lusvarghi, S.; Pierini, A. B.; Brun, R.; Mazzieri, M. R. Bioorg. Med. Chem., 2010, 18,

142;

69

99 Maillard, M. C.; Perlman, M. E.; Amitay, O.; Baxter, D.; Berlove, D.; Connaughton, S.; Fischer, J. B.;

Guo, J. Q.; Hu, L.-Y.; McBurney, R. N.; Nagy, P. I.; Subbarao, K.; Yost, E. A.; Zhang, L.; Durant, G. J.

J. Med. Chem., 1998, 41, 3048;

100 Fukumoto, S.; Imamiya, E.; Kusumoto, K.; Fujiwara, S.; Watanabe, T.; Shiraishi, M.; J. Med. Chem.,

2002, 45, 3009;.

101 Gore, V. K.; Ma, V. V.; Yin, R.; Ligutti, J.; Immke, D.; Doherty, E. M.; Norman, M. H. Bioorg. Med.

Chem. Lett., 2010, 20, 3573;.

102 Faghih, R.; Gopalakrishnan, M.; Briggs, C. A. J. Med. Chem., 2008, 51, 701;.

103 Romero,,A. G.; Darlington, W. H.; McMillan, M. W. J. Org. Chem., 1997, 62, 6582;

104 Ohta, M.; Suzuki, T.; Ohmori, J.; Koide, T.; Matsuhisa, A.; Furuya, T.; Miyata, K.; Yanagisawa, I.

Chem. Pharm. Bull., 1996, 44, 1000;

105 Wallace, O. B.; Lauwers, K. S.; Jones, S. A.; Dodge, J. A. Bioorg. Med. Chem. Lett., 2003, 13, 1907

106 Thevis, M.; Kohler, M.; Schanzer, W. Drug Discovery Today, 2008, 13, 59;

107 Chen, R.; Yang, X.; Tian, H.; Sun, L. J. Photochem. Photobiol. A, 2007, 189, 295;.

108 Pullmann, T.; Engendahl, B.; Zhang, Z.; Holscher, M.; Zanotti-Gerosa, A.; Dyke, A.; Francio, G.;

Leitner, W. Chem.—Eur. J., 2010, 16, 7517;.

109 Liu, W.-B.; He, H.; Dai, L.-X.; You, S.-L. Synthesis, 2009, 12, 2076;

110 Fujita, K.; Yamamoto, K.; Yamaguchi, R. Org. Lett., 2002, 4, 2691;

111 Fujita, K.; Takahashi, Y.; Owaki, M.; Yamamoto, K.; Yamaguchi, R. Org. Lett., 2004, 6, 2785;.

112 Fujita, K.; Furukawa, S.; Yamaguchi, R. J. Organomet. Chem., 2002, 649, 289;

113 Youn, S. W.; Song, J.-H.; Jung, D.-I. J. Org. Chem., 2008, 73, 5658;

114 Davies, S. G.; Mujtaba, N., Roberts, P. M.; Smith, A. D.; Thomson, J. E. Org. Lett., 2009, 9, 1959;

115 Sridharan, V.; Suryavanshi, P. A; Menendez, J. C. Chem. Rev., 2011, 111, 7157–7259;

116 (a) Takeda, Y.; Nakabayashi, T.; Shirai, A.; Fukumoto, D.; Kiguchi, T.; Naito, T. Tetrahedron Lett.,

2004, 45, 3481; (b) Miyata, O.; Shirai, A.; Yoshino, S.; Nakabayashi, T.; Takeda, Y.; Kiguchi, T.;

Fukumoto, D.; Ueda, M.; Naito, T. Tetrahedron, 2007, 63, 10092;

117 Uchikawa, W.; Matsuno, C.; Okamoto, S. Tetrahedron Lett., 2004, 45, 9037;

118 Saavedra, L. A.; Vallejos, G.; Kouznetsov, V. V.; Gutierrez, M.; Gomez, C. M. M.; Mendez, L. Y. V.;

Jaimes, J. H. B. Synthesis, 2010, 4, 593;

119 (a) Kouznetsov, V. V.; Zubkov, F. I.; Nikitina, E. V.; Duarte, L. D. A. Tetrahedron Lett., 2004, 45, 1981;

(b) Zubkov, F. I.; Nikitina, E. V.; Kouznetsov, V. V.; Duarte, L. D. A. Eur. J. Org. Chem., 2004, 24, 5064;

70

120 Togo, H.; Hoshina, Y.; Muraki, T.; Nakayama, H.; Yokoyama, M. J. Org. Chem., 1998, 63, 5193;

121 Wojciechowski, K. Eur. J. Org. Chem., 2001, 19, 3587;

122 Steinhagen, H.; Corey, E. J. Angew. Chem., Int. Ed., 1999, 38, 1928;

123 Zhu, Z.-B.; Shao, L.-X.; Shi, M. Eur. J. Org. Chem., 2009, 15, 2576;

124 Khadem, S.; Udachin, K. A.; Enright, G. D.; Prakesch, M.; Arya, P. Tetrahedron Lett., 2009, 50, 6661;

125 Liu, H.-M.; Liu, F.-W.; Zou, D.-P.; Dai, G.-F. Bioorg. Med. Chem. Lett., 2005, 15, 1821;

Result and discussion

126 Jha, A.; -Yi Chou, T.; Al Jaroudi, Z.; Ellis, B. D.; Cameron, T. S. Beilstein J. Org. Chem. 2014, 10,

848–857;

127 Tarantin, A.V.; Glushkov, V.A.; Mayorova, O. A.; Shcherbininab, I. A.; Tolstikova, A. G. Mendeleev

Commun., 2008, 18, 188–190;

128 Jiang, X.; Wang, C.; Wei, Y.; Xue, D.; Liu, Z.; Xiao, J. Chemistry - A European Journal, 2014 , 20,

58–63;

Characterization of compounds

129 Abu, T. K.;.Musawwer K. Tetrahedron Lett., 2011, 52, 4539–4542;

130 Talwar, D.; Li, H.; Durham,E.; Xiao, J. Chemistry- A European Journal, 2015, 21, 5370-5379;

131 Compain, G.; Bonneau, C.; Martin.Mingot, A.; Thibaudeau, S. J. Org. Chem., 2013, 78, 4463-4472;

132 Ding, F.; Zhang, Y.; Zhao, R.; Jiang, Y.; Bao, R. L.-Y.; Lin, K.; Shi, L. Chem. Comm., 2017, 53, 9262-

9234;

132 Maier, A. F. G.; Tussing, S.; Schneider, T.; Florke, U.; Qu, Z.-W.; Grimme, S.; Paradies, J. Angew.

Chem,- Int. Ed., 2016, 55, 12219-12223; Maier, A. F. G.; Tussing, S.; Schneider, T.; Florke, U.; Qu, Z.-

W.; Grimme, S.; Paradies, J. Angew. Chem., 2016, 128, 12407-12411,5;

134 Gong, T.-J.; Cheng, W.-M.; Su, W.; Xiao, B.; Fu, Y. Tethaedron Lett., 2014, 55, 1859-1862;

135 Min, C.; Mittal, N.; Sun, D. X.; Seidel, D. Angew. Chem.- Int. Ed., 2013, 52, 14084-14088; Min, C.;

Mittal, N.; Sun, D. X.; Seidel, D Angew. Chem., 2013, 125, 14334-14338,5.

71

Annexes

In the Annexes section, 1H and 13C spetra of un precendented products (213b-f) are reported. The

values are reported in Characterization of Compounds section. Other experimental data are

reported in Characterization of Compounds section.

213b

1H

72

13C

213c

1H

73

13C

213d

1H

74

13C

213e

1H

75

13C

213f

1H

76

13C