copyright by kenneth stanley matthews 2005

300
Copyright by Kenneth Stanley Matthews 2005

Upload: others

Post on 31-Dec-2021

3 views

Category:

Documents


0 download

TRANSCRIPT

Copyright

by

Kenneth Stanley Matthews

2005

The Dissertation Committee for Kenneth Stanley Matthews Certifies that this is the

approved version of the following dissertation:

THE TOTAL SYNTHESIS OF (±)-RENIERAMYCIN G AND

STUDIES TOWARD THE SYNTHESIS OF (±)-LEMONOMYCIN

AND (±)-SAFRAMYCIN B

Committee:

Philip D. Magnus

Stephen F. Martin

Eric V. Anslyn

Sean M. Kerwin

Hung-wen Liu

THE TOTAL SYNTHESIS OF (±)-RENIERAMYCIN G AND

STUDIES TOWARD THE SYNTHESIS OF (±)-LEMONOMYCIN

AND (±)-SAFRAMYCIN B

by

Kenneth Stanley Matthews, B.S.

Dissertation

Presented to the Faculty of the Graduate School of

The University of Texas at Austin

in Partial Fulfillment

of the Requirements

for the Degree of

Doctor of Philosophy

The University of Texas at Austin

August, 2005

Dedication

To my father, mother, and brother, for all your love and support.

v

Acknowledgements

Ms. Robin Spalty: you got the ball rolling and I am forever grateful that you

taught chemistry long enough to teach its wonders.

Dr. Christopher Creighton: thank you for giving me a chance, getting me into

organic chemistry, and all the advice that has taken me so far.

Dr. Jamie Reuter: your patience and generosity were exactly what I needed, and

your passionate management style still can not be beat.

Dr. Konrad Fiechtinger: you taught me so much chemistry I could not begin to

thank you. You are a great boss and an inspiring chemist.

Prof. Murray Goodman: you let me in your group with so little experience and I

will forever thank you for giving me that first chance.

Dr. Gary Snyder: the only organic chemistry teacher that was clear, concise, and

thorough. Thank you for giving the best foundation to be a researcher.

Prof. John C. Wheeler: your excitement for chemistry and demand of students

helped me focus and achieve far above my own expectations.

Dr. Luca Araldi: your training has allowed me to understand so much of the

chemistry process and your patience and happy spirit were the best to work with.

Dr. Jean Cui: your drive to do great things is unmatched. I am very grateful to

have learned not only chemistry from you but how to work in the midst of circumstance.

vi

Dr. Odile Levy: a truly great boss, you were so very keen on keeping me focused,

and your leadership showed me how it all gets done.

Dr. Scott Kemp: I can not thank you enough for teaching me the ways of the Jedi.

Without your insight and vigilant teaching I would have been lost in graduate school.

Mr. Maxwell Lawrence: you gave me so much advice on how to be successful in

chemistry and taught me the essential lesson of doing it right the first time.

Dr. Jennifer Kreisburg: you helped me get into a great research group and gave

me great advice throughout graduate school. I am very grateful.

Prof. Cyril Olivier: I owe you for all the helpful discussions and advice. Your

knowledge and passion for chemistry were an inspiration.

Dr. Trevor Rainey: anyone who can stand five years sitting next to me deserves

praise, and thank you for the countless discussions and sharing your knowledge.

Dr. Rachel Turnbull: thank you for all your writing advice and time editing. I

would have been lost without it.

Mr. Vincent Lynch: your skills as an X-ray crystallographer saved the day many a

time, and I thank you for all the helpful discussions.

Mr. Steve Sorey and Dr. Ben Shoulders: thank you for the countless discussions

about NMR interpretation and for dealing with my nagging.

Prof. Philip D. Magnus: you gave me a chance in your group and a wonderful

project. I enjoyed all the guidance and support that you have provided.

vii

THE TOTAL SYNTHESIS OF (±)-RENIERAMYCIN G AND

STUDIES TOWARD THE SYNTHESIS OF (±)-LEMONOMYCIN

AND (±)-SAFRAMYCIN B

Publication No._____________

Kenneth Stanley Matthews, Ph.D.

The University of Texas at Austin, 2005

Supervisor: Philip Douglas Magnus

Herein is described our synthetic approach to the tetrahydroisoquinoline alkaloids.

The first chapter describes relevant background related to the biological significance of

these alkaloids. The analysis of various syntheses of saframycins, renieramycins, and

lemonomycin is also discussed. Chapter 2 describes the development of a new synthesis

of 1,3-cis-substituted tetrahydroisoquinolines and a novel lactam formation in our

approach to (±)-saframycin B. Chapter 3 applies the new tetrahydroisoquinoline

formation toward the synthesis of (±)-lemonomycin. The bicyclo[3.2.1] system is

constructed from an intramolecular N-acyliminium cyclization and leads to the synthesis

(±)-lemonomycinone amide. Chapter 4 reports the total synthesis of (±)-renieramycin G

from an advanced intermediate used in our approach to (±)-lemonomycin, demonstrating

a divergent approach to the tetrahydroisoquinoline alkaloids. Chapter 5 contains the

experimental details and characterization data for all new reported compounds.

viii

Table of Contents

Chapter 1: Tetrahydroisoquinoline Alkaloids..........................................................1 1.0. Introduction ................................................................................................1 1.1. Biological Activity .....................................................................................4 1.2. Mechanism of Action .................................................................................5 1.3. Biosynthesis................................................................................................7 1.4. Synthetic Approaches to Tetrahydroisoquinoline Alkaloids......................9

1.4.1. The Saframycins..............................................................................10 1.4.1.1. Fukuyama’s Total Synthesis of (±)-Saframycin B.................10 1.4.1.2. Kubo’s Total Synthesis of (±)-Saframycin B.........................13 1.4.1.3. Fukuyama’s Total Synthesis of (±)-Saframycin A.................15 1.4.1.4. William’s Studies Toward the Saframycins...........................17 1.4.1.5. Corey’s Enantioselective Synthesis of (–)-Saframycin A......19

1.4.2. The Renieramycins..........................................................................21 1.4.2.1. Fukuyama’s Synthesis of (±)-Renieramycin A ......................21 1.4.2.2. Danishefsky’s Synthesis of Cribrostatin IV ...........................23

1.4.3. Lemonomycin..................................................................................28 1.4.3.1. Stoltz’s Synthesis of (–)-Lemonomycin.................................28 1.4.3.2. Fukuyama’s Studies Toward (–)-Lemonomycin....................31

1.5. Retrosynthetic Analysis ............................................................................35 1.6. Conclusion ................................................................................................37 1.7. References ................................................................................................38

Chapter 2: Toward the Synthesis of Saframycins B ..............................................42 2.0. Introduction ..............................................................................................42 2.1. Background: Enantioselective Alkylation of Isoquinolines.....................43 2.2. Results and Discussion.............................................................................51

2.2.1. Investigation of the Enantioselective Alkylation of Isoquionline ...51 2.2.2. Formation of 1,3-cis-Substituted Tetrahydroisoquinoline ..............55 2.2.3. 3-(Aminomethyl)-Isoquinoline .......................................................59

ix

2.2.4. Oxygen Analogue............................................................................67 2.2.5. Nitrogen Analogue for Pummerer Cyclization ...............................72 2.2.6. Formation of the Real System.........................................................76 2.2.7. Alkylation, Reduction, and Cyclization of the Real System...........80

2.3. Conclusion ................................................................................................85 2.4. References ................................................................................................87

Chapter 3: Toward the Synthesis of Lemonomycin ..............................................89 3.0. Introduction ..............................................................................................89 3.1. Results and Discussion.............................................................................91

3.1.1. Alkylation with Benzyloxymethyllithium.......................................91 3.1.2. Formation 1,3-cis-Substituted Tetrahydroisoquinoline...................97 3.1.3. Amide Coupling ............................................................................103 3.1.4. Stereoselective Incorporation of C14-C15 ....................................107 3.1.5. Formation of the Bicyclo[3.2.1] System .......................................117

3.2. Conclusion ..............................................................................................123 3.3. References ..............................................................................................125

Chapter 4: The Total Synthesis of (±)-Renieramycin G......................................126 4.0. Introduction ............................................................................................126 4.1. Results and Discussion...........................................................................128

4.1.1. Benzyl Chloride Formation ...........................................................128 4.1.2. Formation the Bicyclo[3.3.1] System............................................131 4.1.3. Completion of the Synthesis..........................................................137

4.2. Conclusion ..............................................................................................142 4.3. References ..............................................................................................143

Chapter 5: Experimentals.....................................................................................144 5.0. General Information ...............................................................................144 5.1. Experimental Conditions and Characterization......................................145 5.2. References ..............................................................................................244

x

Appendix A: X-Ray Data for the Chloride 198 .................................................252

Appendix B: X-Ray Data for the Bicyclo[3.3.1] 213 ........................................256

Appendix C: X-Ray Data for the Lactam 229 ...................................................260

Appendix D: X-Ray Data for the Amino Alcohol 283 ......................................264

Appendix E: X-Ray Data for Thioaminal 300....................................................268

Appendix F: X-Ray Data for the Allylated Product 301 ...................................273

Appendix G: X-Ray Data for Oxime 314 ..........................................................278

Appendix H: X-Ray Data for the Diol 334 ........................................................283

Abbreviations ......................................................................................................288

Vita .....................................................................................................................290

1

Saframycins A (1a) R1=CN,R2=H B (1b) R1=R2=H C (1c) R1=H,R2=OMe G (1d) R1=CN,R2=OH S (1e) R1=OH,R2=H

Renieramycins

A (2a) R1=R2=H,R3=OH B (2b) R1=R2=H,R3=OMe C (2c) R1=R2=O,R3=OH D (2d) R1=R2=O,R3=OEt E (2e) R1=OH,R2=R3=H F (2f) R1=OH,R2=H,R3=OMe G (2g) R1=R2=O,R3=H

Safracins A (3a) R1=R2=H B (3b) R1=H,R2=OH (3c) R1=CN,R2=H

Ecteinascidin

Et743 (4a) R1=OH,R2=Me Et729 (4b) R1=OH,R2=H Et745 (4c) R1=H,R2=Me Et770 (4d) R1=CN,R2=Me

Chapter 1: Tetrahydroisoquinoline Alkaloids

1.0. INTRODUCTION

The tetrahydroisoquinoline alkaloids are a broad family of natural products with

nearly 60 members isolated and more than a hundred derivatives. Members of this family

include those with a bis-tetrahydroisoquinoline core; the saframycins, renieramycins,

safracins, and ecteinascidins (Figure 1.01), and those with a mono-tetrahydroisoquinoline

core; naphthyridinomycin, cyanocycline, the bioxalomycins, the quinocarcins,

tetrazomine, and lemonomycin (Figure 1.02).1-10

Figure 1.01. Bis-Tetrahydroisoquinolines

NN

Me

O

OOMe

O

O

Me

MeO

HH

H

NH

H

1

3

OO

Me

Me

R2

R1

NN

Me

O

OOMe

O

O

Me

MeO

HH

H

O

H

1

3

OMe

MeR3

R1

H

MeR2

NN

Me

R2

HOOMe

O

O

Me

MeO

HH

H

NH

H

1

3

ONH2

Me

Me

R1

NN

MeHOOMe

OAcMe

HH

HR1

OHS

NHMeO

HO

O

13 R2

OO

2

naphthridinomycin (5) R1=OH,R2=Me,R3=Mecyanocycline A (6a) R1=CN,R2=Me,R3=Me

B (6b) R1=CN,R2=Me,R3=H F (6c) R1=CN,R2=H,R3=Me

bioxalomycin β1 (7a) R=H bioxalomycin β2 (7b) R=Me

quinocarcin (8) tetrazomine (9) lemonomycin (10)

Figure 1.02. Mono-Tetrahydroisoquinolines

The first tetrahydroisoquinoline alkaloid structure elucidated was in 1974 with the

X-ray analysis of the unstable ruby red crystal of naphthyridinomycin.11 This compound

was isolated from an unknown Streptomycete strain, Streptomyces lusitanus AYB-1026,

found in an Easter Island soil sample.12 Within the next 10 years cyanocyclines,13,14

quinocarcin,15 safracins16 and numerous saframcyins9 were isolated and characterized

from strains of Streptomyces. During the 1990’s further fermentation studies led to the

discovery of tetrazomine17 and bioxalomycins.18 One of the most recent additions to the

family was lemonomycin which was first isolated in 196019 from Streptomyces candidus

however the structure was not elucidated until 2000.20 Lemonomycin is the only

tetrahydroisoquinoline alkaloid to possess a glycidic linkage in the C1 side-chain and the

rare aldehyde hydrate functionality.

N

OMe

HH

HO

H

CO2H

N3

1

Me

H

NNR

O

O

Me

MeO

H

H

1

3

N

O

HH

OH

N

OMe

HH

HO

HN3

1

Me

HNH

O

NH

OH

H

H

OH

NNR3

O

O

Me

R2O

H

R1

OH

H

1

3

N

O

HH

N

O

Me

MeO

HH

HOH

O

H

OOH

NMe2

HOOH

NH3

1

O

3

Tetrahydroisoquinoline alkaloids are also found as marine natural products. All of

the renieramycins were isolated from marine sources: A-F from the bright blue sea

sponge Reniera sp. found in Mexico,21,22 G from the Fijian sponge Xestospongia

caycedoi23 and most recently H and I from the sponge Haliclona cribricutis found off the

coast of India.24 The renieramycins possess a similar bis-isoquinolinequinone core to the

saframycins. The main difference is an angelate C1 side-chain in place of a pyruvamide.

The most structurally complex tetrahydroisoquinoline alkaloids, the ecteinascidins which

are actually tris-tetrahydroisoquinolines, were isolated from a Caribbean tunicate

Ecteinascidia turbinata in 1990.25,26

4

1.1. BIOLOGICAL ACTIVITY

The tetrahydroisoquinoline alkaloids possess a range of biological activity as

potent cytotoxic agents. This includes antitumor, antibiotic and antimicrobial activity

through the inhibition of RNA, DNA, and protein synthesis. A recent review by

Williams1 gives a comprehensive account of the biology of tetrahydroisoquinoline

antitumor antibiotics. While many of the tetrahydroisoquinoline alkaloids have well

investigated in vitro activity, saframycin and the ecteinascidins have the most well

studied in vivo biological activity in this family. However none of these alkaloids or

derivatives are currently on the market as therapeutics.

Ecteinascidin 743 (Et743, 4a) is currently in Phase III clinical trials for the

treatment of ovarian, endometrial and breast cancer.27 The antiproliferative activity of

Et743 is greater than that of taxol, mitomycin C and cisplatin (Table 1.01). Furthermore,

the unique mode of action of Et743 is thought to be responsible for its high level of

activity in advanced sarcomas that had relapsed or were resistant to conventional therapy.

cell line drug

P388 leukemia

L1210 leukemia

A549 lung cancer

Et743 0.34 nM28 0.66 nM28 0.26 nM28

saframycin A N/A N/A 133 nM29

taxol 16.9 nM30 41.0 nM31 2.0 nM32

mitomycin C 14.0 nM30 4.2 μM33 0.23 μM34

cisplatin 0.11 μM35 7.0 μM35 18.1 μM36

Table 1.01. Comparison of Inhibitory Concentrations (IC50)

5

1.2. MECHANISM OF ACTION1,9,37

Multiple theories have been proposed for the mechanism of action leading to the

observed biological activity of the tetrahydroisoquinoline alkaloids. The current accepted

theory for Et743 involves binding to the minor groove of DNA through hydrogen

bonding interactions between the A and E-ring of Et743 (4a) and three base pairs, leading

to unique sequence specificities (Scheme 1.01). Protonation at N12 and subsequent acid

catalyzed dehydration of carbinolamine 11 leads to formation of iminium 12. The

exocyclic 2-amino group of guanine 13 attacks the C21 iminium to form a covalently

bonded adduct 15. The initial Et743/DNA complex is formed reversibly under non-

denaturing conditions, migrating from a non-favored binding sequence to the favored

DNA target site (5′-AGC). The Et743/DNA covalent adduct 15 has been shown to form

reversibly with DNA denaturization.38

NN

MeHOOMe

AcO

O

Me

O

HH

HOH

OHS

NHMeO

HO

O

13 MeA

E18

2113

11

87

6

5

14

15

16

17

B

H

GF

C

NN

MeHOOMe

OH

MeH

5´-AGC

NN

MeHOOMe

Me

HN

N

N

NH2N

O

5´-AGC

H

OH

NN

MeHOOMe

Me

HN

N

N

NHN

O

5´-AGC

H

OHH

NN

MeHOOMe

Me

HN

N

N

NHN

O

5´-AGC

H

Scheme 1.01. Et743 DNA Alkylation

4a

14

13

15

12

11

6

The saframycins possess a similar mechanism of binding as Et743, involving

reversible formation of a noncovalent complex (hydrogen bonded) and an acid-promoted

covalent binding to guanine. In addition, the saframycins possess a quinone functionality

which allows for an alternative method of covalent bonding. Using saframycin A (1a) as

an example, reducing cofactors such as dithiothreitol can lead to the formation of a

hydroquinone 16 (Scheme 1.02). At this oxidation level a ring fragmentation and loss of

cyanide can occur, forming the electrophilic iminium 18 via intermediate 17. Alkylation

of the 2-amino group of guanine 13, as before, generates the covalently bonded adduct

19. Yet another possible mechanism for biological activity is through single strand

scission of DNA caused by reactive oxygen species (O2−·) produced by the redox cycling

of the quinone functionality present in saframcyins.39

NN

Me

O

OOMe

O

O

Me

MeO

HH

H

NH

H

1

3

OO

Me

Me

CN87

6

5 4

18 17

1615

142113

11

NN

Me

O

OOMe

OH

O

Me

MeO

HH

H

NH

H

OO

Me

Me

CN

[H]

H

NN

Me

O

OOMe

OH

O

Me

MeO

HH

H

NH

OO

Me

Me

H

NN

Me

O

OOMe

OH

OH

Me

MeO

HH

H

NH

OO

Me

Me

HN

N

N

NH2N

O

NN

Me

O

OOMe

OH

OH

Me

MeO

HH

H

HN

OO

Me

Me

HN

N

N

NH2N

O

Scheme 1.02. Saframycin A DNA Alkylation

1a

19 13

17 16

18

7

1.3. BIOSYNTHESIS1,9

The biosynthesis of some tetrahydroisoquinoline alkaloids, namely the

saframycins, safracins, and cyanocycline A is well investigated, while that of the more

structurally complex Et743 has not been fully realized. In 1985, 14C-labeled experiments

demonstrated that the biosynthesis of saframycin A involves the amino acids tyrosine,

glycine, alanine, and methionine.40 Two tyrosine residues 20 cyclize to form a dimeric

intermediate (Scheme 1.03), followed by incorporation of a glycine-alanine dipeptide 21

which becomes the pyruvamide side chain. The five methyl groups were shown to be

incorporated from methionine units 22.

NN

Me

O

OOMe

O

O

Me

MeO

HH

H

NH

H

OO

Me

Me

CN

CO2H

NH2HO

CO2H

NH2HO O

NH

ONH2

Me

OH

MeS NH2

CO2H

*

SMeH2N

CO2H

*

*

**

*

*

Scheme 1.03. Biosynthesis of Saframycin A.

The biosynthesis of unstable naphthyridinomycin was elucidated by analogy with

the more stable cyanocycline A (6a). In 1982, 14C-labeled and 15N-labeled experiments

showed the involvement of tyrosine, methionine, glycine, and ornithine (Scheme 1.04).41

The glycine unit 23 is converted to serine (24) which becomes the oxazolidine portion of

20

22 1a 21

22

20

8

6a.42 The ornithine unit 25 forms the bicyclo[3.2.1] system, while the three methyl

groups were shown to be incorporated from methionine units 22. Studies showed that

methylation of tyrosine (20) was followed by hydroxylation to form catechol 27 before

incorporation into cyanocycline A.43

NNMe

O

O

Me

MeO

H

CNOH

H

N

O

HH

HO2C 15NH2

*

HO2C 15NH2

OH

*

*

HO2C NH2

H2N

CO2H

NH2HO

CO2H

NH2HO

CO2H

NH2HO

Me

Me

OH

*

*

MeS NH2

CO2H

*

Scheme 1.04. Biosynthesis of Cyanocyline A

23

25 27

6a

22

20

26

24

9

29

1.4. SYNTHETIC APPROACHES TO TETRAHYDROISOQUINOLINE ALKALOIDS

Numerous syntheses of tetrahydroisoquinolines have been reported since the first

synthesis of saframycin B by Fukuyama in 1982.44 The emergence of Et743 as a

promising therapeutic agent and the discovery of new structures, including lemonomycin,

have kept a number of research groups publishing in this area. While a variety of

different synthetic approaches have been published there are fundamental similarities in

strategy to address the formation of key stereogenic centers. Formation of the 1,3-cis-

substituted tetrahydroisoquinoline ring system of the western fragment has proven to be

the primary focus of many strategies (Scheme 1.05). The most common strategy is

formation via a Pictet-Spengler cyclization late in the synthesis. This is used in the

syntheses of saframycins, renieramycins, and lemonomycin. The 1,3-cis-substituted

tetrahydroisoquinoline ring system of the eastern fragment is also often formed via a

Pictet-Spengler cyclization or by a Mannich reaction. These approaches and other

solutions to stereochemical control will be discussed in detail in the proceeding sections.

NN

Me

O

OOMe

O

O

Me

MeO

HH

H

X

H

1

3 R

R

HNN

Me

OMe

HOOMe

OMe

OH

Me

MeO

HH

H

R

R

1

3 HNN

Me

OMe

HOOMe

OMe

OH

Me

MeOH

R

R

1

3

1st 1,3-cis-substituted tetrahydroisoquinoline

2nd 1,3-cis-substituted tetrahydroisoquinoline

X

OHC+

H1

3

Scheme 1.05. Pictet-Spengler Approach to Tetrahydroisoquinoline Alkaloids

28

30

31

10

Saframycins

A (1a) R1=CN, R2=H B (1b) R1=R2=H C (1c) R1=H, R2=OMe G (1d) R1=CN, R2=OH S (1e) R1=OH,R2=H

1.4.1. THE SAFRAMYCINS

OMe

MeOO

NN

OOMe

Me

OMe

H

H

H

H

NH

OO

Me

R1

R21

11

13

1615

8

75

4

3

19

24

22

18

17

21

10

1.4.1.1. Fukuyama’s Total Synthesis of (±)-Saframycin B

Fukuyama and co-workers were the first to publish the total synthesis of a

saframycin, saframycin B.44 The synthesis starts with the formation of intermediates 32

and 34, each from aldehyde 33 (Scheme 1.06). The symmetric structure of the arene units

of the saframycins enable their synthesis from the same aldehyde 33. A condensation of

32 and 34 followed by acetate formation gave amide 35. Formation of aldehyde 36 was

performed in a three-step sequence; ozonolysis and treatment with dimethyl sulfide gave

the correct oxidation state at the C11 stereocenter (saframycin numbering), followed by

elimination of acetic acid with DBU. N-Acyliminium cyclization via a Pictet-Spengler

ring closure was promoted by heating 36 in formic acid to give the bicyclo[3.3.1] 37.

Fukuyama notes that a similar intermediate 38 does not undergo the analogous

cyclization to 39 (Scheme 1.07). The lack of reactivity was attributed to steric bulk

around the C3 stereocenter and the arylbenzyl ether.

11

MeOMe

MeOOBn

CHO MeOMe

MeOOBn

MeOMe

MeOOBn

NH2

PhOH

CO2H

NHCbz

4 steps

76% yield

6 steps

84% yield

MeOMe

MeOOBn

HN

PhOAc

O

NHCbz

OMeMe

OMeOBn

MeOMe

MeOOBn

HN

O

NHCbz

OMeMe

OMeOBn OMe

Me

MeOOBn

HN

O

N

BnOOMe

Me

OMeCbz

H

H

O

i, ii iii, iv

v11

Reaction Conditions: i) DCC, CH2Cl2 (83%); ii) Ac2O, pyr., 60 °C (98%); iii) O3, −78 °C; then Me2S; iv) DBU, CH2Cl2, 0 °C; v) HCO2H, 60 °C (74% over 3 steps).

Scheme 1.06. Fukuyama’s Synthesis of (±)-Saframycin B

OMeMe

MeOOBn

N

O

N

BnOOMe

Me

OMeCO2Me

H

H

MeOMe

MeOOBn

N

O

N

OMeMe

OMeOBn

H

HOH

CO2Me Hi3 3

Reaction Conditions: i) HCO2H, 60 °C, 20 min.

Scheme 1.07. Fukuyama’s Unsuccessful Pictet-Spengler Cyclization

32 33 34

32 + 34

35

36 37

38 39

12

41

The synthesis continued with catalytic hydrogenation of 37 which

stereoselectively formed the C3 center (Scheme 1.08). Reductive alkylation followed by

reduction of the lactam gave N-methylamine 40. Hydrogenolysis and a second Pictet-

Spengler cyclization with aldehyde 41 gave predominantly the desired cyclized product

42 in a ratio of 6:1 over the α-isomer. The final steps included removal of the Cbz group,

pyruvamide formation, and final oxidation to the bis-isoquinolinequinone to give

(±)-saframycin B (1b). The total synthesis was completed in 18 steps with an 8% overall

yield from 33.

CbzHN CHO

i-iiiiv

v-vii

OMeMe

MeOOH

HNN

HOOMe

Me

OMeMe

H

H

H

OMeMe

MeOOH

NN

HOOMe

Me

OMeMe

H

H

H

H

NHCbz

OMe

MeOO

NN

OOMe

Me

OMe

H

H

H

H

NHCOCOMe

3

Reaction Conditions: i) H2 (1000 psi), Raney Ni, 100 °C; ii) H2 (1000 psi), 37% aq. HCHO, Raney Ni; iii) AlH3, THF (75%, 3 steps); iv) 41, CH3CN, 70 °C (75%); v) H2, 10% Pd/C, AcOH vi) ClCOCOMe, PhNMe2 (72%, 2 steps); vii) CAN, 0 °C (37%).

Scheme 1.08. Fukuyama’s Synthesis (±)-Saframycin B

42

37

1b

40

13

1.4.1.2. Kubo’s Total Synthesis of (±)-Saframycin B

The next total synthesis of a saframycin was of (±)-saframycin B by Kubo and

co-workers.45 In this new approach, the C2-symmetrical diketopiperazine 44 (Scheme

1.09) was the key starting building block. A sequence of condensations with aldehyde 43

led to arylidenediketopiperazine 45. Benzyl protection of the amide followed by

conversion of the N-acetyl protected amide furnished the isopropylcarbamate 46. The

first Pictet-Spengler ring closure was carried out by selectively reducing the C11

carbonyl moiety (saframycin numbering) to form a hemi-aminal which cyclized, as in

Fukuyama’s work, upon heating in formic acid. The isopropylcarbamate was then

removed and the secondary amine methylated. Reduction of the amide and

stereoselective catalytic hydrogenation gave 47.

AcNNAc

O

O

MeOMe

MeOOMe

CHO

OMeMe

MeOOMe

HNN

MeOOMe

Me

OMeMe

H

H

+Me

OMe

MeOOMe

HN

O

NAc

OMeMe

OMeOMeO

i-iv

MeOMe

MeOOMe

BnN

O

N

OMeMe

OMeOMeO

CO2iPr

v-vii

viii-xiii11

Reaction Conditions: i) t-BuOK, t-BuOH, DMF, 25 °C (66%); ii) 10% Pd/C, EtOH, DMF; iii) Ac2O, 110 °C (80% yield 2 steps); iv) t-BuOK, t-BuOH, DMF, 25 °C (81%); v) BnBr, NaH; vi) NH2NH2·H2O (94%, 2 steps); vii) ClCO2

iPr, DMAP, TEA (94%); viii) LiAlH(OtBu)3; ix) HCO2H, 60 °C (52%, 2 steps); x) H2SO4, TFA xi) 37% aq. HCOH, HCO2H, 70 °C (96%, 2 steps); xii) AlH3, THF, 0 °C (93%); xiii) 20% Pd/C, H2 (4 atm), EtOH, 80 °C (99%).

Scheme 1.09. Kubo’s Synthesis of (±)-Saframycin B

47 46

4544 43

14

48 49

At this stage in the synthesis Kubo used an alternative substrate than Fukuyama to

install the side-chain group, butyl glyoxylate. Kubo used a two step procedure that

coupled the n-butyl glyoxylate to 47 and then a second Pictet-Spengler ring closure was

promoted by treatment with TFA (Scheme 1.10). In contrast to Fukuyama’s work, Kubo

only observed the opposite stereochemistry at the newly formed C1 stereocenter, giving

solely the 1,3-trans-substituted tetrahydroisoquinoline 48. Numerous attempts to

epimerize the stereocenter under basic conditions were unsuccessful. Complete inversion

of the stereocenter was achieved by oxidation to the iminium with mercury acetate and

reduction with sodium borohydride yielding solely 1,3-cis-49.

i, ii iii, iv

v-viii ix, xOMe

Me

MeOOMe

NN

MeOOMe

Me

OMeMe

H

H

H

H

NHCOCOMe

OMeMe

MeOOMe

NN

MeOOMe

Me

OMeMe

H

H

H

CO2nBu

OMeMe

MeOOMe

NN

MeOOMe

Me

OMeMe

H

H

H

CO2nBu

H1

Reaction Conditions: i) CHOCO2Bu, K2CO3, BuOH; ii) TFA, 25 °C (70%, 2 steps); iii) Hg(OAc)2, 5% AcOH/H2O, 90 °C; iv) NaBH4, EtOH/H2O (71%, 2 steps); v) LAH, THF, reflux (77%); vi) DEAD, PhtNH, PPh3, THF; vii) NH2NH2·H2O, EtOH, reflux; viii) ClCOCOMe, DMAP, TEA, CH2Cl2 (76%, 3 steps); ix) BBr3, CH2Cl2, −78 °C to 0 °C; x) 10 M HNO3, 25 °C (41%, 2 steps). Scheme 1.10. Kubo’s Synthesis of (±)-Saframycin B

50

1b

47

15

The side-chain pyruvamide was installed by reduction of the ester, Mitsunobu

displacement of the alcohol with phthalamide, hydrazinolysis and treatment with

pyruvoyl chloride to give 50. The final oxidation to the quinone was accomplished by

first deprotecting the ArOMe with BBr3. Direct oxidation of the trimethoxybenzene gave

only decomposition. Oxidation of the phenol with nitric acid gave (±)-saframycin B (1b).

The total synthesis was accomplished in 22 steps with a 3% overall yield.

1.4.1.3. Fukuyama’s Total Synthesis of (±)-Saframycin A

Soon after Kubo’s synthesis Fukuyama and co-workers reported a total synthesis

of (±)-saframycin A.46 Their approach utilized a similar initial strategy previously

reported by Kubo, using the C2-symmetrical diketopiperazine 44 and aldehyde 33

(Scheme 1.11). A similar sequence of 11 steps afforded amide 51. At this stage

Fukuyama addressed the key difference between saframycin A and B, the oxidation state

at C21. Boc protection of the phenols and amide of 51 allowed reduction of the amide

carbonyl to the ring opened alcohol 52. Boc deprotection followed by a Pictet-Spengler

with aldehyde 53, stereoselectively formed the 1,3-cis-substituted tetrahydroisoquinoline

54 with only trace amounts of the unwanted α-isomer. Swern oxidation formed an

unstable hemi-aminal which was stereoselectively converted to the amino nitrile 55

providing the correct stereochemistry at C21. The final steps involved deprotection of the

side-chain, pyruvamide formation, and oxidation with DDQ to give (±)-saframycin A

(1a). The total synthesis was completed in 20 steps with 9% overall yield.

16

OMeMe

MeOOH

HN

O

N

HOOMe

Me

OMeMe

H

H

H

OMeMe

MeOOBoc

NHBoc

H NH H

MeOH

OMeMe

MeO

BocO OMeMe

MeOOH

NH

H NH H

MeOH

OMeMe

MeO

HO

BocHN CHO

i, ii

iii

ivH

NHBoc

viiiv-vii

11 steps

41% yield

OMeMe

MeOOH

NN

HOOMe

Me

OMeMe

H

H

H

H

NHCOCOMeCN

21

Reaction Conditions: i) Boc2O, DMAP, DMF, 60 °C (81%); ii) NaBH4, EtOH, 0 °C (92%); iii) TFA; iv) 53, MeOH, 60 °C (82%, 2 steps); v) Swern oxid., then NaCN, MeOH (67%); vi) TFA; vii) ClCOCOMe, NaHCO3, CH2Cl2 (86%, 2 steps); viii) DDQ, 0 °C (60%).

Scheme 1.11. Fukuyama’s Synthesis of (±)-Saframycin A

33

52

53

54

51

1a

55

44 +

17

1.4.1.4. William’s Studies Toward the Saframycins

Another approach to the saframycins that involved Pictet-Spengler ring closure

was reported by Williams.47 This enantioselective approach started from the condensation

of aldehyde 33 (Scheme 1.12). Treatment of the resulting imine with the ketene, formed

from the reaction of acid chloride 56 with triethylamine at −78 °C, gave β-lactam 57. The

chiral auxiliary was removed under hydrogenolysis and the free amine underwent Pictet-

Spengler ring closure with methyl glyoxylate to give a single diastereomer 58, possessing

the unwanted α-orientation at C1. Contrary to Kubo’s inversion of C1, basic conditions

were sufficient to epimerize the C1 center as a 3:1 mixture in favor of the cis-isomer 59.

The side-chain and phenol were converted to benzyl ethers in four steps to arrive at 60.

MeMeO

MeOOBn

N

OPh

PhO

COCl

i, ii

BnNH

NH

O

OPh

Ph

O

MeMeO

MeOOH

NH

CO2Me

BnN OH

HMe

MeO

MeOOH

NH

CO2Me

BnN OH

HMe

MeO

MeOOBn

NH

BnN OH

H

OBn

H H

iii, iv

v vi-ix1

Reaction Conditions: i) BnNH2, PhH, reflux; ii) TEA, CH2Cl2, −78 °C to 0 °C (99%, 2 steps); iii) Pd(OH)2, H2 (60 psi), MeOH/THF; iv) CHOCO2Me, MeOH (84%, 2 steps); v) DBU, THF (75%); vi) Boc2O, EtOH; vii) LiBH4, MeOH, Et2O, reflux (78%, 2 steps); viii) NaH; BnBr, nBu4NI; ix) TMSOTf, 2,6-lutidine (55%, 2 steps).

Scheme 1.12. William’s Studies Toward Saframycins

58 59

57

60

56

33

18

A coupling between amine 60 and amino acid analogue 61 afforded amide 62

(Scheme 1.13). In order to promote the second Pictet-Spengler cyclization the TBS and

Fmoc protecting groups were simultaneously removed using TBAF. The product was

carefully treated with Super-Hydride® to open the β-lactam and eliminate BnNH2 to form

intermediate 63. Upon aqueous workup the bicyclo[3.3.1] product 64 was formed. This

one-pot procedure formed the carbon skeleton of the saframycins very efficiently but the

key C3 stereogenic center was lost. Williams reported that attempted reduction of the

3,4-olefin was unsuccessful without degradation and no further work was reported.

MeNFmoc

Cl

O

OMeOTBSMe

+ MeMeO

MeOOBn

N

BnN OH

H

OBn

H

O

MeN

H

Fmoc

TBSOOMe

Me

MeMeO

MeOOBn

N

OBn

H

O

NHMe

O

H

MeOMe

OH

OMeMe

MeOOBn

NN

HOOMe

Me

Me

H

H

OBn

H

O

i ii, iii3

Reaction Conditions: i) DMAP, CH2Cl2 (84%); ii) TBAF, THF (80%); iii) LiBEt3H, THF, 0 °C, then aqueous workup (49%).

Scheme 1.13. William’s Studies Toward Saframycins

64

6261

60

63

19

1.4.1.5. Corey’s Enantioselective Synthesis of (–)-Saframycin A

In 1996 E. J. Corey and co-workers published the enantioselective synthesis of

ecteinascidin 743.48 Three years later Corey reported a divergent approach in this

synthesis that allowed access to the saframycins.49 Corey demonstrated this by

synthesizing (−)-saframycin A. The enantioselective synthesis was a convergent approach

utilizing amino acid analogues 67 and 68 (Scheme 1.14). The highlight of this strategy

employed dimethyl acetal 66, formed in a seven step sequence from phenol 65, for the

intramolecular Pictet-Spengler ring closure to form the 1,3-cis-substituted

tetrahydroisoquinoline 67. With the C3 stereocenter already established, formation of the

trans-isomer during the Pictet-Spengler was not a possibility, resulting in complete

control of diastereoselectivity. Amine 67 was condensed with aldehyde 68 in the

presence of acetic acid and KCN to stereoselectively form the aminonitrile 69. Formation

of the bicyclo[3.3.1] system required reduction of the lactone to the lactol, followed by

desilylation and acid catalyzed Pictet-Spengler, which gave solely the cis-isomer 70 due

to the established C13 stereocenter. At this stage the backbone structure of the

saframycins was established. A 16 step sequence of functional group interconversions

successfully completed the enantioselective synthesis of (−)-saframycin A (1a). From

aldehyde 65 the synthesis was completed in 31 steps with an overall yield of 0.4%.

20

MeOBn

HNO

O

OBn

O

OMe

OMe

H

NH

OO

H

HMe

OHOMe

OTBSTBSO

CHO

AllocHN N

OO

H

MeOAllyl

HN

TBSOOMe

OTBS

CN

AllocH

H

+

7 steps

42% yield

i-iii

iv, v

vi-viii 16 steps

4% yield

OH

OMeMe

NN

HOOMe

OH

Alloc

H

H

H

H

OHCN

OO

OO

OO

OO

OO

3

13

Reaction Conditions: i) BF3·OEt2, H2O; ii) BF3·OEt2, 4Å molecular sieves (73%, 2 steps); iii) 10% Pd/C, H2 (100%); iv) HOAc, KCN (61%); v) allyl bromide, Cs2CO3, DMF (87%); vi) DIBAL, PhMe, −78 °C; vii) KF·2H2O, MeOH; viii) CH3SO3H, 3Å molecular sieves, CH2Cl2 (55%, 3 steps).

Scheme 1.14. Corey’s Enantioselective Synthesis of (–)-Saframycin A

67

70

68 69

1a

6665

21

Renieramycins

A (2a) R1=R2=H, R3=OH B (2b) R1=R2=H, R3=OMe C (2c) R1=R2=O, R3=OH D (2d) R1=R2=O, R3=OEt E (2e) R1=OH, R2=R3=H F (2f) R1=OH, R2=H, R3=OMe G (2g) R1=R2=O, R3=H

1.4.2. THE RENIERAMYCINS

OMe

MeOO

NN

OOMe

Me

OMe

H

H

H

H

O

OMe

R1

R31

11

13

1615

8

75

4

3

19

24

22

18

17

21

10

R2

H

Me

26

1.4.2.1. FUKUYAMA’S SYNTHESIS OF (±)-RENIERAMYCIN A

The only total synthesis of a renieramycin before 2005 was a stereoselective

synthesis of (±)-renieramycin A (2a) completed by Fukuyama and co-workers in 1990.50

Although much of the strategy is similar to Fukuyama’s previous work in the area, the

synthesis was essential to unambiguously prove the stereochemistry at the C1 center. The

stereochemistry of the angelate side-chain of renieramycins was originally assigned as

the α-isomer,51 however further review of spectroscopic data revealed similarities

between saframycin C and renieramycin A, suggesting that both share the same

β-configuration. Faulkner revised the assignment,52 and Fukuyama proved this to be

correct upon completion of his total synthesis.

The synthesis began from readily available aldehyde 71 (Scheme 1.15). In

anticipation of installation of the C14 hydroxyl late in the synthesis, Fukuyama takes

great measures to synthesize differentially protected phenol 72, so that C17-OH can be

generated to facilitate benzylic oxidation. Conversion of 71 to 72 requires 10 steps,

followed by a condensation with diketopiperazine 44. Similar steps to the previous

synthesis of saframycin A led to phenol 73.

22

CHO

OMe

AcNNAc

O

OMe

OMe

10 steps

55% yield

OMOMOR

MeOMe

R = (CH2)3ODMTS

+

OMeMe

MeOOBn

HN

O

NR

BnOOH

Me

OMeH

H

H

11 steps

13% yield

OMeMe

MeOOBn

HN

O

N

BnOOMe

Me

OMeMe

H

H

H

OMeMe

MeOOH

HNN

HOOMe

Me

OMeMe

H

H

H

OMeMe

MeOOH

NN

HOOMe

Me

OMeMe

H

H

H

H

O

OMe

Me

OHOH

OH

v, vii-iv

vii viii

R = CO2(CH2)2CO2Me

OO

Me

Me

CHO

HH

Reaction Conditions: i) DDQ, THF/H2O (8:1); ii) MeI, K2CO3, DMF; iii) DBU, MeOH; iv) HCHO, NaBH3CN, TFA, MeOH (45%, 4 steps); v) AlH3, THF vi) H2, Pd/C, EtOH (64%, 2 steps) vii) glycolaldehyde angelate, CH3CN, 50 °C (66%) viii) DDQ, actone/H2O (20:1) (48%).

Scheme 1.15. Fukuyama’s Synthesis of (±)-Renieramycin A

The C18 phenol was protected as the benzyl ether, while the C17 phenol existed

as the free alcohol, which upon treatment with DDQ stereoselectively hydroxylates from

the exo-face. The phenol was alkylated with MeI and the carbamate converted to the

2a

4471 72

74

75

77

73

76

23

N-methylamine 74. Reduction of the amide with AlH3 and diastereoselective catalytic

hydrogenation gave amine 75. The angelate side-chain was installed by Pictet-Spengler

reaction with glycolaldehyde angelate 76 to furnish a 5:1 mixture of 77 and its α-isomer.

Fukuyama was able to establish the relative stereochemistry by a single crystal X-ray

analysis of 77. (±)-Renieramycin A (2a) was formed upon oxidation with DDQ giving a

structure identical to that of the authentic sample. The synthesis was completed in 29

steps with an overall yield of 0.7%.

1.4.2.2. Danishefsky’s Synthesis of Cribrostatin IV (Renieramycin H)

OMe

MeOO

NN

HOOMe

Me

OHMe

H

H

H

O

OMe

Me

O

O

H

1

11

13

1615

8

7

5

43

19

24

22

18

17

21

10

26

Danishefsky and co-workers have recently reported the synthesis of cribrostatin

IV (renieramycin H, 78).53 Pettit reported the isolation and structural elucidation of this

compound from a blue marine sponge, Cribrochalina.54 However, around the same time

Kubo had semi-synthetically derived the same compound and characterized it as

renieramycin H.55 The significant differences when compared with renieramycin A are

the 3,4-olefin, the C14 ketone and the existence of the hydroquinone, whose presence is

attributed to the adjacent ketone.

Cribrostatin IV (Renieramycin H, 78)

24

Danishefsky used a strategy he developed in the 1980’s in his approach to the

quinocarcins.56 The enantioselective synthesis began with the construction of

tetrahydroisoquinoline 81 derived from phenol 79 (Scheme 1.16). A 12 step sequence of

reactions, in which a Noyori protocol57 enantioselectively installed the C1 stereocenter,

gave dimethylacetal 80. The tetrahydroisoquinoline 81 was formed from a Bobbitt

modified Pomeranz-Fritsch cyclization.58 The amine of 81 was coupled with amino acid

analogue 82, then PMB removal followed by Dess-Martin oxidation formed the C4-keto-

benzaldehyde 83.

MeCHO

HOOH

MeOAllyl

MeOOMe

NH

OMe

OMe

OBn

MeAllylO

MeOOMe

NH

OBn

OH

H

BnOOMe

Me

BocHNHO

O

PMBO

Me

H

BnOOMe

Me

BocN

O

Me

H

MeAllylO

MeOOMe

N

OBn

O

H

O

12 steps

22% yield

i

+ii-iv

Reaction Conditions: i) 8.0 M HCl/dioxane (97%); ii) BOPCl, TEA, CH2Cl2 (89%); iii) DDQ, CH2Cl2, pH = 7 buffer (90%); iv) DMP, 2,6-lutidine, CH2Cl2 (84%).

Scheme 1.16. Danishefsky’s Synthesis of Cribrostatin IV

79

83

80 81

81

82

25

Formation of the bicyclo[3.3.1] system was achieved by treatment of aldehyde 83

with formic acid at 100 °C (Scheme 1.17). This led to loss of the N-Boc protecting group,

imine formation and Mannich closure to form 84. The diastereoselective ring closure

formed solely the 1,3-trans-substituted tetrahydroisoquinoline, which is normally the

undesired orientation. However, the target molecule possesses unsaturation at the C3-C4

position making the stereochemistry at the newly formed C3 stereocenter irrelevant.

Reduction of the C4 ketone and elimination was facilitated by first deprotecting the

epi-allylether to generate the phenol. The phenol was reprotected as the TBS ether and

selective debenzylation of the arylbenzyl ether over the alkylbenzyl ether afforded 85. In

order to install the C14 ketone the phenol was first oxidized to the quinone with Fremy’s

salt. Benzylic oxidation to the ketone was promoted with selenium dioxide followed by

oxidation with Dess-Martin periodinane. The quinone was reduced back to the

hydroquinone and the alkylbenzyl ether was hydrogenolyzed in the same step under

catalytic hydrogenation conditions, producing alcohol 86. The angelate ester was readily

formed by simply allowing a solution of alcohol 86 and angeloyl chloride (87) to stand

for 12 hours, followed by TBS removal to afford phenol 88. A three step sequence was

necessary to furnish the mono-quinone cribrostatin IV. Oxidation to the bis-

isoquinolinequinone was necessary to remove the arylmethyl ether. Reduction of the bis-

isoquinolinequinone and air oxidation selectively oxidized the A-ring hydroquinone to

furnish cribrostatin IV (78). The synthesis was completed in 31 steps with an overall

yield of 2%.

26

AllylOMe

MeOOMe

NN

BnOOMe

Me

Me

H

H

H

H

OBnO

O OTBSMe

MeOOMe

NN

HOOMe

Me

Me

H

H

H

OBnO

OTBSMe

MeOOMe

NN

HOOMe

Me

Me

H

H

H

OHO

OHMe

MeOOMe

NN

HOOMe

Me

OHMe

H

H

H

O

OMe

Me

O

O

OH

O

i ii-vi

vii-x

Cl

O

MeH

Me

xi, xii

xiii-xv

H

OMe

MeOO

NN

HOOMe

Me

OHMe

H

H

H

O

OMe

Me

O

O

H

Reaction Conditions: i) HCO2H, 100 °C (59%) ii) NaBH4, THF/H2O; iii) AcOH, Bu3SnH, (Ph3P)2PdCl2, CH2Cl2 (98%, 2 steps) iv) CSA, PhH, 80 °C (80%); v) TBSOTf, TEA, CH2Cl2 (90%); vi) 5% Pd/C, H2 (1 atm), EtOAc (90%); vii) Fremy’s salt, KH2PO4, CH3CN/H2O (84%); viii) SeO2, dioxane, 100 °C (87%) ix) DMP, CH2Cl2; x) 10% Pd/C H2 (1 atm), MeOH (89%, 2 steps); xi) 87, CH2Cl2; xii) AcOH, TBAF, THF (75%, 2 steps); xiii) PIFA, CH3CN/H2O; xiv) Zn, AcOH; xv) air, DMF (65%, 3 steps).

Scheme 1.17. Danishefsky’s Synthesis of Cribrostatin IV

88

86

87

85 84

83

78

27

This strategy was successful in the synthesis of Cribrostatin IV, due to the lack of

stereochemistry at the C3 center, but Danishefsky’s attempts to synthesize other

tetrahydroisoquinoline alkaloids employing the same methodology were not as

successful. In his approach to the saframycins59 a similar Mannich cyclization on a

slightly different substrate 89 yielded only the 1,3-cis-isomer 90 rather than the trans-

isomer observed above (Scheme 1.18). In further contrast, this same strategy was

employed in Danishefsky’s synthesis of quinocarcins.56 However, dimethylacetal 91

cyclized to give only the trans-isomer on formation of the bicyclo[3.2.1] 92 (Scheme

1.19).

MeOOMe

Me

BocN

O

Me

H

MeMeO

MeOOMe

N

OBn

O

H

OOMe OMe

Me

MeOOMe

NN

MeOOMe

Me

Me

H

H

H

OBnO

OMeH

OHCO2H, reflux

Scheme 1.18. Danishefsky’s Approach Toward Saframycins

MeOBocN

O

Me

HOMe

N

O

H

MeOCO2Me

CO2Me

HCO2H, reflux

OMe

N

O

HNMe

HH

CO2MeMeO2C

HO

Scheme 1.19. Danishefsky’s Approach Toward Quinocarcin

89 90

92 91

28

The strategy outlined by Danishefsky was the first to combine the two subclasses

of tetrahydroisoquinoline alkaloids, the mono- and bis-tetrahydroisoquinoline alkaloids.

Unfortunately, the stereoselectivity necessary for formation of the

1,3-cis-tetrahydroisoquinoline was substrate dependent such that in the bicyclo[3.2.1]

ring formation the undesired stereochemistry was observed.

1.4.3. LEMONOMYCIN

NNH

H

HOH

MeO

Me

O

H

H

O

O

Me

OHMe

NMe2

O

HOOH

1

11

13

16

15

8

75

4

3

22

21

10

1.4.3.1. Stoltz’s Synthesis of (–)-Lemonomycin

The structure of (–)-lemonomycin (10) is quite unique in that it is the only

tetrahydroisoquinoline alkaloid to possess a glycosidic linkage and the rare aldehyde

hydrate functionality. The enantioselective synthesis of (–)-lemonomycin by Stoltz

further confirmed the rare functional groups.60 The synthetic strategy proved to be not

nearly as unique but rather an efficient combination of known strategies in the synthesis

of tetrahydroisoquinoline alkaloids. The early enantioselective formation of the

bicyclo[3.2.1] system was a slight modification of the strategy used by Garner and

co-workers for the synthesis of (−)-quinocarcin.61 Garner assembles the bicyclo[3.2.1]

(–)-Lemonomycin (10)

29

system via a 1,3-dipolar addition of azomethine ylide 94, formed from irradiation of

aziridine 93, with Oppolzer’s chiral acryloyl sultam 95 to diastereoselectively form the

bridged ring system 96 (Scheme 1.20).

hν (2537 Å)

dioxane+

N

SO2

ON

O

OOMe

Me

OH

NMe

N

OMeOH

NMeO

O

Me

NN

H

HO

O

OMe

MeH

OH

ON

SO2Me

61% yield

Scheme 1.20. Garner’s Approach to Quinocarcin

The cycloaddition employed by Stoltz used the Joule oxidopyrazinium62 97 and

the same Oppolzer’s chiral acryloyl sultam 95 to arrive at bicycle 98 in 94% ee (Scheme

1.21). Conversion to vinyl iodide 99 was followed by a Suzuki coupling with boronate

100, to give eneamide 101. The diastereoselectivity of the 3,4-olefin reduction followed

known observations,61 giving solely amide 102. Formation of the 1,3-cis-substituted

tetrahydroisoquinoline used methodology developed by Fukuyama in his synthesis of

renieramycin A; employing a Pictet-Spengler with the fully constructed side-chain

tethered to an aldehyde.50 Unfortunately the Pictet-Spengler ring closure between 102 and

104 proved unsuccessful, as the nitrogen amide was not reactive. Amide 102 was then

converted to amine 103 in five steps.

9495 93

96

30

HNN

O

BnBr

N

O

SOO

HNNBn

H

HO

OH

HNNBn

H

HO

OTIPS

I

OMeBMe

MeOOTs

O

O

HNNBn

H

HO

OTIPS

MeO

MeOMe

OTs

HNNBn

H

HO

OTIPS

MeO

MeOMe

OTs

H

H2NN

H

HOH

OH

MeO

MeOMe

OH

HCbz

+i, ii iii, iv

+ v

vi vii-xi

Reaction Conditions: i) N-Me-morpholine, CH3CN, −20 °C; ii) NaBH4, EtOH (72%, 2 steps); iii) TIPSOTf, CH2Cl2, 2,6-lutidine (82%); iv) ICl, CH2Cl2, 0 °C (81%); v) Pd(PPh3)4, K2CO3, PhH, MeOH, 70 °C (69%); vi) Pd/C, H2 (1000 psi), TFA, EtOH (72%); vii) CbzCl, DMAP, CH3CN; viii) KOTMS, CH3CN (87%, 2 steps); ix) Boc2O, DMAP, CH3CN; x) NaBH4, EtOH; xi) HCl, MeOH (81%, 3 steps).

Scheme 1.21. Stoltz’s Synthesis of (–)-Lemonomycin

The Pictet-Spengler cyclization with the more reactive amine 103 and aldehyde

104 gave the 1,3-cis-substituted tetrahydroisoquinoline 105 with excellent

diastereoselectivity (Scheme 1.22). Completion of the synthesis followed with N-Cbz

removal, bis-Swern oxidation, equilibration to the bicyclo[3.2.1] ring system and

oxidation to give (−)-lemonomycin (10). This very efficient enantioselective synthesis

was completed in 15 steps from 97 with an overall yield of 3%.

95 98

101 10099

97

103102

31

NHN

H

HOH

OH

MeO

MeOMe

OH

HCbz

H

O

O

Me

OHMe

NMe2

CHO

O

O

Me

OHMe

NMe2

+i ii-iv

Reaction Conditions: i) EtOH (63%) ii) Pd/C, H2, EtOH iii) Swern, then aq. HCl (52%); iv) CAN, 0 °C (51%).

Scheme 1.22. Stoltz’s Synthesis of (–)-Lemonomycin

1.4.3.2. Fukuyama’s Approach Toward the Synthesis of (–)-Lemonomycin

Fukuyama reported a partial synthesis of (–)-lemonomycin in 2005 employing a

strategy his group had previously developed.63 In their work on Et743, a synthetic

strategy using the Ugi four-component condensation was successful in building the core

ring structure.64 The approach to (–)-lemonomycin began with phenol 106 (Scheme 1.23)

which undergoes a Mannich-type reaction with chiral template 107 to yield

phenylglycine derivative 108. A five-step sequence installed the requisite differential

protecting groups and side-chain oxidation state to form the phenylglycinol derivative

109.

The Ugi four-component condensation was effected by heating amine 109,

aldehyde 110, isonitrile 111, and amino acid 112 to provide in excellent yield amide 113

(Scheme 1.24). This efficient procedure installed in one step all of the necessary atoms

needed to form the core of lemonomycin. Unfortunately, arranging the atoms to the

correct connectivity proved tedious. Mild acid hydrolysis of the dimethyl acetate formed

cyclized product enecarbamate 114.

105

104

103 10

32

MeOMe

MeOOH

O

N

O

Ph MeOMe

MeOOH

O

HN

O

Ph+

i MeOMe

MeOOMs

OTBDPS

NH2

ii-vi

Reaction Conditions: i) TFA, CH2Cl2, −10 °C (93%); ii) MsCl, TEA, CH2Cl2, 0 °C; iii) NaBH4, MeOH; iv) TBDPSCl, imid., DMF (90%, 3 steps); v) Pb(OAc)4, CH3CN 0 °C; vi) NH2OH·HCl, NaOAc, EtOH, 0 °C (74%, 2 steps).

Scheme 1.23. Fukuyama’s Approach to (–)-Lemonomycin

MeO CHO

OMe

OCO2PhCN

HO2C

NHBoc i

MeMeO

MeOOMs

OTBDPS

NO

HNOMeMeO OCO2Ph

O

NHBocMe

MeO

MeOOMs

NNBoc

OOTBDPS

HN

OCO2Ph

Oii

MeOMe

MeOOMs

OTBDPS

NH2

Reaction Conditions: i) CF3CH2OH, 50 °C; ii) CSA, quinoline, PhMe, reflux (73%, 2 steps).

Scheme 1.24. Fukuyama’s Approach to (–)-Lemonomycin

108107

106 109

112

111109

110

114113

33

Prior to the essential formation of the bicyclo[3.2.1] system the C4 amide was

converted to the acetate protected alcohol, as was the TBDMS ether. A metathesis

reaction was used to install the allyltrimethylsilane moiety providing cyclization

precursor 115 (Scheme 1.25). Exposure of 115 to BF3·Et2O generated a reactive

N-acyliminium intermediate 116 which was trapped by the allylsilane to give cyclized

product 117. This cyclization proved diastereoselective in formation of the C15

stereocenter, with no discussion of the observed selectivity. To achieve the required

formation of the 1,3-cis-substituted tetrahydroisoquinoline Fukuyama had previously

developed a method that treated an analogous eneamide with DMDO in methanol,

followed by an acidic reduction to reduce the C3 stereocenter.65 Unfortunately in this

synthesis the protecting groups proved to be incompatible. The acid sensitive protecting

groups were converted to more stable protecting groups and treatment with DMDO

formed methyl-aminal 118. Reduction with NaCNBH3 in TFA/THF provided the

requisite stereochemistry at C3. The final ring closure required a more electron-donating

aromatic ring thus the mesyl group was replaced by the corresponding benzyl ether.

Oxidation of the primary alcohol generated the aldehyde which upon exposure to TFA

cyclized to 119. No further work was reported on this intermediate.

34

NN

H

HO

MeO

Me

OMs

BocH

OAc

MeMeO

MeOOMs

NNBoc

OOAc

TMSOAc

MeMeO

MeOOMs

NNBoc

OOAc

Me3Si

NN

HO

MeO

MeOMe

OMs

CbzH

OTIPS

OHOMe

NN

HO

MeO

MeOMe

OBn

CbzH

OTIPS

OHH

i-v vi

vii-xi

xii-xvi3

OMe 15

Reaction Conditions: i) t-BuOK, 4Å molecular sieves, THF, 0 °C; ii) NaBH4, THF; iii) TBAF, THF, 50 °C; iv) Ac2O, pyr. (68%, 4 steps); v) Grubbs 2nd generation catalyst (2 mol%), allyltrimethylsilane, CH2Cl2, reflux (51%); vi) BF3·Et2O, CH2Cl2, −78 °C (95%); vii) TMSOTf, CH2Cl2, 0 °C; viii) CbzCl, DMAP, CH3CN; ix) K2CO3, MeOH; x) TIPSOTf, 2,6-lutidine, CH2Cl2, 0 °C (55%, 4 steps); xi) DMDO, Na2SO4, MeOH, −78 °C; CSA; xii) NaBH3CN, TFA/THF, 0 °C; xiii) KOSiMe3, CH3CN, 0 °C; xiv) BnBr, 50 °C (53%, 4 steps); xv) DMP, CH2Cl2; xvi) TFA/CH2Cl2 (76%, 2 steps).

Scheme 1.25. Fukuyama’s Approach to (–)-Lemonomycin

114

115

116 117

119118

35

1.5. RETROSYNTHETIC ANALYSIS

In our approach to the tetrahydroisoquinoline alkaloids we envisioned an

alternative route to form the key 1,3-cis-substituted tetrahydroisoquinoline core 122

(Scheme 1.26). With the exception of Corey’s work, the tetrahydroisoquinoline ring

forming reaction via Pictet-Spengler or Mannich cyclization has been shown to give both

the desired 1,3-cis and undesired 1,3-trans product depending on the substrate, thus

planning a strategy which predicts the stereoselectivity of these cyclizations a priori is

not possible. To avoid this potential problem our aim was to develop an alternative

method for the synthesis of the 1,3-cis-substituted tetrahydroisoquinoline ring system.

Unlike most syntheses, we planned to enantioselectively install the C1

stereocenter first. This chiral center would then be used to direct the formation of

subsequent stereocenters. The C3 stereocenter could be obtained via a reduction where

the steric bulk of the C1 side-chain would provide means for diastereoselectivity. For

installation of the C1 substituent we envision an enantioselective alkylation of

3-substituted-isoquinoline 125 with a nucleophile, M-CH2-X, to generate the

1,2-dihydroisoquinoline 124 followed by an ionic hydrogenation to establish the

1,3-cis-substituted tetrahydroisoquinoline core 123.

By building the western fragment first and elaborating the eastern bicycle last we

will have a potentially divergent route that allows formation of both the bicyclo[3.3.1]

system 120 and the bicyclo[3.2.1] system 121 late in the synthesis. Construction of the

bridged ring system will be realized from tricycle 122 by a carbon-carbon bonding

forming reaction at either C11 or C13, or by a cycloaddition generating both bonds at the

same time. The diastereoselectivity of these reactions will be essential but difficult to

36

predict. Our strategy has great potential to provide an efficient approach to both mono-

and bis-tetrahydroisoquinoline alkaloids.

OMe

MeOO

NN

OOMe

Me

OMe

H

H

H

X

H

Y Z

NNH

H CHO

HMeO

MeO

O

H

H

XY Z

NNR3

MeO

MeOMe

OR1

H

H

XO

NR2Y

MeO

MeOMe

OR1

H

H

XNR2Y

MeO

MeOMe

OR1

H

X

NY

MeO

MeOMe

OR1

1

11

13

3

1

3 1

3

1

11

13

3

1

11

13

3

1

3

Scheme 1.26. Retrosynthetic Analysis

122

121 120

124

123

125

37

1.6. CONCLUSION

The tetrahydroisoquinoline alkaloids have been of biological and chemical

significance for over 25 years. Their potent biological properties make them promising

therapeutics, while their structural complexity provides challenging synthetic targets.

Numerous research groups, far more than have been mentioned above, have made

important contributions in the synthesis of both mono- and bis-tetrahydroisoquinoline

alkaloids. Fukuyama has been the most prolific researcher publishing the first synthesis

of a tetrahydroisoquinoline in 1982 and a partial synthesis of lemonomycin using an

altogether different route in 2005. The strategy employed by Danishefsky had the most

potential for efficient access to both mono- and bis-tetrahydroisoquinoline alkaloids from

a common advanced intermediate, but the requisite diastereoselectivity during the

formation of the 1,3-cis-substituted tetrahydroisoquinoline could not be attained.

The results of our efforts to combine an efficient strategy for the synthesis of

mono- (lemonomycin) and bis-tetrahydroisoquinolines (saframycin B and renieramycin

G) will be discussed in the proceeding chapters.

38

1.7. REFERENCES

1) The chemistry and biology of tetrahydroisoquinoline alkaloids has been extensively reviewed in following review and references 2 through 10: Williams, R. M.; Scott, J. D. Chem. Rev. 2002, 102, 1669. 2) Kubo, A.; Arai, T. The Alkaloids; Brossi. A., Ed.; Academic Press: New York, 1983; Vol. 21, p 55. 3) Remers, W. A. The Chemistry of Antitumor Antibiotics; Wiley: New York, 1988, Vol. 2, p 93. 4) Remers, W. A. The Chemistry of Antitumor Antibiotics; Wiley: New York, 1988, Vol. 2, p 120. 5) Arai, T. Journal of Chromatography Library: Natural Product Isolation; Wagman, G. H., Cooper, R., Eds.; Elsevier: New York, 1989; Vol. 43, p 191. 6) Kubo, A.; Saito, N. Studies in Natural Products Chemistry; Elsevier: New York, 1992; Vol. 10, p 77. 7) Fukuyama, T. Adv. Heterocycl. Nat. Prod. Synth. 1992, 2, 189. 8) Katoh, T.; Terashima, S. Studies in Natural Products Chemistry; Elseveir: New York, 1997; Vol. 19, p 289. 9) Ozturk, T. The Alkaloids; Brossi, A., Ed.; Academic Press: New York, 2000; Vol. 53, p 119. 10) Rinehart, K. L. Med. Res. Rev. 2000, 20, 1. 11) Sygusch, J.; Brisse, F.; Hanessian, S.; Kluepfel, D. Tetrahedron Lett. 1974, 15, 4021. 12) Kluepfel, D.; Baker, H. A.; Piattoni, G.; Sehgal, S. N.; Sidorowicz, A.; Singh, K.; Vezina, C. J. Antibiot. 1975, 28, 497. 13) Zmijewski, M. M., Jr.; Goebel, M. J. Antibiot. 1982, 35, 771. 14) Hayashi, T.; Noto, T.; Nawata, T.; Okazaki, H.; Sawada, M; Ando, K. J. Antibiot. 1982, 35, 771. 15) Tomita, F.; Takahashi, K.; Shimizu, K. J. Antibiot. 1983, 36, 463.

39

16) Ikeda, T.; Idemoto, H.; Hirayama, F.; Yamamoto, K.; Iwao, K. Asao, T.; Munakata, T. J. Antibiot. 1983, 36, 1279. 17) Suzuki, K.; Sato, T.; Morioka, M.; Nagai, K.; Abe, K.; Yamaguchi, H.; Saito, T. J. Antibiot. 1991, 44, 479. 18) Bernan, V. S.; Montenegro, D. A.; Korshalla, J. D.; Maiese, W. M.; Steinberg, D. A.; Greenstein, M. J. Antibiot. 1994, 47, 1417. 19) Whaley, H. A.; Patterson, E. L.; Dann, M.; Shay, A. J.; Porter, J. N. Antimicrob. Agents Chemother. 1964, 8, 83. 20) He, H.; Shen, B.; Carter, G. T. Tetrahedron Lett. 2000, 41, 2067. 21) Frincke, J. M.; Faulkner, D. J. J. Am. Chem. Soc. 1982, 104, 265. 22) He. H.; Faulkner, D. J. J. Org. Chem. 1989, 54, 5822. 23) Davidson, B. S. Tetrahedron Lett. 1992, 33, 3721. 24) Parameswaran, P. S.; Naik, C. G.; Kamat, S. Y.; Pramanik, B. N. Ind. J. Chem. 1998, 37B, 1258. 25) Original isolation: Rinehart, K. L.; Holt, T. G.; Fregeau, N. L.; Stroh, J. G.; Kieffer, P. A.; Sun, F.; Li, L. H.; Martin, D. G. J. Org. Chem. 1990, 55, 4512. 26) Structural revision: Rinehart, K. L.; Holt, T. G.; Fregeau, N. L.; Stroh, J. G.; Kieffer, P. A.; Sun, F.; Li, L. H.; Martin, D. G. J. Org. Chem. 1991, 56, 1676. 27) Phase I clinical data, see: Ryan, D. P.; Supko, J. G.; Eder, J. P.; Seiden, M. V.; Demetri, G.; Lynch, T. J.; Fischman, A. J.; Davis, J.; Jieno, J.; Clark, J. W. Clin. Cancer Res. 2001, 7, 231. 28) Rinehart, K. L. Med. Res. Rev. 2000, 20, 1. 29) Myers, A. G.; Plowright, A. T. J. Am. Chem. Soc. 2001, 123, 5114. 30) Pastrone, I.; Viale, M; Cafaggi, S.; Mariggio, M. A.; Parodi, A.; Esposito, M. Invest. New Drugs 1999, 16 (4), 297. 31) Martoli, M. H.; Boitard, M.; Fessi, H.; Beriel, H.; Devissaguet, J. P.; Picot, F.; Puisieux, F. J. Microencapsul. 1990, 7 (2), 191.

40

32) Martello, L. A.; McDaid, H. M.; Regl, D. L.; Yang, C. P.; Meng, D.; Pettus, T. R.; Kaufman, M. D.; Arimoto, H.; Danishefsky, S. J.; Smith, A. M.; Horwitz, A. B. Clin. Cancer Res. 2000, 6, 1978. 33) Dorr, R. T.; Liddil, J. D.; Trent, J. M.; Dalton, W. S. Biochem. Pharmacol. 1987, 36 (19), 3115. 34) Kohn, H.; Na, Y.; Li, V.-S.; Nakanishi, Y.; Bastow, K. F. J. Med. Chem. 2001, 44, 3453. 35) Viale, M.; Vannozzi, M. O.; Merlo, F.; Cafaggi, S.; Parodi, B.; Esposito, M. Eur. J. Cancer 1996, 32 (13), 2327. 36) Ying, Y.; Qiu-Jun, L.; Qing-You, D., Bing-Hu, Y.; Ru-Xian, L.; Sheng-Qi, W. World J. Gastroenterol 2005, 11 (16), 2491. 37) Aune, G. J.; Furuta, T.; Pommier, Y. Anti-Cancer Drugs 2002, 13, 545. 38) Pommier, Y.; Kohlhagen, G.; Bailly, C.; Waring, M.; Mazumder, A.; Kohn, K. W. Biochemistry, 1996, 35, 13303. 39) Bernan, V. S.; Montenegro, D. A.; Korshalla, J. D.; Maiese, W. M.; Steinberg, D. A.; Greenstein, M. J. Antibiot. 1994, 47, 1417. 40) Mikami, Y.; Yazawa, K.; Takahashi, K.; Arai, T.; Namikoshi, M.; Iwasaki, S.; Okuda, S. J. Biol. Chem. 1985, 260, 334. 41) Zmijewski, M. J., Jr.; Mikolajczak, M.; Viswanatha, V.; Hruby, V. J. J. Am. Chem. Soc. 1982, 104, 4969. 42) Zmijewski, M. J., Jr.; Palaniswamy, V. A.; Gould, S. J. J. Chem. Soc., Chem. Comm. 1985, 1261. 43) Palaniswamy, V. A.; Gould, S. J. J. Am. Chem. Soc. 1986, 108, 5651. 44) Fukuyama, T.; Schleben, R. A. J. Am. Chem. Soc. 1982, 104, 4957. 45) Kubo, A.; Saito, N.; Yamato, H.; Masubuchi, K.; Nakamura, M. J. Org. Chem. 1988, 53, 4295. 46) Fukuyama, T.; Yang, L.; Ajeck, K. L.; Sachleben, R. A. J. Am. Chem. Soc. 1990, 112, 3712. 47) Williams, R. M.; Jin, W.; Metobo, S. Org. Lett. 2003, 5, 2095.

41

48) Corey, E. J.; Gin, D. Y.; Kania, R. S. J. Am. Chem. Soc. 1996, 118, 9202. 49) Corey, E. J; Martinez, E. J. Org. Lett. 1999, 1, 75. 50) Fukuyama, T.; Linton, S. D.; Tun, M. M. Tetrahedron Lett. 1990, 31, 5989. 51) Faulkner, D. J.; Frincke, J. M. J. Am. Chem. Soc. 1982, 104, 265. 52) Faulkner, D. J.; He, H. J. Org. Chem. 1989, 54, 5822. 53) Danishefsky, S. J.; Chan, C.; Heid, R.; Zheng, S.; Guo, J.; Zhou, B.; Furuuchi, T. J. Am. Chem. Soc. 2005, 127, 4596. 54) Pettit, G. R.; Knight, J. C.; Collins, J. C.; Herald, D. L.; Pettit, R. K.; Boyd, M. R.; Young, V. G. J. Nat. Prod. 2000, 63, 793. 55) Kubo, A.; Saito, B.; Sakai, H.; Suwanborirux, K.; Pummangura, S. Heterocycles 2001, 55, 21. 56) Danishefsky, S. J.; Harrison, P. J.; Webb, R. R. II; O’Neill, B. T. J. Am. Chem. Soc. 1985, 107, 1421. 57) Noyori, R.; Fujii, A.; Hashiguchi, S.; Uematsu, N.; Ikariya, T. J. Am. Chem. Soc. 1996, 118, 2521. 58) Bobbitt, J. M.; Moore, T. E. J. Org. Chem. 1968, 33, 2958. 59) Danishefsky, S. J.; Guo, J.; Zhou, B. Tetrahedron Lett. 2000, 41, 2043. 60) Stoltz, B. M.; Cruz, E. G.; Ashley, E. R. J. Am. Chem. Soc. 2003, 125, 15000. 61) Garner, P.; Ho, W. B.; Shin, H. J. Am. Chem. Soc. 1993, 115, 10742. 62) Joule, J. A.; Yates, N. D.; Peteres, D. A.; Allway, P. A.; Beddeoes, R. L.; Scopes, D. I. C. Heterocycles 1995, 40, 331. 63) Fukuyama, T.; Rikimaru, K.; Mori, K.; Kan, T. Chem. Comm. 2005, 394. 64) Fukuyama, T.; Endo, A.; Yanagisawa, A.; Abe, S.; Toma , T. K. J. Am. Chem. Soc. 2002, 142, 6552. 65) Fukuyama, T.; Mori, K.; Rikimaru, K.; Kan, T. Org. Lett. 2004, 6, 3095.

42

Chapter 2: Toward the Synthesis of Saframycin B

2.0. INTRODUCTION

In our retrosynthetic analysis of the tetrahydroisoquinoline alkaloids we plan to

form the first stereocenter at C1 early in the synthesis via an enantioselective alkylation

of an isoquinoline derivative 128 (Scheme 2.01). The stereoselective reduction of the

3,4-olefin 127 will be essential for obtaining the requisite 1,3-cis-substituted

tetrahydroisoquinoline core 126. We anticipate the C1 substituent will provide steric bulk

for facial selectivity during reduction. Saframycin B (1b) was chosen as our initial target

because of the simplicity of the structure, lacking oxidation at C20, C14, C4, and

possessing the pyruvamide side-chain. The alkylation and reduction chemistry will be

investigated with isoquinoline, a 3-substituted isoquinoline, and a substituted

isoquinoline representative of the real system.

OMe

MeOO

NN

OOMe

Me

OMe

H

H

H

NHCOCOMe

H

1

11

13

314

4

21NR2

YH

H

X

NR2Y

H

X

NY

1

3

1

3

1

3

R1

R1 R1

Scheme 2.01. Synthetic Approach Toward Saframycin B

126

127128

1b

43

2.1. BACKGROUND: ENANTIOSELECTIVE ALKYLATION OF ISOQUINOLINES

The alkylation of isoquinolines at C1 is a well documented reaction.1 When

treated with a strong organometallic nucleophile (130) isoquinoline (129) is alkylated

solely at the C1 position giving 131 (Scheme 2.02). In theory, if one shifts the aromatic

resonance to the pyridine portion of isoquinoline (132), one can envision alkylation at the

C3 position. However, this is not observed because the alkylated product 133 would no

longer possess an aromatic ring. The activation energy necessary to alkylate the C3 center

is significantly higher than alkylation at the C1 center.

N M X+ N

X

M

NM

N M XX+

Scheme 2.02. C1 Selectivity in Isoquinoline Alkylation

The stereoselective alkylation of an isoquinoline at C1 has been demonstrated on

few substrates and with limited success. In contrast, the enantioselective alkylation of

3,4-dihydroisoquinolines is much more well studied and will not be discussed here.2 One

of the oldest and well studied methods to alkylate isoquinolines is the Reissert reaction.3

Treatment of isoquinoline with an acid chloride in the presence of a cyanide source

generates the 1-cyano-1,2-dihydroisoquinoline 135 via the N-acylisoquinolinium

activated intermediate 134 (Scheme 2.03). Shibasaki and co-workers reported an

129 130

132 133

131

130

44

enantioselective Reissert reaction by catalyzing the alkylation in the presence of a chiral

lewis acid 137 (Scheme 2.03).4 The alkylation with 3-methylisoquinoline 136 under

optimized conditions gave the 1,2-dihydroisoquinoline 138 in 99% yield and 77% ee. The

Reissert reaction is a viable method for installation of the C1 stereocenter, and there is

potential for the formation of the requisite aminomethyl side-chain upon reduction.

However a drawback of this method is the use of an acid chloride which generates an

amide, a rather robust functional group, which could be potentially difficult to remove.

NKCN, AcCl

N Me

O

N Me

OCN

CN

N

Me TMSCN, AcCl

137, CH2Cl2N

MeO

N

Me

Me

OCN

OO

P(O)Ph2

P(O)Ph2

AlCl

Me3Si CN

Scheme 2.03. Variants of the Reissert Reaction

Reissert Reaction

Enantioselective Reissert Reaction

135

134

138 99% yield, 77% ee

137

136

129

45

Another method for the alkylation of activated isoquinolines involves the use of a

chiral acid chloride or chloroformate. Comins and co-workers have developed an

efficient method for the diastereoselective addition of Grignard reagents to homochiral 1-

acylpyridinium salts.5 Comins published one paper on the application of this

methodology to isoquinolines.6 Isoquinoline was treated with (−)-8-phenylmenthyl

chloroformate, forming a chiral N-acylisoquinolinium. Addition of MeMgI gave an 80%

yield of the alkylated product 139 with 60% de under optimized conditions (Scheme

2.04). This route is potentially attractive because the more readily cleaved carbamate is

formed rather than an amide. However, there is little potential for greater than 60% ee

and the alkylations are reported to work for Grignards but not alkyllithium nucleophiles.

Organolithium nucleophiles are reported to alkylate at the acyliminium carbonyl not at

the C1 imine carbon.7

N1) R*OCOCl, PhMe/THF, −23 °C

2) MeMgI

R* = (−)-8-phenylmenthyl

N

MeCO2R*

Scheme 2.04. Comins’ Diastereoselective Alkylations

139 80% yield, 60% de

129

46

The use of a chiral auxiliary is another potential option for the enantioselective

alkylation of isoquinolines. Marazano and co-workers reported modest

diastereoselectivity in the alkylation of the isoquinolinium salt 140 (Scheme 2.05).8 The

addition of RMgI formed the 1-substituted-1,2-dihydroisoquinoline 141 in 32-57% yield

and 38-90% de. The modest yield and diastereoselectivity make this route less attractive.

In addition, formation of the chiral salt 140 required a three step procedure from

isoquinoline.

NPhOH(CH2)11

OSO3

RMgI, THF

0 °CN

PhOH

HH

R

Scheme 2.05. Chiral Isoquinolinium Salts

A recent publication by the Alexakis group offered the most general method to

enantioselectively alkylate isoquinolines.9 Alexakis and Amiot applied known

methodology used for the enantioselective alkylation of acyclic and cyclic imines to

isoquinolines.2 They reported an alkylation of isoquinoline with an alkyllithium

nucleophile in the presence of a chiral bidentate ligand, however the initial alkylated

product could not be isolated. Alexakis observed that treatment of isoquinoline with

MeLi resulted in the rearomatised product 142 upon workup (Scheme 2.06) and not the

1,2-dihydroisoquinoline 143. In order to obtain the 1,2-dihydroisoquinoline the

intermediate alkylated product 146 was acylated with methyl chloroformate, however

acylation occurred at both C2 and C4 forming a mixture of 144 and 145. After MeLi

addition to isoquinoline the N2-lithio 146 is in equilibrium with the C4-lithio 147 which

141 32-57% yield, 38-90% de

140

47

142

is then acylated. The resulting nucleophilic nitrogen is acylated a second time to give

intermediate 148 which can be readily deprotonated to generate the diacylated product

145.

N N

MeLi

N

Me

Li

MeLi

Et2O25 °C

N

Me

air

ClCO2Me

N

MeCO2Me N

Me

CO2Me

+CO2Me

N

Me

MeCO2 H

CO2Me

NH

Me

Scheme 2.06. Alexakis Alkylation of Unactivated Isoquinoline

It was observed that the alkylation of isoquinoline with MeLi was catalyzed by

the addition of a bidentate ligand such as dimethoxyethane. Furthermore, the use of a

chiral bidentate ligand, namely (−)-sparteine (149) (Table 2.01), resulted in

enantioselective alkylation. The results are summarized in Table 2.1.

143

145144

147

148

146129

48

N

i) MeLi, solventtemp., 149

+ii) ClCO2Me

N N

entry solvent temp. (°C)

equiv. of 149 yield (%) ratio

144/145 ee (%)

144

1 PhMe −40 1 77 75/25 42

2 PhMe −40 0.25 89 70/30 36

3 Et2O −40 0.2 76 70/30 23

4 Et2O −78 0.2 7 70/30 21

5 PhMe −78 0.2 54 70/30 29

6 PhMe −10 0 45 80/20 N/A

Table 2.01. Alexakis’ Enantioselective Alkylation of Isoquinoline7,9

The shortcomings of this approach include modest ee’s, inseparable mixtures of

mono- and di-acylated products and only a limited number of nucleophiles were

investigated (MeLi, n-BuLi, and PhLi). Although the reaction proved catalytic for

enantioselective alkylation (entries 2-5), variance of solvent and temperature did very

little to improve the ee. The alkylations with PhLi and n-BuLi gave similar results to

MeLi. The Alexakis group was able to prove through further experiments that the

addition of MeLi occurred from the top face, producing the desired absolute

stereochemistry at C1 in agreement with the tetrahydroisoquinoline alkaloid side-chain.

We hoped to improve upon this methodology with the use of a more

functionalized nucleophile, improving the overall yield of mono-acylated product and

increasing the ee for alkylation of unactivated isoquinolines. The tetrahydroisoquinoline

144 145

149 129

49

alkaloids have a C1 side-chain with a methylene then a heteroatom (N or O), suggesting

that our organolithium nucleophile should have the general structure Li-CH2-X. An initial

search in the literature found that Li-CH2-SPh (phenylthiomethyllithium or

lithiothioanisole, 150) was a well investigated nucleophile suitable for our requirements.

Phenylthiomethyllithium (151) can be generated by treatment of thioanisole with

n-BuLi in diethyl ether with stirring for 15 hours (Table 2.02). The lithiation takes place

on the aromatic ring as well as the methyl group generating a mixture of products.10 The

aryl-lithiated products 152 slowly equilibrate to the more thermodynamically favored

methyl-lithiated product over a period of 15 h. Corey and Seebach reported that under

similar conditions in the presence of a tertiary diamine, 1,3-diazabicyclo[2.2.2]octane 153

(DABCO), phenylthiomethyllithium was formed in 45 minutes.11 Peterson observed

similar results when tetramethylethylenediamine (TMEDA) was employed instead of

DABCO.12

S n-BuLi

Et2O

S

Li

S Li+

observed product composition (mole%) time

(min.) 153

(equiv.) methyl ortho and meta para

5 0 63 37 trace

1 0 90 9 trace

15 0 96 4 trace

45 min. (in THF) 1 98.5 trace trace

Table 2.02. Thioanisole Metalation

NN

150 153 151 152

NN

50

129

A further literature search revealed, to the best of our knowledge, that

(−)-sparteine has never been used in the lithiation of thioanisole. It was our hope to

combine the isoquinoline alkylation work of Alexakis and lithiation results of Corey to

enantioselectively alkylate isoquinoline with phenylthiomethyllithium formed in the

presence of (−)-sparteine (Scheme 2.07).

S

n-BuLi, THFS Li

N NN

NN

N

PhS

Li

ClCO2MeN

PhS

CO2Me

Scheme 2.07. Planned Enantioselective Alkylation of Isoquinoline

149

150 154

156155

51

2.2. RESULTS AND DISCUSSION

2.2.1. INVESTIGATION OF THE ENANTIOSELECTIVE ALKYLATION OF ISOQUINOLINE

During our initial investigation into the alkylation of isoquinoline with

phenylthiomethyllithium we chose to use the known methods of DABCO and TMEDA to

generate the lithiated nucleophile. When a solution of isoquinoline in toluene at −78 °C

was treated with phenylthiomethyllithium the alkylation proceeded slowly (Table 2.03).

Stirring at −20 °C for more than 24 hours was needed for greater than 80% conversion,

when either DABCO or TMEDA were used. A methyl chloroformate quench gave a

mixture of mono-acylated product 157 and di-acylated product 158, as observed by

Alexakis. In contrast to Alexakis’ results, these products could be separated and

characterized. When (−)-sparteine was used in place of DABCO/TMEDA the results

were similar although the yield of diacylated product was slightly lower. In an effort to

minimize the reaction time the reaction was allowed to warm to room temperature. It was

observed that the starting isoquinoline was rapidly consumed when warmed to room

temperature and upon quenching with methyl chloroformate only the mono-acylated

product 157 was observed.

These results suggest that the intermediate alkylated product, which exists in an

equilibrium as the N2-lithio or C4-lithio adducts, is temperature dependent. The C4-litho

product is either less stable at room temperature or the N2-lithio adduct is significantly

more reactive at room temperature such that none of the C4 acylated product is observed.

52

N N

PhS

CO2MeN

PhS

CO2Me

CO2Me

+

1) PhSCH2Li (1.5 equiv.), PhMe, diamine, temp., time

2) ClCO2Me

diamine temp. (°C) time yield (%) 157 yield (%) 158

DABCO −78 to −20 1 day 33 52

TMEDA −78 to −20 2 days 39 28

(−)-sparteine −78 to −20 1 day 45 10

(−)-sparteine −78 to −40 3 days 56 12

(−)-sparteine −78 to 25 1 h 91 0

Table 2.03. Alkylation of Isoquinoline with Phenylthiomethyllithium

With the success of the alkylation of isoquinoline with phenylthiomethyllithium

in the presence of (−)-sparteine we next had to determine if the alkylation was

enantioselective. We first attempted to resolve enantiomers in the 1H NMR using a

europium based chiral shift reagent, but this was unsuccessful. Instead, analytical HPLC

with a chiral column (Diacel, Chiracel OD) was employed and the 1:1 mixture of

enantiomers was readily separable. The separation was also performed on the product

formed from the use of DABCO and identical results were observed. A variety of

conditions were employed; varying reaction temperature, equivalents of chiral ligand and

nucleophile, and solvent in an effort to achieve some level of enantioselectivity without

success.

157 158 129

53

In a different approach to the enantioselective alkylation of isoquinolines we

investigated the use of chiral sulfoxides. Sulfoxides are more readily lithiated at the

methyl position and chiral sulfoxides have shown diastereoselectivity in the alkylation of

imines.13 Methyl phenyl sulfoxide (159) was used as a model (Scheme 2.08), but the

reaction was not analogous to the phenylthiomethyllithium reaction. The starting

isoquinoline was consumed by TLC analysis and a variety of products were observed, but

upon methyl chloroformate quench none of the desired product 160 was isolated.

SO i) LDA, THF, −78 °C

ii) isoquinoline (129), −78 °C to 25 °C

iii) ClCO2Me

N

SPh

OCO2Me

Scheme 2.08. Isoquinoline Alkylation with Methyl Phenyl Sulfoxide

There are a number of potential side reactions stemming from use of the lithiated

methyl phenyl sulfoxide 161 that are less likely and hence not observed with thioanisole.

Assuming the initial alkylation was successful, the sulfoxide adduct 162 (Scheme 2.09)

would have α-methylene protons which are still quite acidic. This could lead to not only

an equilibrium with C4-lithiation but also lithiation at C22 (saframycin numbering) to

give 165. Once lithiation at C22 occurs α-elimination to form 166 would be possible.

Furthermore, sulfoxides are also known to β-eliminate14 and 164 could potentially form

from deprotonation at C1 of intermediate 163. All approaches to enantioselective

alkylation of isoquinoline with a sulfoxide were abandoned owing to the wide variety of

possible by-products.

159 160

54

129 N

SPh

OLi

N

SPh

O

Li

NH

SPh

OLi

HN

SPh

OLi

H

N

LiSO

LiN

22

Scheme 2.09. Potential Side-Reactions of Sulfoxide Alkylation

Our investigation into the enantioselective alkylation of isoquinoline with

phenylthiomethyllithium failed to show any facial selectivity, but we were able to

successfully install a functionalized C1 side-chain and minimize the amount of diacylated

product. At this point we decided further investigation into the formation of the

1,3-cis-substituted tetrahydroisoquinoline would prove more fruitful and a potential

reinvestigation of the enantioselective alkylation could be done at a later stage.

166165

162161

164

163

55

2.2.2. FORMATION OF 1,3-CIS-SUBSTITUTED TETRAHYDROISOQUINOLINE

To form the 1,3-cis-substituted tetrahydroisoquinoline we needed to establish a

method of reduction. As a model system we used the previously formed

1,2-dihydroisoquinoline 157. We anticipated potential difficulties with reduction of the

hindered di-substituted cyclic olefin (and tri-substituted for the real system). In addition it

was postulated that the presence of the sulfur could poison catalytic hydrogenation

methods.

Initial investigations found that palladium catalyzed hydrogenations were

unsuccessful for obtaining the reduced product. Wilkson’s catalyst has been used to

successfully reduce olefins in the presence of sulfides.15 For our substrate no reduction

was observed. We next turned our attention to ionic hydrogenation with the use of a

strong acid and a trialkylsilane. A recent publication by Molinski et al. gave promise for

the reduction of our substrate.16 It was reported that the reduction of enecarbamates 167

with TFA and triethylsilane, through the intermediate iminium 168, gave carbamates 169

in excellent yields (Scheme 2.10). The reaction is the acyclic equivalent to our substrate

and uses reagents that should be compatible with the sulfide side-chain.

HN

CO2Et

R

Et3SiH

TFA, −10 °C

HN

CO2Et

R

NCO2Et

R

HH

H SiEt3O2CCF3

94-99% yield

Scheme 2.10. Molinski’s Ionic Hydrogenation of Enecarbamates

169

168

167

56

Upon treatment of 1,2-dihydroisoquinoline 157 with triethylsilane and TFA clean

reduction of the 3,4-olefin was observed, to give the tetrahydroisoquinoline 170 in

excellent yield (Scheme 2.11).

NCO2Me

SPh

Et3SiH, TFA

CH2Cl2, 0 °C to 25 °C, 1 hN

CO2Me

SPh95% yield

Scheme 2.11. Reduction of 1,2-Dihydroisoquinoline 157

In order to establish the stereoselectivity for this reduction a 3-substituted

isoquinoline was necessary. The simple, commercially available 3-methyl isoquinoline

(171), was chosen for the model study. Treatment of 3-methyl isoquinoline with

phenylthiomethyllithium and methyl chloroformate quench as before, afforded the

desired product 172 in excellent yield (Scheme 2.12).

NCO2Me

SPh

N

90% yield

Me Me i) PhSCH2Li, (−)-sparteinePhMe, −78 °C to 25 °C, 1 h

ii) ClCO2Me

Scheme 2.12. Alkylation of 3-Methyl Isoquinoline

It should be noted that although enantioselectivity was not observed for the

alkylation of isoquinolines with phenylthiomethyllithium generated in the presence of

(−)-sparteine, these alkylations gave superior results than the DABCO or TMEDA

promoted alkylations. DABCO is a hygroscopic solid which made for tedious reaction

170

171

157

172

57

setup on a small scale and capricious reaction results were observed. With TMEDA more

by-products were observed during alkylation. (−)-Sparteine is readily purified by

distillation, it is not particularly expensive, and the best, reproducible results were

observed using (−)-sparteine as the diamine for phenylthiomethyllithium formation.

The reduction of 3-methyl-1,2-dihydroisoquinoline 172 was conducted with

Et3SiH/TFA as before, to give in excellent yield the reduced 173 as what appeared to be

one diastereomer by TLC (Scheme 2.13). Further confirmation of the structure by 1H

NMR analysis was difficult because strong carbamate resonance led to an observed

broadening of signals. However, upon performing a 1H NMR acquisition at 100 °C, the

signals sharpened and it was obvious that only one diastereomer was formed. A NOESY

experiment was conducted to determine the stereochemistry at the C3 center. Irradiation

of the C3 methyl protons showed diagnostic nuclear Overhauser enhancements of C22

methylene protons, as well as C3 and C4 protons, but no enhancement of the C1 proton

was observed. These results suggested that indeed we had formed the 1,3-cis-substituted

tetrahydroisoquinoline 173.

NCO2Me

NCO2Me

SPh

Me Et3SiH, TFA

CH2Cl2, 0 °C to 25 °C, 45 min.

87% yield

MeH

HH H

HPhSH

Scheme 2.13. Stereoselective Reduction of 3-Methyl-1,2-Dihydroisoquinoline 172

observed nOe’s of C3-methyl

172 173

58

The observed cis-relationship is due to the steric bulk of the C1 side-chain. The

reduction occurs on the iminium intermediate 172 (Scheme 2.14) which possesses four

sp2 hybridized carbons leading to a flat structure, as modeled by Chem3D (Figure 2.01).

The C1 side-chain exists in the axial orientation hindering hydride addition to the C3

iminium from the top face which would give the trans-isomer. Instead only the

cis-isomer is observed from hydride addition to the bottom face.

NCO2Me

SPh

Me

NCO2Me

SPh

MeHN

CO2Me

SPh

MeH SiEt3 H

H

Scheme 2.14. Ionic Hydrogenation of 172

Figure 2.01 Chem3D Representation of the N-Acyliminium Intermediate 174

172 173 174

59

We have successfully demonstrated an efficient route to 1,3-cis-substituted

tetrahydroisoquinolines from alkylation of unactivated isoquinolines with

phenylthiomethyllithium followed by methyl chloroformate quench to give the

1,2-dihydroisoquinoline. A stereoselective 3,4-olefin reduction by ionic hydrogenation

provides the 1,3-cis-substituted tetrahydroisoquinoline. Investigation of this chemistry

using a more functionalized 3-substituted isoquinoline which is representative of the real

system was required.

2.2.3. 3-(AMINOMETHYL)-ISOQUINOLINE

Moving to an isoquinoline system that closely resembles saframycin B we wanted

to investigate the alkylation and reduction of a 3-(aminomethyl)-isoquinoline 178. The

nitrogen is in the correct orientation to generate the piperazine ring system present in

saframycin B. In addition, we anticipated that the benzyl protecting groups would be

stable to the strong basic conditions used for alkylation and the strong acidic conditions

used for reduction.

The initial synthesis of 178 followed a literature preparation from 1994 (Scheme

2.15).17 The problems with this route included the high cost of starting material, the

number of steps involved, and inconsistent yields for reduction of the nitrile 176 were

observed. Improvements in the synthesis of 3-substituted isoquinolines were essential for

this route to be viable.

60

NOH

O1) ClCO2Et, TEA, THF

2) NH4OH3) TFAA, Et3N, CH2Cl2

95% yield

N

CN

H2, Pd/C

HCl, MeOH NH·HClNH2·HCl

48-78% yield

NNBn2

Scheme 2.15. Synthesis of 3-(Aminomethyl)-Isoquinoline

The solution came in a timely publication by Larock.18 He reported a very

efficient synthesis of 3-substituted isoquinolines 181 from ortho-iodo imines 179 and

various acetylenes 180 in a two step process (Scheme 2.16). The first step involved a

Sonagashira coupling of the aryl iodide 179 and acetylene 180 to form the coupled

product 182. After filtration and removal of triethylamine the coupled product was

redissolved in DMF and heated in the presence of catalytic CuI. The copper complex 183

cyclized to 184 and elimination formed the isoquinoline 181. Larock did not report the

synthesis of isoquinoline 178, nor did he demonstrate that this methodology could be

used with a nitrogenous acetylene.

176, $128/1

178 177

175, $78/1 g

61

I

N+ RH

1) 2% PdCl2(PPh3)2, 1% CuI Et3N, 50 °C

2) 10% CuI, DMF, 100 °CN

R

81-95% yield

N

R

N

RCuLn

N

RCu

H

HSonagashira

CuI

Scheme 2.16. Larock Isoquinoline Synthesis

To generate the required isoquinoline 178, ortho-iodo imine 179 was synthesized

from inexpensive ortho-iodo benzoic acid 185 (Scheme 2.17). Following published

procedures the acid was reduced to the corresponding alcohol with borane19 then oxidized

to the aldehyde 186 with PCC20 in 89% yield over the two steps. The Larock procedure

for imine formation was modified; the aldehyde was stirred with tert-butylamine in

pentane over 4Å molecular sieves for 12 hours at room temperature. Upon filtration and

concentration, a 92% yield of imine 179 was obtained with no aqueous workup or

distillation, as the excess tert-butylamine is removed under aspirator vacuum. The

standard Larock conditions were used with dibenzylpropargylamine21 (187) for the

Sonagashira coupling, to give in near quantitative yield the coupled product after two

hours at 55 °C. The copper catalyzed cyclization step proceeded slower than expected

and was not complete until after heating at 100 °C for 14 hours. Under optimized

conditions isoquinoline 178 was isolated in 84% from imine 179.

182

180181179

183 184

62

I

CO2H

I

O

H1) BH3·SMe2, THF, reflux

2) PCC, CH2Cl2, 25 °C

t-butylamine4Å molecular sieves

25 °C, 16 h

I

N

1) 2% PdCl2(PPh3)2, 1% CuI Et3N, 55 °C, 2 h

2) 10% CuI, DMF, 100 °C, 14 h NNBn2+

NBn2

89% yield92% yield

84% yield

Scheme 2.17. 3-(Aminomethyl)-Isoquinoline Formation

The alkylation of isoquinoline 178 with 1.5 equivalents of

phenylthiomethyllithium gave the desired 1,2-dihydroisoquinoline 188 in 86% yield upon

quenching with methyl chloroformate (Scheme 2.18). From ortho-iodo benzoic acid, a

six step sequence yielded the 1,2-dihydroisoquinoline 188 in 59% yield.

NNBn2

NCO2Me

SPh86% yield

NBn2

i) PhSCH2Li, (−)-sparteinePhMe, −78 °C to 25 °C, 30 min.

ii) ClCO2Me

Scheme 2.18. Alkylation of Isoquinoline 178

The ionic hydrogenation conditions which worked so effectively for the 3-methyl-

1,2-dihydroisoquinoline 172 gave only starting material when applied to 188 (Scheme

2.19). Treatment of 188 with Et3SiH and TFA, even under refluxing conditions, gave

mostly starting material with only slight degradation. This dramatic change in reactivity

can be attributed to the tertiary amine. The strong acidic conditions the nitrogen will be

185, $15/25 g 186

179 187 178

178 188

63

protonated such that in solution the starting material exists as the fully protonated

intermediate 189. In order for the reduction to occur a second protonation at the C4 center

must occur generating a di-cationic species 190. This di-cation may be too high in energy

to form under these conditions, hence reduction is not observed.

NCO2Me

SPh

NBn2 Et3SiH, TFA

CH2Cl2, reflux NCO2Me

SPh

NBn2H

H

NCO2Me

SPh

NBn2

H

NCO2Me

SPh

NBn2

H

H

H

H

Scheme 2.19. Attempted Ionic Hydrogenation of 1,2-Dihydroisoquinoline 188

In an attempt to circumvent this problem we planned to convert the tertiary amine

to a carbamate via a known modification of the Von Brun reaction.22 Benzyl amines can

be removed in a two step procedure by first reacting with a chloroformate to form a

carbamate, followed by hydrolysis to generate the debenzylated amine. It was anticipated

that treatment of amine 188 with phenylchloroformate in refluxing 1,2-dichloroethane

would promote debenzylation via N-acyliminium 192 to generate carbamate 193 which

we hoped would undergo ionic hydrogenation more readily.

190189

191 188

64

NCO2Me

SPh

NBn2 ClCO2Ph

DCE, reflux NCO2Me

SPh

NBn

CO2Ph

NN

PhSCO2Me

Cl

CO2PhPh

Ph

Scheme 2.20. Attempted Formation of Carbamate 193

Unfortunately the desired product was not observed. Initial inspection of

spectroscopic data revealed that one benzyl group had been removed and IR revealed a

new stretch consistent with a carbamate carbonyl. This seemingly correct product was

isolated after chromatography to separate it from unreacted starting material. Upon

reduction under ionic hydrogenation conditions the expected product could not be

verified. Difficulty with characterization ensued as a mixture of inseparable products was

isolated and carbamate resonance distorted room temperature NMR. An X-ray structure

analysis revealed the answer. The product isolated after reduction was a 6:1 mixture of

chloride 198 and methyl 173 (Scheme 2.22). The attempted debenzylation with

phenylchloroformate (Scheme 2.21) was not successful; chloride 194 and carbamate 195

were formed and could not be separated by chromatography. The apparent loss of the

benzyl methylene in the 1H NMR came from symmetrization as the starting material had

diastereotopic benzylic methylenes, and the new carbonyl IR stretch was from the

carbamate 195. The preference for allylic chlorination over benzylic chlorination is

potentially due to neighboring group participation. After acylation to generate the

188

192

193

65

N-acyliminium 196 the N2 carbamate can displace the quaternized amine. Chlorination of

the intermediate 197 would occur readily to give 194.

N

Bn2N CO2Ph

O

OMePhS

NCO2Me

SPh

NBn2

N

PhS

O

OMe

ClCO2Ph

DCE, reflux NCO2Me

SPh

Cl+ Bn2N

CO2Ph

Cl

58% yield

Scheme 2.21. Formation of Chloride 194

Reduction of chloride 194 with TFA and triethylsilane yielded the reduced

compound 198 and the over-reduced compound 173 (Scheme 2.22). The reduction

proceeds via the expected pathway of ionic hydrogenation and enecarbamate protonation

followed by hydride addition. The over reduced product was potentially formed from the

same oxazoline intermediate 197, derived as above from neighboring group participation

of the methyl carbamate. Hydride addition to oxazoline 197 forms the 3-methyl-1,2-

dihydroisoquinoline 172, and further reduction gives the previously synthesized

isoquinoline 173. The X-ray structure of 198 and 173 (appendix A) resolved the

confusion observed from analysis of spectroscopic data. In addition the X-ray data

proved unambiguously that the ionic hydrogenation stereoselectivity forms the

1,3-cis-substituted tetrahydroisoquinoline.

188 195 194

197196

66

NCO2Me

SPh

ClN

CO2Me

SPh

MeH

H

H

H

+

45% yield6:1, X-ray

Et3SiH, TFA

CH2Cl2

N

PhS

O

OMe

NCO2Me

SPh

Cl

N

PhS

Me

CO2Me

NCO2Me

SPh

Cl

H H

NCO2Me

SPh

MeH

H

H

Scheme 2.22. Possible Mechanism for Formation of Chloride 198

The reduction of 188 proved troublesome and an alternative route for the

synthesis of 1,3-cis-substituted tetrahydroisoquinoline 191 was sought. We turned our

attention to the synthesis of an oxygen analogue of 188 speculating that reduction would

occur more readily in the absence of the basic amine. It was anticipated that after

stereoselective reduction the oxygen atom could be converted to the requisite nitrogen

atom.

198

172

173194

197 174

199

67

2.2.4. OXYGEN ANALOGUE

The new route required synthesis of the requisite isoquinolines 201 and 203. The

preferred route would utilize isoquinoline 201 as it is anticipated that during the reduction

step the silyl protecting group will be lost to afford the reduced and deprotected alcohol,

ready for conversion to a nitrogen atom.

The isoquinoline synthesis proceeded well for both the triisopropylsilylpropargyl

ether (200) and the propargylbenzyl ether23 (202) to afford 201 and 203 in excellent yield

(Scheme 2.23). These isoquinolines were alkylated as before to give the

1,2-dihydroisoquinolines 204 and 205 in good yield (Scheme 2.24).

1) 2% PdCl2(PPh3)2, 1% CuI Et3N, 55 °C, 6 h

2) 10% CuI, DMF, 100 °C, 9 h NOBn

+OBn

84% yield

1) 2% PdCl2(PPh3)2, 1% CuI Et3N, 55 °C, 1 h

2) 10% CuI, DMF, 100 °C, 4 h NOTIPS

+OTIPS

94% yield

Scheme 2.23. Isoquinoline Synthesis

NOR

NCO2Me

SPh

i) PhSCH2Li, (−)-sparteinePhMe, −78 °C to 25 °C, 30 min. OR

ii) ClCO2Me

Scheme 2.24. 1,2-Dihydroisoquinoline Synthesis

203

200

202

179

201

179

204 R = TIPS, 89% yield205 R = Bn, 76% yield

201 R = TIPS 203 R = Bn

68

207, 29% yield

The ionic hydrogenation was first attempted with 204. Treatment of a solution of

204 and Et3SiH in CH2Cl2 with TFA at −10 °C led to immediate deprotection of the silyl

ether (Scheme 2.25). Upon warming to 25 °C reduction was observed as well as the

formation of a by-product. Reduction gave the desired 1,3-cis-substituted

tetrahydroisoquinoline 206 as a single diastereomer in 53% yield and isoquinoline 207 in

29% yield. After silyl ether deprotection to form 208 an acid catalyzed acyl migration can

lead to the formation of by-product 207. Protonation of the methyl carbamate 208 gives

209 which can undergo hydroxyl addition to form intermediate 210 followed by a ring

opening to generate 211. The 1,2-dihydroisoquionoline 211 can readily undergo air

oxidation to form isoquinoline 207.

NCO2Me

SPh

OTIPSEt3SiH, TFA

NCO2Me

SPh

OH

NH

H

+ OCO2Me

−10 °C to 0°C1 h

CH2Cl2

NCO2Me

SPh

OH

−TIPS

NOH

PhSO

OMe

H

N

PhS

O

OHOMe

NH

PhS

OCO2Me

H

PhS

[O]

Scheme 2.25. Reduction of Silyl Ether 204

211 208

209 210

204

206, 53% yield

69

In dramatic contrast to the triisopropylsilyl ether 204, the benzyl ether analogue

205 proved more stable during reduction. When enecarbarmate 205 was subjected to the

same ionic hydrogenation conditions clean reduction was observed to give a single

diastereomer 212 in excellent yield (Scheme 2.26). As with previous reductions the

1,3-cis relative stereochemistry was based on the analogy with earlier

tetrahydroisoquinoline formation.

NCO2Me

SPh

OBn Et3SiH, TFA

CH2Cl2, −10 °C to 0°C, 3 h NCO2Me

SPh

OBnH

H

97% yield

Scheme 2.26. Reduction of Benzyl Ether 205

We now wanted to investigate the reactivity of the C1 and C3 substituents toward

each other to confirm their 1,3-cis relationship. To explore this reactivity we chose to

form a Pummerer intermediate. Treatment of sulfide 212 with N-chloro-succinimide 214

forms the α-chloro-sulfide 218 (Scheme 2.27). Addition of SnCl4 generates the reactive

thionium ion 217, the Pummerer intermediate, which quickly cyclizes to 219 followed by

debenzylation to afford 213 as a single diastereomer in 74% yield. The cyclized product

213 was isolated as a crystalline white solid and an X-ray structure (appendix B)

confirmed the formation of the bicyclo[3.3.1] system.

212205

70

NCO2Me

SPh

OBnH

H

1) NCS, PhCl, 2 h, 25 °C

2) SnCl4, 0 °C, 5 min.

N

ClO

O

NCO2Me

SPh

OBnH

H

Cl

H

N

PhS

OBnH

H

NOO

NO

CO2Me

PhS

NO

CO2Me

SPh

SnCl5

Ph

74% yield

Cl

N

PhS

OBnH

H

Cl

CO2Me

CO2Me

N

PhS

OBnH

H

CO2Me

SnCl4

Scheme 2.27. The Pummerer Cyclization

The cyclization introduced another potential route to the tetrahydroisoquinolines.

The structure of the cyclization product is that of a bicyclo[3.3.1] system, which is very

similar to that found in the saframycins except for the presence of the oxygen atom

instead of a nitrogen atom (Figure 2.02). The pseudo-symmetry of the saframycins makes

our strategy to synthesize the 1,3-cis-substituted tetrahydroisoquinoline potentially

applicable to both the western and eastern fragments of saframycin B. Although we

initially planned to synthesize the western fragment first we hoped to apply this

cyclization methodology to the nitrogen analogue, to form the 1,4-diaza-

bicyclo[3.3.1]system found in saframycin B (Scheme 2.28).

212

218

219

217

214

213, X-ray

217215

216

71

OMe

MeOO

NN

OOMe

Me

OMe

H

H

H

NHCOCOMe

H

OMe

OMe

O

NN

OMeO

MeO Me

H

H

H

NHCOCOMe

H

NO

CO2Me

SPh

Figure 2.02. Pseudo-Symmetry of Saframycin B

NCO2Me

SPh

NBn2H

H

R

NNBn

CO2Me

SPh

R

Scheme 2.28. Nitrogen Analogue to Form Bicyclo[3.3.1] System

1b

213

Saframycin B (1b)

221220

72

2.2.5. NITROGEN ANALOGUE FOR PUMMERER CYCLIZATION

To investigate the Pummerer cyclization on the nitrogen analogue conversion of

the oxygen atom was required. Previously, alcohol 206 was successfully generated from

simultaneous reduction and deprotection of silyl ether 204. However, modest yields for

this product were observed due to by-product formation. The benzyl ether proved to be a

more efficient route to alcohol 206. Ionic hydrogenation gave tetrahydroisoquinoline 212,

and benzyl ether deprotection was promoted by treatment with BCl3 to give alcohol 206

in 97% yield (Scheme 2.29). This two step procedure to form alcohol 206 was more

efficient than the one step procedure from silyl ether 204. The alcohol was then converted

to the tosylate 222 in 86% yield using standard conditions.

NCO2Me

SPh

OBnH

HBCl3, CH2Cl2

−78 °C to 25 °C, 30 min.

0 °C to 25 °C, 4 h

95% yield

NCO2Me

SPh

OHH

H

Me

SO2Cl

pyridine, CH2Cl2

86% yield

NCO2Me

SPh

OTsH

H

Scheme 2.29. Tosylate Formation

222

212 206

73

Treatment of tosylate 222 with sodium azide in DMF at 55 °C gave only modest

yields of the desired azide 223 (Scheme 2.30) and the by-product 224 in 31% yield. The

formation of this by-product was likely to have come from a similar neighboring group

participation as previously observed. Heating the tosylate 222 in DMF could lead to an

intramolecular displacement of the tosylate generating intermediate 225. Azide can then

demethylate the activated intermediate to give the oxazolidinone 224. It should be noted

that without heating no reaction occurs between sodium azide and tosylate 222.

NCO2Me

SPh

OTsH

H

NCO2Me

SPh

N3H

H

N

SPh

H

HNaN3, DMF

55 °C, 12 hO

O

N

PhS

TsO

H

H

O

OMe

N

PhS

H

H

O

O Me

N3

+

Scheme 2.30. Azide Formation

A more efficient synthesis of the azide was found using modified Mitsunobu

conditions.24 Treatment of alcohol 206 with diethylazodicarboxylate (DEAD),

triphenylphosphine, and diphenylphosphorylazide gave azide 223 (Scheme 2.31). Under

these conditions, which did not require heating, the oxazolidinone product 224 was not

observed.

223, 65% yield

225

222 224, 31% yield

74

(PhO)2PON3, DEAD

PPh3, THF0 °C to 25 °C, 2 h

NCO2Me

SPh

OHH

H

NCO2Me

SPh

N3H

H

82% yield

Scheme 2.31. Efficient Azide 223 Formation

With azide 223 in hand the initial plan was to investigate the Pummerer

cyclization which involved reduction of the azide to the amine followed by dibenzylation.

However, a thorough investigation of the Pummerer cyclization directly with azide 223

gave interesting results.

Azide 223 was treated under identical Pummerer cyclization conditions as benzyl

ether 212, NCS followed by SnCl4 (Scheme 2.32). The evolution of gas was observed and

the reaction was complete upon initial TLC analysis. After aqueous workup and

chromatography the unstable iminosulfide 226 was isolated in 49% yield. The

spectroscopic data appeared consistent with the cyclized product. The mechanism for this

reaction is outlined in Scheme 2.32 and proceeds through an analogous Pummerer

intermediate 227 as before. The azide cyclizes to form intermediate 228 which can

undergo rapid and irreversible loss of nitrogen gas to generate the iminosulfide 226. It

was observed that iminosulfide 226 readily hydrolyzed to lactam 229 under acidic

conditions. Treatment of the crude cyclized product with 5% aq. sodium hydrogen sulfate

rapidly formed the lactam 229 (Scheme 2.33), which was characterized by X-ray

crystallography (appendix C).

206 223

75

NCO2Me

SPh

N3H

H

1) NCS, PhCl, 2 h, 25 °C

2) SnCl4, 0 °C, 5 min.

49% yield

NN

CO2Me

SPh

NCO2Me

S

NH

H

Ph

N NN

N

CO2Me

PhSH

N2

SnCl5

Scheme 2.32. Azide Cyclization

NN

CO2Me

SPh

5% aq. NaHSO4

Et2O, 5 min.

>95% yield

NNH

CO2Me

O

Scheme 2.33. Lactam Formation

This lactam formation was unprecedented and the cyclization of an azide with a

Pummerer intermediate has, to the best of our knowledge, never been reported. In

addition cyclization gave the desired 1,4-diazabicyclo[3.3.1] system. Application of this

methodology to the real system was necessary to determine its potential utility in the

synthesis of saframycin B.

229, X-ray 226

226

228227

223

76

2.2.6. FORMATION OF THE REAL SYSTEM

Moving away from a model system necessitated the formation of a substituted

analogue of the bicyclo[3.3.1] system 229. To achieve this goal we required the synthesis

of hexasubstituted benzene 234 (Scheme 2.34). The arylbenzyl ether was important as it

provides easy access to the phenol, prior to oxidation to the quinone in the final steps of

the synthesis of saframycin B. Oxidation to the quinone has been shown to proceed in

higher yields from a phenol than from an aryl ether.25

In our first approach to the formation of 234 we started with commercially

available aldehyde 230 which was converted to phenol 231 via a known method of

Baeyer-Villiger oxidation, followed by formate ester hydrolysis.26 Phenol 231 was

formylated under Duff conditions27 and protected as the benzyl ether to give aldehyde

233. The installation of the aryl iodide was attempted with an electrophilic iodine source,

iodine, NIS, or AgO2CCF3/I2, but only complex reaction mixtures were isolated.

Me

MeOOH

OMe

Me

MeOOBn

OMe

O

N

NN

N

AcOH, CH2Cl2, 100 °C

1)

2) BnBr, Na2CO3, DMF

Me

MeOOBn

OMe

O

I

Me

MeOCHO

OMe

1) m-CPBA, CH2Cl2

2) NaOH, MeOH

>95% yield 48% yield

Scheme 2.34. Attempted Formation of Hexasubstituted Benzene 234

233 234

230 231

232

77

Further examination of the literature showed that ortho-iodination of similar

aldehydes was not known. More common was a two step procedure utilized by Suzuki in

his work on a similar benzaldehyde.28 A solution of phenol 231 and paraformaldehyde in

CH2Cl2 was treated with neat diethylaluminum chloride to afford diol 235 (Scheme 2.35).

The hydroxymethylated product was formed very cleanly but found to be unstable at

room temperature. The crude phenol was immediately protected as the arylbenzyl ether to

give alcohol 236 in 89% yield over the two steps. The alcohol 236 underwent facile aryl

iodination with silver trifluoroacetate and iodine to give iodide 237 in 82% yield.

Oxidation of the primary alcohol to the aldehyde with PCC gave the desired

hexasubstituted benzene 234 in 98% yield. This procedure provided a high yielding and

scaleable route to the benzaldehyde required for isoquinoline formation.

Me

MeOOH

OMe

OH

Me

MeOOH

OMe

(CH2O)n, Et2AlCl

CH2Cl2, 0 °C to 25 °C

Me

MeOOBn

OMe

OH

BnBr, K2CO3

acetone, 60 °C

Me

MeOOBn

OMe

OH

IAgOOCCF3

I2, CHCl3

Me

MeOOBn

OMe

CHO

IPCC

CH2Cl2

89% yieldtwo steps

82% yield 98% yield

Scheme 2.35. Synthesis of Hexasubstituted Benzene 235

237236

235231

234

78

With aldehyde 234 in hand we turned our attention to the synthesis of

isoquinoline 240. The imine 238 was formed as before in quantitative yield (Scheme

2.36) and then treated with acetylene 202 using the Larock isoquinoline procedure.18

Unfortunately, the reaction did not proceed smoothly. A mixture of hydrolyzed starting

material 234, hydrolyzed coupled product 239, and desired isoquinoline 240 were

isolated after aqueous workup. In previous isoquinoline formation, upon addition of the

palladium and heating to promote the Sonagashira coupling the solution became pale

yellow in color with a fluffy precipitate forming (triethylamine hydroiodide). However

for the substituted imine 238 the reaction appeared black in color and a small amount of

black solid formed suggesting that palladium(0) was precipitating out of solution.

Me

MeOOBn

OMe

CHO

I Me

MeOOBn

OMeI

Nt-butylamine, PhMe4Å molecular sieves

25 °C, 3 h

OBn 2% PdCl2(PPh3)2, 1% CuI, Et3N, 55 °C, 3 h

2) 10% CuI, DMF, 100 °C, 5 h

1)

Me

MeOOBn

OMe

CHO

OBnMe

MeOOBn

OMe

CHO

I Me

MeOOBn

OMe

NOBn

+ +

Scheme 2.36. Larock Conditions for Isoquinoline Synthesis

240, 33% yield 239, 18% yield 234, 41% yield

238234

202

79

To avoid the problems seemingly associated with the use of palladium we decided

to leave the palladium out of the reaction mixture. In the absence of palladium a

stoichiometric amount of copper was used to conduct the Castro-Stevens reaction.29

Under optimized conditions it was found that stirring imine 238 with acetylene 202 in

triethylamine with 1.2 equivalents of CuI at room temperature for 24 hours gave the

cleanly coupled product in near quantitative yield (Scheme 2.37). A filtration step was no

longer necessary as the same reaction flask was heated to 85 °C, after complete coupling,

and stirred for one hour to promote cyclization to isoquinoline 240. Attempts to heat the

reaction before complete imine 238 coupling led to significant by-product formation. The

yield for the one-pot procedure was 91% yield.

Me

MeOOBn

OMeI

N

OBn1.2 equiv. CuI, Et3N, 25 °C, 24 h

then 85 °C, 1 h

Me

MeOOBn

OMe

NOBn

91% yield

Scheme 2.37. Castro-Stevens Conditions for Isoquinoline Synthesis

Castro-Stevens reactions often require heating for the coupling to occur. With the

imine substrate 238 simply stirring at room temperature generated the coupled product in

near quanitiative yield. However it was noted that room temperature coupling with

aldehyde 234 was unsuccessful and only unreacted starting material was observed after

stirring overnight. It appears the imine nitrogen coordinates with the CuI to lower the

activation energy for acetylene insertion into the aryl iodine bond.

238 240

80

2.2.7. ALKYLATION, REDUCTION, AND CYCLIZATION OF THE REAL SYSTEM

The substituted isoquinoline could now be obtained very cleanly and efficiently

using a modification of the Larock isoquinoline synthesis. Alkylation of isoquinoline 240

with phenylthiomethyllithium in the presence of (−)-sparteine gave

1,2-dihydroisoquinoline 241 in 83% yield (Scheme 2.38). Three equivalents of

phenylthiomethyllithium were used to hasten the alkylation of the much less reactive

electron rich isoquinoline 240. The enantioselectivity of this reaction was also

investigated, but unfortunately separation of the enantiomers by chiral HPLC revealed a

1:1 mixture.

Me

MeOOBn

OMe

NOBn

1) PhSCH2Li (3.0 equiv.) (−)-sparteine (3.0 equiv.) PhMe, −78 °C to 25 °C, 15 min.

2) ClCO2Me

83% yield

Me

MeOOBn

OMe

NOBn

CO2Me

SPh

Scheme 2.38. Alkylation of Substituted Isoquinoline 240

The alkylated product 241 was then subjected to ionic hydrogenation conditions,

previously used with much success for the unsubstituted analogue 205. Treatment of a

solution of 241 and Et3SiH in CH2Cl2 with TFA gave a mixture of two products (Scheme

2.38). The over-reduced 3-methyl tetrahydroisoquinoline 242 was isolated in 61% yield

and the desired product 243 was isolated in 31% yield. The 3-methyl product 242 was

unexpected as no by-products were observed in the model system. This suggests that

neighboring group participation of the methyl carbamate is not likely as it would have

241 240

81

been observed in the model system. The unwanted product was attributed to the electron

rich aromatic ring.

Me

MeOOBn

OMe

NOBn

CO2Me

SPh

Me

MeOOBn

OMe

N

Me

CO2Me

SPh

Me

MeOOBn

OMe

NOBn

CO2Me

SPh

H

H

H

H

TFA, Et3SiH, CH2Cl2−10 °C to 25 °C, 3 h

+

Scheme 2.39. Ionic Hydrogenation on the Real System

The 3-methyl tetrahydroisoquinoline 242 could be formed via the pathway

outlined in Scheme 2.40. In the presence of a strong acid small amounts of the protonated

ether 245 may exist. The lone pair of electrons on either arylmethoxy of 245 can donate

into the ring system and eliminate benzyl alcohol, forming oxonium intermediate 246.

Hydride addition to the activated intermediate would give 247. Subsequent ionic

hydrogenation of 247 leads to the over-reduced product 242. The reaction mechanism

suggests that the pathway for elimination of benzyl alcohol remains reversible until the

first hydride addition generates 247. In order to minimize the amount of eliminated

benzyl alcohol intermediate we conducted the reaction in excess benzyl alcohol, hoping

to employ Le Chatelier’s principle. By conducting the reaction with benzyl alcohol as a

co-solvent the yield of the desired product 243 was increased to 71% (Scheme 2.41) and

by-product 242 was isolated in only 22% yield.

241

242, 61% yield

243, 31% yield

82

241

Me

MeOOBn

OMe

NOBn

CO2Me

SPh

TFA, Et3SiH

−10 °C to 25 °C, 3 h

H

H

Me

MeOOBn

OMe

NOBn

CO2Me

SPh

H

Me

MeOOBn

OMe

NOBn

CO2Me

SPh

Me

MeOOBn

OMe

NCO2Me

SPh

Me

MeOOBn

OMe

N

Me

CO2Me

SPhH

H

Scheme 2.40. Ionic Hydrogenation on the Real System

Me

MeOOBn

OMe

NOBn

CO2Me

SPh

TFA, Et3SiH, BnOH

−10 °C to 25 °C, 16 h

Scheme 2.41. Improved Reduction Conditions

243 +

247

246

245

242

243 + 242

71% yield 22% yield

244

241

83

With an improved synthesis of the desired 1,3-cis-substituted

tetrahydroisoquinoline 243 enough material was available to investigate the formation of

the requisite azide. Benzyl ether 243 required deprotection, however in the real system an

additional benzyl ether is present on the aryl ring. Fortunately the previous conditions

established for benzyl ether deprotection, BCl3, provided the required selectivity. Boron

trichloride can selectively deprotect alkylbenzyl ethers over arylbenzyl ethers because the

mechanism first involves coordination of the boron with oxygen to form an activated

intermediate 249 (Scheme 2.42), and the alkylbenzyl ether oxygen has the most

nucleophilic lone pairs. Treatment of benzyl ether 243 with BCl3 at −40 °C, with careful

monitoring of the reaction, gave the desired alcohol 248 in 88% yield.

Me

MeOOBn

OMe

NOBn

CO2Me

SPh

Me

MeOOBn

OMe

NOH

CO2Me

SPh

H

H

H

HBCl3, CH2Cl2

−40°C, 2 h

Me

MeOOBn

OMe

NO

CO2MeSPh

H

H

Ph

BCl2Cl

BCl2Cl

88% yield

Scheme 2.42. Alkylbenzyl Ether Deprotection

243 248

249

84

Alcohol 248 was then converted to azide 250 in 85% yield (Scheme 2.43) using

the conditions from the model system with diisopropyl azodicarboxylate (DIAD)

replacing DEAD. DIAD could be stored longer than DEAD and gave identical results.

Azide 250 was subjected to the Pummerer cyclization conditions previously used. The

desired cyclized product 251 was isolated in 51% yield. For this substrate the

iminosulfide could not be isolated nor observed by TLC analysis. Lactam 251 was

characterized and the spectroscopic data was consistent with the model lactam system.

Me

MeOOBn

OMe

NOH

CO2Me

SPh

H

H

1) NCS, PhCl, 2 h, 25 °C

2) SnCl4, 0 °C, 5 min.

51% yield

NNH

CO2Me

O

(PhO)2PON3, DIADPPh3, THF

0 °C to 25 °C, 1.5 h

85% yield

Me

MeOOBn

OMe

NN3

CO2Me

SPh

H

H

MeOMe

MeOOMe

Scheme 2.43. Cyclization of the Real System

248

251

250

85

2.3. CONCLUSION

The model conditions proved successful in their application to the real system

leading to the synthesis of the bicyclo[3.3.1] system present in saframycin B. Further

work was needed to install the second 1,3-cis-substituted tetrahydroisoquinoline which

would likely go through intermediate 252 (Scheme 2.44). However, after the change in

our strategy we found our approach resembled the diketopiperazine derived intermediates

used by Kubo and Fukuyama for their work in the saframycins.

NNH

CO2Me

O

MeOMe

MeOOBn

NNR

CO2MeMeOMe

MeOOBn

Ar

ArHN

O

N

BnOOMe

Me

OMeCbz

H

H

ArRN

N

BnOOMe

Me

OMeCO2Me

H

H

ArHN

N

MeOOMe

Me

OMeMe

H

H

Scheme 2.44. Other Intermediates in the Synthesis of Saframycin B

252

Fukuyama’s Intermediate

251

Kubo’s Intermediate

86

At this stage, the approach to saframycin B was terminated. We were successful

in developing a new stereoselective approach for the formation of the 1,3-cis-substituted

tetrahydroisoquinoline core of the tetrahydroisoquinoline alkaloids by alkylating

unactivated isoquinolines with phenylthiomethyllithium followed by ionic hydrogenation.

The lack of enantioselectivity was disappointing but we hoped to reinvestigate this issue.

In addition a new lactam synthesis was developed from the reaction of an azide with a

Pummerer intermediate to form the bicyclo[3.3.1] system found in saframycin B. The

project now turned toward the synthesis of an alternative tetrahydroisoquinoline alkaloid.

87

2.4. REFERENCES

1) Dyke, S. F.; Kinsman, R. G. Heterocyclic Compounds; Wiley-Interscience: New York, 1981; Vol. 38, Part 1-3. 2) Kobayashi, S.; Ishitani, H. Chem. Rev. 1999, 99, 1069. 3) Blasko, G.; Kerekes, P.; Makleit, S. Alkaloids 1987, 31, 1. 4) Shibasaki, M.; Takamura, M.; Funabashi, K.; Kania, M. J. Am. Chem. Soc. 2000, 122, 6327. 5) Comins, D. L.; Goehring, R. R.; Joseph, S. P.; O’Connor, S. J. Org. Chem. 1990, 55, 2574. 6) Comins, D. L.; Badawi, M. M. Heterocycles 1991, 32, 1869. 7) Frank Amiot’s Dissertation, 2002, Universite de Geneve, Switzerland. 8) Marazano, C.; Barbier, D.; Riche, C.; Das, B. C.; Potier, P. J. Org. Chem. 1998, 63, 1767. 9) Alexakis, A.; Amiot, F. Tetrahedron: Asymmetry 2002, 13, 2117. 10) Shirley, D. A.; Reeves, B. J. J. Organometallic Chemistry 1969, 16, 1. 11) Corey, E. J.; Seebach, D. J. Org. Chem. 1966, 31, 4097. 12) Peterson, D. J. Org. Chem. 1967, 32, 1717 13) Pyne, S. G.; Dikic, B. J. Org. Chem. 1990, 55, 1932. 14) Solladié, G. Synthesis 1981, 185. 15) Birch, A. J.; Walker, K. A. M. Tetrahedron Lett. 1967, 8, 1935. 16) Molinski, T. F.; Masuno, M. N. Tetrahedron Lett. 2001, 42, 8263. 17) Langy, K. C. Org. Prep. Proc. Int. 1994, 26, 429. 18) Larock, R. C.; Roesch, K. R. J. Org. Chem. 2002, 67, 86. 19) Bacon, R. G. R.; Lindsay, W. S. J. Chem. Soc., 1958, 1375.

88

20) Larock, R. C.; Doty, M. J.; Cacchi, S. J. J. Org. Chem. 1993, 58, 4579. 21) Ireland, R. E.; Anderson, R. C.; Badoud, R.; Fitzsimmons, B. J.; McGarvey, G. J.; Thaisrivongs, S.; Wilcox, C. S. J. Am. Chem. Soc. 1983, 105, 1988. 22) Cooley, J.; Evain, E. J. Synthesis 1989, 1. 23) Turos, E.; Ren, X. F.; Lake, C. H.; Churchill, M. R. J. Org. Chem. 1995, 60, 6468. 24) Bose, A. K.; Lal, B.; Pramanik, B. N.; Manhas, M. S. Tetrahedron Lett. 1977, 18, 1977. 25) Kubo, A.; Saito, N.; Yamato, H.; Masubuchi, K.; Nakamura, M. J. Org. Chem. 1988, 53, 4295. 26) Godfrey, I. M.; Sargent, M. V.; Elix, J. A. J. Chem. Soc., Perkin Trans. 1 1974, 1353. 27) Kubo, A.; Kitahara, T.; Nakahara, S.; Numata, R. Chem. Pharm. Bull. 1985, 33, 2122. 28) Suzuki, K.; Saito, T.; Morimoto, M.; Akiyama, C.; Matsumoto, T. J. Am. Chem. Soc. 1995, 117, 1075. 29) Castro, C. E.; Havlin, R.; Honwad, V. K.; Malte, A.; Moje, S. J. Am. Chem. Soc. 1969, 91, 6464.

89

Chapter 3: Toward the Synthesis of Lemonomycin

3.0. INTRODUCTION

The new target tetrahydroisoquinoline alkaloid, lemonomycin (10, Scheme 3.01),

was chosen because at the beginning of the project the total synthesis of this unique

structure had not been reported. It was not until halfway through the project that Stoltz

reported his enantioselective total synthesis.1

In our retrosynthetic analysis of lemonomycin, the western fragment was to be

synthesized employing the chemistry previously developed. We anticipated the necessity

for an alternative nucleophile than phenylthiomethyllithium for the alkylation of

isoquinoline 257 (Scheme 3.01). The presence of the resulting sulfide side-chain would

require extra steps for conversion to the requisite ether. Once the 1,2-dihydroisoquinoline

256 was formed, finding appropriate conditions for reduction that would provide an

efficient yield of the 1,3-cis-substituted tetrahydroisoquinoline 255 would be important.

The current conditions for ionic hydrogenation of the electron rich system led to

elimination of benzyl alcohol. We anticipate that the elimination by-product would be

observed no matter which protecting group was used. Further elaboration to

lemonomycin (10) would proceed through intermediate 254 followed by formation of the

bicyclo[3.2.1]system 253.

90

NNH

H

HOH

MeO

MeO

O

H

H

O

O

Me

OHMe

NMe2

HOOH

NN

H

HO

MeOMe

OBn

H

H

OBn

CHO

R2

NNR2

OMeO

MeOMe

OBn

H

H

OBn

OR3

NHMeO

MeOMe

OBn

H

H

X

NMeO

MeOMe

OBn

H

X

OHN

MeO

MeOMe

OBn

OR1 OH

1

11

13

314

4

17

15

16

18

6 10

8

1

3

1

3

1

3

1

3

1

3

MeO

CO2Me

Scheme 3.01. Retrosynthesis of Lemonomycin (10)

10

255

253 254

256257

91

3.1. RESULTS AND DISCUSSION

3.1.1. ALKYLATION WITH BENZYLOXYMETHYLLITHIUM

The alkylation of an isoquinoline with a nucleophile other than

phenylthiomethyllithium would still require the nucleophile to have the general structure

Li-CH2-X. The preference for our approach to lemonomycin would be where X is an

oxygen atom. A literature search revealed that benzyloxymethyllithium2 (BOM-Li, 260,

Scheme 3.02) was a well investigated nucleophile and a suitable choice. The

organolithium nucleophile is formed from trans-metallation of the stannane 259 with

n-BuLi. Stannane 259 is formed from stannylation of benzyl chloromethyl ether3 (258)

with Bu3SnLi.

O

ClBu3Sn-Li, THF

O

SnBu3n-BuLi

O

Li

−78 °C to 25 °C, 30 min. THF, −78 °C

70% yield

Scheme 3.02. Formation of Benzyloxymethyllithium

The alkylation of isoquinoline with benzyloxymethyllithium was used as a model

system to determine the reactivity of the nucleophile. A solution of stannane 259 and 1.7

equivalents of (−)-sparteine in toluene at −78 °C was treated with one equivalent of

n-BuLi (Scheme 3.03). After 10 minutes a solution of isoquinoline in THF was added and

the reaction warmed to room temperature. The starting material was consumed and the

reaction was quenched with methyl chloroformate to give the 1,2-dihydroisoquinoline

261 in 66% yield. The enantiomers were separated by chiral HPLC (Diacel, Chiracel OD

column) in a ratio of 2:1, revealing an enantioselectivity of 33% for the conversion.

258 259 260

92

229

O

i) n-BuLi, (−)-sparteine PhMe, −78 °C, 10 min.

SnBu3ii)

N

−78 °C to 25 °C, 15 min.

, THF

iii) ClCO2Me

N

OBn

CO2Me

Scheme 3.03. Alkylation of Isoquinoline with BOM-Li

These promising results demonstrated the utility of the nucleophile and that under

unoptimized conditions some enantioselectivity could be achieved. The observed

enantioselectivity was unexpected because the analogous alkylation with

phenylthiomethyllithium showed no enantioselectivity under a variety of conditions. If

the steric differences of the nucleophiles are considered negligible, then the main

difference is the heteroatom; sulfur and oxygen have different electronic properties that

could effect the coordination of (−)-sparteine to the respective lithiated species. The

better coordinating sulfur lone pairs could be stabilizing the anion without assistance

from the nitrogen lone pairs of (−)-sparteine, while in the case of oxygen the nitrogen

atoms of (−)-sparteine provide stabilization and form a chiral complex that shows facial

selectivity in the alkylation of isoquinoline. The reaction was not optimized for the model

system because application of this alkylation to the real system was of more importance.

In our approach to lemonomycin we wanted to use an alternative substituted

isoquinoline to 240, used in our work toward saframycin B. Using the same isoquinoline

for the alkylation with BOM-Li the product would possess two alkylbenzyl ethers which

could prove difficult to differentiate at a later stage. Instead a triisopropylsilyl protected

66% yield, 33% ee

261

259

93

alcohol at the C3 position was preferred, which would provide differentiation as well as

stability to the alkylation conditions.

The synthesis of isoquinoline 262 (Scheme 3.04) was first attempted using a

modified one-pot procedure of the Larock conditions that worked well before. The

Castro-Stevens reaction between imine 238 and acetylene 200 appeared to be consistent

with prior substrates, but upon heating the reaction to promote cyclization degradation

was observed and the desired isoquinoline 262 was not isolated. The only difference from

the prior isoquinoline synthesis was the presence of the triisopropylpropargyl ether.

During the Castro-Stevens coupling one equivalent of triethylamine hydroiodide was

formed, which at room temperature did not appear to deprotect the silyl ether. However,

heating to 80 °C could lead to acid catalyzed deprotection of the silyl ether. Larock

reported that propargyl alcohol does not undergo isoquinoline formation.4

Me

MeOOBn

OMeI

N

OTIPSCuI (1.2 equiv.), Et3N, 25 °C, 24 h

then 80 °C, 1 h

Me

MeOOBn

OMe

NOTIPS

Scheme 3.04. Attempted Synthesis of Isoquinoline 262

To solve this problem a basic workup was conducted after the Castro-Stevens

room temperature coupling was complete. The coupled intermediate was then dissolved

in DMF with catalytic CuI and heated, now in the absence of triethylamine hydroiodide,

to afford the desired isoquinoline 262 in 76% yield (Scheme 3.05).

238

200

262

94

Me

MeOOBn

OMeI

N

OTIPSCuI (1.2 equiv.), Et3N, 25 °C, 24 h

2) CuI (0.2 equiv.), DMF 100 °C, 1 h

Me

MeOOBn

OMe

NOTIPS

1)

76% yield

Scheme 3.05. Synthesis of Isoquinoline 262

The alkylation of isoquinoline 262 with BOM-Li in the presence of (−)-sparteine

was performed under the conditions used for the previous model isoquinoline (Scheme

3.06). The starting material was consumed but a mixture of unidentified products was

isolated.

Me

MeOOBn

OMe

NOTIPS

CO2Me

OBn

O

i) n-BuLi, (−)-sparteine PhMe, −78 °C, 10 min.

SnBu3ii)

−78 °C to 25 °C, 15 min.262, THF

iii) ClCO2Me

Scheme 3.06. Attempted Synthesis of 263

The reaction conditions were simplified by removing (−)-sparteine and using only

THF as solvent. A variety of conditions were employed varying the equivalents of

stannane 259, the equivalents of n-BuLi, and reaction temperature (Scheme 3.07). The

results are summarized in Table 3.01.

262 238

259

263

200

95

Me

MeOOBn

OMe

NOTIPS

CO2Me

OBni) n-BuLi (equiv.), THF, −78 °C, 10 min.

ii)THF, temp.262 (1.0 equiv.),

iii) ClCO2Me (5.0 equiv.)

Me

MeOOBn

OMe

NOTIPS

CO2Me

Me

MeOOBn

OMe

NOTIPS

CO2Me

OBn

CO2Me

Scheme 3.07. Alkylation of 262

yield (%) entry 259

(equiv.) n-BuLi (equiv.)

temp. (°C) 262 263 264 265

1 1.2 1.2 −78 to 25 68 0 8 12

2 3 1 −78 to 25 43 0 32 0

3 3 3 −78 to 25 31 0 38 8

4 3 1 −78 28 57 0 0

5 3 3 −78 0 45 0 33

6 4 3 −78 0 79 0 trace

Table 3.01. Alkylation of 262

The subtle reaction changes demonstrate the capricious reactivity of BOM-Li and

isoquinoline 262. The conditions for entries 1 to 3 allowed warming to room temperature

using either one equivalent of BOM-Li or an excess, but a large percentage of recovered

starting material was isolated. It is likely that BOM-Li is not stable at room temperature

and the isoquinoline 262 is not consumed before degradation. As observed in the

264

263

265

259

96

alkylations with phenylthiomethyllithium, the electron rich isoquinoline 262 is less

reactive than the unsubstituted isoquinoline. The isoquinoline 262 that was alkylated gave

mainly diacylated product 264 upon methyl chloroformate quench. Keeping the reaction

at −78 °C and generating one equivalent of BOM-Li (entry 4) gave better conversion and

57% yield of the desired mono-acylated product 263. The diacylated product 264 was not

observed, however, nearly 30% of the starting isoquinoline was isolated. In an attempt to

drive the reaction to completion, three equivalents of BOM-Li were used (entry 5) which

did succeed in consuming all of the starting isoquinoline. Upon quenching with methyl

chloroformate the n-butyl adduct 265 was isolated in 33% yield along with the desired

product 263 in 45% yield. The origin of 265 was initially thought to be due to excess

n-BuLi in the reaction arising from improper titration of the commercial n-BuLi solution

in hexanes. After multiple reactions using a one to one ratio of carefully titrated n-BuLi

and stannane 259 the n-butyl adduct 265 was always isolated in significant yield, which

suggested that an aggregate could be present. It was then observed that a slight excess of

stannane 259 over n-BuLi (entry 6) minimized the amount of 265 and gave the desired

1,2-dihydroisoquinoline 263 in 79% yield.

97

3.1.2. FORMATION 1,3-CIS-SUBSTITUTED TETRAHYDROISOQUINOLINE

When 263 was treated under ionic hydrogenation conditions none of the desired

1,3-cis-substituted tetrahydroisoquinoline 266 could be isolated or observed (Scheme

3.08). The ionic hydrogenation of 1,2-dihydroisoquinoline 263 was anticipated to be

problematic, as consistent with the observations made in our approach to saframycin B.

Two possible by-products were the acyl-migration adduct 269 and the over-reduced

3-methyl adduct 272. The former coming from loss of the silyl ether and acid catalyzed

migration of the methyl carbamate to form the methyl carbonate 269. The latter could

arise from elimination of H2O or HOTIPS and reduction of the oxonium intermediate 271

leading to C3-methyl tetrahydroisoquinoline 272.

Me

MeOOBn

OMe

NOTIPS

CO2Me

OBn

TFA, Et3SiH, CH2Cl2−10 °C to 25 °C, 1 h

H

Me

MeOOBn

OMe

NO

CO2MeOBn

H Me

MeOOBn

OMe

NCO2Me

OBn

Me

MeOOBn

OMe

N

Me

CO2Me

OBn

Me

MeOOBn

OMe

NOH

CO2Me

OBn

H

H

Me

MeOOBn

OMe

NOH

OBn

Me

MeOOBn

OMe

N

OBn

O

OHOMe

OMe

O

Me

MeOOBn

OMe

N

OBn

OCO2Me

-TIPS

H

H

H

Scheme 3.08. Failed Ionic Hydrogenation

266263

269

272 271

268267

270

98

The acyl-migration product could readily be avoided by replacing the acid

sensitive protecting group on the alcohol. The elimination product could not be avoided

without changing the electronics of the electron rich isoquinoline ring system. The first

solution to the problem was to change the reduction conditions and not the substrate.

Attempts to reduce the 3,4-olefin under catalytic hydrogenation conditions were

unsuccessful (Scheme 3.09). The cyclic tri-substituted olefin is quite hindered and

literature suggested hydrogen pressures over 1000 psi would have been necessary.5 In

addition, deprotection of the benzyl ethers was observed.

Me

MeOOBn

OMe

NOTIPS

CO2Me

OBn

Me

MeOOH

OMe

NOTIPS

CO2Me

OH

PtO2, EtOH

H2 (1 atm)

87% yield

Scheme 3.09. Attempted Hydrogenation

273263

99

An effort was made to utilize the proximity of the C3 hydroxymethyl substituent,

which, when the silyl ether was removed by treatment with TFA gave allylic alcohol 274

in 88% yield (Scheme 3.10). Catalytic hydrogenation conditions were employed with the

hopes that neighboring group assistance would promote reduction. Reduction was not

observed only benzyl ether deprotection resulting in 275.

Me

MeOOBn

OMe

NOTIPS

CO2Me

OBn

TFAMe

MeOOBn

OMe

NOH

CO2Me

OBn88% yield

CH2Cl2, 0 °C

5% Pt/C, EtOH

H2 (1 atm), 16 h

87% yield

Me

MeOOH

OMe

NOH

CO2Me

OBn

Scheme 3.10. Allylic Alcohol Formation and Attempted Reduction

275

274263

100

Reduction of the allylic alcohol was then attempted with NaBH4, which has

shown to be successful in berberine alkaloids for the reduction of eneamines.6 When

alcohol 274 was treated with NaBH4 in MeOH a new compound was formed (Scheme

3.11). However it proved not to be the reduced compound 266 but rather the cyclized

product 276. NaBH4 was reacting as a base generating the alkoxide 277, which forms the

cyclized intermediate 278 then the oxazolidinone derivative 276.

Me

MeOOBn

OMe

NOH

CO2Me

OBn

NaBH4, MeOH

25 °C, 15 h

Me

MeOOBn

OMe

N

OBn

O

O86% yield

Me

MeOOBn

OMe

NO

OBn

OMe

O

Me

MeOOBn

OMe

N

OBn

O

OOMe

Scheme 3.11. Unexpected Formation of Oxazolidinone 276

274

277 278

276

101

276

The new compound 276 was subjected to ionic hydrogenation conditions and

clean reduction to the tetrahydroisoquinoline 279 as one diastereomer was observed

(Scheme 3.12). It was expected that reduction of the oxazolidinone derivative 276 was

not likely to give the acyl-migration product, but by-products from elimination were

anticipated. One could imagine protonation of the carbonyl moiety resulting in

intermediate 280, which could lead to the ring-opened intermediate 281 and

decarboxylated intermediate 282. However, none of the anticipated by-products from the

elimination pathway were observed.

TFA, Et3SiH

−10 °C to 25 °C, 3 h

Me

MeOOBn

OMe

N

OBn

O

O

Me

MeOOBn

OMe

N

OBn

O

O95% yield

H

H

Me

MeOOBn

OMe

N

OBn

O

O H

Me

MeOOBn

OMe

N

OBn

O

OH

H

Me

MeOOBn

OMe

N

OBn

Me

-CO2

H

Scheme 3.12. Successful Ionic Hydrogenation

279

280

281

282

102

Conversion to the oxazolidinone 279 was optimized by treating silyl ether 263

with TBAF to give 276 in 91% yield (Scheme 3.13). Reduction with Et3SiH and TFA in

CH2Cl2 gave the tetrahydroisoquinoline 279, which was hydrazinolyzed to yield the

amino alcohol 283 in 86% yield over the two steps. The amino alcohol 283 was

recrystallized and an X-ray structure (appendix D) proved that the relative

stereochemistry was the anticipated cis-isomer.

Me

MeOOBn

OMe

NOTIPS

CO2Me

OBn

Me

MeOOBn

OMe

NHOH

OBn

H

H

Me

MeOOBn

OMe

N

OBn

O

O−10 °C to 25 °C

TBAF, THF

91% yield

1) TFA, Et3SiH−10 °C to 25 °C, 2 h

2) H2NNH2·H2O, KOHethylene glycol150 °C, 3 h

86% yield

Scheme 3.13. Optimized Formation of Amino Alcohol 283

283, X-ray

276 263

103

3.1.3. AMIDE COUPLING

In our approach to lemonomycin we intended to construct the bicyclo[3.2.1]

system 284 from a hemi-aminal 285, which would come from an amide coupling of the

amino alcohol 283 (Scheme 3.14).

Me

MeOOBn

OMe

NHOH

OBn

H

H

NN

H

HO

MeOMe

OBn

H

H

OBn

R2

R1

NNR1

OMeO

MeOMe

OBn

H

H

OBn

OH

MeO

Scheme 3.14. Retrosynthesis of Bicyclo[3.2.1] System

A literature search showed precedence for performing amino acid couplings with

hindered amines using PyBOP® as an activating reagent.7 A solution of amino alcohol

283 and one equivalent of BocGlyOH in CH2Cl2 was treated with PyBOP® and Hünig’s

base (Scheme 3.15). The only product isolated was ester 286, none of the amide 287 was

observed.

Me

MeOOBn

OMe

NHOH

OBn

H

HMe

MeOOBn

OMe

NHO

OBn

H

HNHBoc

O1.0 equiv. BocGlyOH

1.0 equiv. PyBOP®

Hünig's base, CH2Cl20 °C to 25 °C, 4 h

82% yield

Scheme 3.15. Ester Formation

285284 283

286283

104

287

The amide 287 was predicted to be more thermodynamically stable than the ester,

so conditions were investigated to promote thermal acyl-migration (Scheme 3.16).

Heating ester 286 in dichlorobenzene at 150 °C gave only unreacted starting material.

Heating in the presence of thiopyridone at 100 °C was also unsuccessful in migrating the

glycine to the amine.

Me

MeOOBn

OMe

NHO

OBn

H

HNHBoc

O dichlorobenzene

150 °C Me

MeOOBn

OMe

NOH

OBn

H

H

ONHBoc

N SHPhMe, 100 °C

Scheme 3.16. Attempted Acyl-Migration

It was observed that the use of an excess of BocGlyOH and PyBOP® during the

coupling with amino alcohol 283 gave a dicoupled product 288 in 85% yield (Scheme

3.17). The ester was hydrolyzed by treatment with NaOH to afford amide 287 in 60%

yield. While this two step procedure succeeded in providing the desired amide product it

was not particularly efficient, so an alternative method was developed.

Me

MeOOBn

OMe

NO

OBn

H

HNHBoc

O3.0 equiv. Boc-GlyOH

3.0 equiv. PyBOP®

Hünig's base, CH2Cl20 °C to 25 °C, 15 h

85% yield

ONHBoc

NaOH

DMSO

60% yield

Scheme 3.17. Unoptimized Formation of Amide 287

283 287

286

288

105

283 287

289

The previous results demonstrated that the first coupling occurred at the alcohol to

give the ester, but the amine was not too hindered to undergo a second coupling to

provide the amide 288. If the alcohol was protected and treated under coupling conditions

then amide 287 could be isolated directly. A multiple step sequence involving selective

protection of the alcohol would prove tedious. Thus, the best approach would be an in

situ protection of the alcohol during amide coupling and subsequent removal in the

workup.

The answer came by the way of silyl-activated amide coupling conditions, taking

advantage of the known silyl activation of amines.8 Treatment of amino alcohol 283 with

two equivalents of chlorotrimethylsilane gave the disilylated intermediate 290 (Scheme

3.18). The silyl ether prevents ester formation, while silylation of the amine leads to

activation of the nitrogen.

Me

MeOOBn

OMe

NHOH

OBn

H

H 1) TMSCl, Et3N, THF 0 °C to 25 °C, 3 h

2)

THF, −65 °C to 25 °C, 13 h

O

O ONHBoc

Me

MeOOBn

OMe

NOH

OBn

H

H

ONHBoc

Me

MeOOBn

OMe

NTMSOTMS

OBn

H

H

TMSCl

Me

MeOOBn

OMe

NOTMS

OBn

H

H

ONHBoc

82% yield H

Scheme 3.18. Silyl-Activated Amide Coupling Conditions

290 291

289

106

292 21% yield

The reaction conditions were carefully optimized to improve the yield and prevent

by-product formation. The disilylated intermediate 290 was added with filtration to a

solution of the mixed anhydride 289 at −65 °C and then slowly warmed to room

temperature with stirring overnight. Addition of the disilylated 290 to mixed anhydride

289 at −20 °C or warmer gave the tert-butyl amide product 292 from addition at the

wrong carbonyl (Scheme 3.19).

THF, −20°C to 25 °C, 14 h

Me

MeOOBn

OMe

N

OBn

H

H

O

OH

Scheme 3.19. tert-Butyl Amide By-Product Formation

The primary alcohol of amide 287 was then oxidized under Swern conditions9 to

afford hemi-aminal 293 as a mixture of diastereomers (3:2) in 94% yield (Scheme 3.20).

Care was taken during the oxidation to avoid dehydration as this is known to occur with

prolonged stirring at room temperature or with the use of Dess-Martin periodinane.10

Me

MeOOBn

OMe

NOH

OBn

H

H

ONHBoc

(COCl)2, DMSOEt3N, CH2Cl2

−78 °C to 25 °C, 8 min.

87% yield

Me

MeOOBn

OMe

N

OBn

H

HNBoc

OH

O

11

Scheme 3.20. Hemi-Aminal Formation

287 293, 3:2 mixture

at C11

290 287 57% yield

289 +

107

3.1.4. STEREOSELECTIVE INCORPORATION OF C14-C15

The strategy for building the bicyclo[3.2.1] system 294 (Scheme 3.21) in the

eastern fragment of lemonomycin was not planned from the beginning. The biggest

challenge lies with stereoselectivity for the incorporation of the three carbon unit. The

1,3-cis-substituted tetrahydroisoquinoline 295 possesses most of the steric bulk on the top

face, suggesting potential problems with stereoselectivity during the carbon-carbon bond

forming reactions at either C11 or C13.

Me

MeOOBn

OMe

N

OBn

H

HNBoc

OH

O

NN

H

HO

MeOMe

OBn

H

H

OBn

R1

Boc

1

3

MeO1

3 11 11

1313

15

14

Scheme 3.21. Alkylation Across C11 and C13

A cycloaddition process was avoided as this strategy has been shown to work best

on substrates which cannot undergo β-elimination at the C3 center.11 Instead, alkylation

at C11 was investigated to determine the reactivity and stereoselectivity at that center.

The first approach involved N-acyliminium alkylation with allylsilane (Scheme 3.22),

however, the alkylated product 296 was not observed and only starting material was

recovered. The starting hemi-aminal is identical to the hydrolyzed iminium intermediate

so it was not possible to determine if the N-acyliminium ion had formed.

294 295

108

Me

MeOOBn

OMe

N

OBn

H

HNBoc

OH

O

Si(Me)3Tf2O, DABCOEt2O, −78 °C

Me

MeOOBn

OMe

N

OBn

H

HNBoc

O

Scheme 3.22. Attempted Allylation

The hemi-aminal 293 was converted to the methyl aminal 297 in 95% yield on

treatment with CSA and trimethylorthoformate in methanol (Scheme 3.23). The methyl

aminal was formed in a ratio of 6:1 but the relative stereochemistry at C11 was not

determined, nor could the diastereomers be separated by flash chromatography. The

product was then subjected to allylation under a variety of conditions.

Me

MeOOBn

OMe

N

OBn

H

HNBoc

OH

O

HC(OMe)3, CSA

MeOH, 0 °C, 30 min.

95% yield

Me

MeOOBn

OMe

N

OBn

H

HNBoc

OMe

O

11

RLA,

R = Si(Me)3 Sn(Me)3LA = BF3·OEt2 TiCl4 SnCl4

TMSOTf

OTBS

Me

MeOOBn

OMe

N

OBn

H

HNBoc

O

CHO

Scheme 3.23. Attempted Allylations using Methyl Aminal 297

293 296

293

297296

298

297, 6:1 mixture at C11

299

109

Alkylation was not observed under any conditions. The main product isolated was

always hemi-aminal 293, which suggests that the N-acyliminium intermediate was

forming but not reacting. A more reactive nucleophile than allylsilane and allylstannane,

the silyl enol ether of acetaldehyde 298,12 was employed but again without success. An

observation made under heating was the apparent loss of the benzyl ethers, which may

have been from reaction with the lewis acid catalyst. To avoid this we investigated an

alternative method for the incorporation of the three carbon unit.

The hemi-aminal 293 was converted to thioaminal 300 by treatment with

thiophenol and para-toluenesulfonic acid in 86% yield from alcohol 287, under

optimized conditions (Scheme 3.24). Only one diastereomer was observed, which upon

recrystallization and X-ray analysis (appendix E) proved to be the cis-isomer. This

stereochemical result was surprising because the X-ray suggests that the axial orientation

of the C1 substituent ought to provide enough steric bulk to destabilize the axial

orientation of the C11 thiophenol (Figure 3.01). The likely cause of this preferred

orientation is the planar N12-nitrogen. Strong carbamate resonance was observed in 1H

NMR, suggested the nitrogen-carbon bond to the N-Boc carbonyl has imine character. As

a result the N-Boc is coplanar with the N12-nitrogen making the neighboring equatorial

positions more sterically hindered than the axial positions.

1) (COCl)2, DMSO, Et3N, CH2Cl2 −78 °C to 25 °C, 8 min.

2) PhSH, pTSA 0 °C, 1.5 h

86% yield

Me

MeOOBn

OMe

N

OBn

H

HNBoc

O

SPhMe

MeOOBn

OMe

NOH

OBn

H

H

ONHBoc

Scheme 3.24. Optimized Thioaminal Formation

287 300, X-ray

110

Figure 3.01. ORTEP Representation of Thioaminal 300 X-ray Structure

With application of the thioaminal we hoped to use a thiophilic lewis acid that

could selectively generate the N-acyliminium ion without reacting with the benzyl ethers

or N-Boc protecting group. Silver tetrafluoroborate was chosen as the thiophile.

Unfortunately treatment of thioaminal 300 with silver tetrafluoroborate in the presence of

tributylallylstannane gave only hemi-aminal 293 after an aqueous quench, 296 was not

observed (Scheme 3.25). Benzyl ether or N-Boc deprotection were not observed and the

C11 position proved unreactive toward alkylation by N-acyliminium chemistry.

Me

MeOOBn

OMe

N

OBn

H

HNBoc

O

SPh

Sn(Bu)3

Et2O, 0 °C

Me

MeOOBn

OMe

N

OBn

H

HNBoc

O

AgBF4

11 11

Scheme 3.25. Attempted Allylation of Thioaminal 300

300 296

111

300 301, X-ray

Our next approach focused on alkylation at the C13 position by enolization of the

amide and alkylation with an electrophile. Hemi-aminal 293 and methyl aminal 297 were

not used for this alkylation as they were never isolated as a single diastereomer, however

the thioaminal 300 was isolated as the cis-isomer exclusively. Examination of the X-ray

structure showed the expected stereochemistry for alkylation at C13 would be from the

bottom face to give the undesired isomer. Thioaminal 300 was deprotonated with lithium

hexamethyldisilazide at −78 °C then treated with allyl bromide to furnish the alkylated

product 301 in 70% yield (Scheme 3.26). The alkylation was complete after 20 minutes

and only one diastereomer was isolated. NMR analysis did not provide conclusive

evidence to determine the relative stereochemistry at the C13 position. However, X-ray

analysis of the recrystallized product (appendix F) showed the stereochemistry at the C13

position to be that of the expected trans-isomer (Figure 3.02).

Me

MeOOBn

OMe

N

OBn

H

HNBoc

O

SPhi) LiHMDS, THF

−78 °C, 15 min.

ii) Br

Me

MeOOBn

OMe

N

OBn

H

HNBoc

O

SPh

70% yield

13 13

Scheme 3.26. Allylation of Thioaminal 300

112

Figure 3.02. ORTEP Representation of Allylated Product 301 X-ray Structure

Since the alkylation was completely diastereoselective it was anticipated that

deprotonation of the C13 hydrogen and reprotonation would occur from the bottom face.

The alkylated product 301 was treated with potassium tert-butoxide, but no reaction

occurred at room temperature. However, heating at 55 °C for 16 hours produced a 1:1

mixture of diastereomers (Scheme 3.27).

Me

MeOOBn

OMe

N

OBn

H

HNBoc

O

SPh

t-BuOK, t-BuOH

THF, 55 °C, 16 h

Me

MeOOBn

OMe

N

OBn

H

HNBoc

O

SPh

13

Scheme 3.27. Epimerization of C13 Center

301 301/302, 1:1 mixture at C13

113

As an alternative to the thermodynamic conditions applied above, a

diastereoselective reprotonation was performed with complete deprotonation at C13 and

reprotonation in a second step. This reaction initially proved difficult because the C13

hydrogen of 301 was not as easily deprotonated as 300. Examination of the X-ray

structure (Figure 3.02) revealed the C13 hydrogen was equatorial and coplanar with

respect to the C17 carbonyl. Such alignment makes enolization more difficult because of

poor orbital overlap with the carbonyl. Treatment of 301 with LiHMDS, KHMDS, or

LDA at −78 °C showed no signs of deprotonation. Attempted deprotonation at elevated

temperatures led to degradation. A stronger base was used in conjunction with n-BuLi but

multiple by-products were observed.

Success was found using a procedure developed by S. G. Davies13 involving

treatment with t-BuLi at −78 °C followed by immediate quench with a bulky proton

source, butylated hydroxytoluene (BHT, 303, Scheme 3.28). Inversion of the C13

stereocenter afforded 302 in 79% yield, but it was observed that at least two equivalents

of t-BuLi were necessary to obtain complete inversion. Efforts to elaborate 302 to the

bridged system were unsuccessful and attempts to proceed with the synthesis from

allylated product 301 were abandoned.

Me

MeOOBn

OMe

N

OBn

H

HNBoc

O

SPh−78 °C, 30 sec.

i) 2.4 equiv. t-BuLi, THF

ii) OH

Me

MeOOBn

OMe

N

OBn

H

HNBoc

O

SPh

79% yield

Scheme 3.28. Inversion of C13 Stereocenter

301 302 303

114

An alternative electrophile to allylbromide was chosen that would require less

manipulation to form the bicyclo[3.2.1] system. Thioaminal 300 was alkylated with

bromide 304 (Scheme 3.29), but initial results were poor. The optimized conditions

included deoxygenation of the starting solution, the use of KHMDS instead of LiHMDS,

and the use of iodide 306 instead of the analogous bromide 304 (Scheme 3.30). Under

these conditions the alkylated product 305 was isolated in 78% yield, along with 10%

recovered starting material. The remaining starting material could not be minimized by

the use of excess base or iodide 306. It is likely that O-alkylation is concomitant with

C-alkylation and the O-alkylated intermediate would be readily hydrolyzed upon aqueous

workup to give starting material.

Me

MeOOBn

OMe

N

OBn

H

HNBoc

O

SPhi) LiHMDS, THF

−78 °C, 5 min.

ii)Br

Me

MeOOBn

OMe

N

OBn

H

HNBoc

O

SPh

48% yield

OTIPSOTIPS

Scheme 3.29. Initial C13 Alkylation

Me

MeOOBn

OMe

N

OBn

H

HNBoc

O

SPhi) KHMDS, THF

−78 °C, 5 min.

ii)I

78% yield

OTIPS

Scheme 3.30. Optimized C13 Alkylation

305

300

306

304

305300

115

Inversion of the C13 stereocenter was performed as before, with care taken to

deoxygenate the reaction solution. The inverted product 307 was isolated in 73% yield

after purification and characterized as the cis-isomer (Scheme 3.31). On scale-up, a two

step procedure starting from 305 inverted the C13 stereocenter, then treatment of the

crude product mixture with 48% aqueous HF in acetonitrile removed the triisopropylsilyl

protecting group to furnish alcohol 308 in 83% yield over the two steps.

Me

MeOOBn

OMe

N

OBn

H

HNBoc

O

SPh

OTIPS

−78 °C, 30 sec.i) 2.4 equiv. t-BuLi, THF

ii) BHT

73% yield

Me

MeOOBn

OMe

N

OBn

H

HNBoc

O

SPh

OH

−78 °C, 30 sec.1) i) 2.2 equiv. t-BuLi, THF

ii) BHT

2) 48% aq. HF, CH3CN0 °C, 30 min.

83% yield

Scheme 3.31. Optimized C13 Inversion

305

305

307

308

116

With the three carbon unit installed with correct C13 stereochemistry the focus

turned to the formation of the C11-C15 bond. The approach would be to form the silyl

enol ether of the side-chain then cyclize to form the bicyclo[3.2.1] system. Alcohol 308

was converted to the silyl enol ether by oxidation to aldehyde 309 under Swern

conditions (Scheme 3.32),14 then treatment with TIPSOTf and triethylamine in diethyl

ether. The two step procedure furnished the trans-silyl enol ether in 88% yield.

TIPSOTf, Et3N0 °C to 25 °C, 16 h

Me

MeOOBn

OMe

N

OBn

H

HNBoc

O

SPh

OTIPS

Me

MeOOBn

OMe

N

OBn

H

HNBoc

O

SPh(COCl)2, DMSO

Et3N, CH2Cl2−78 °C to 25 °C, 9 min. O

Me

MeOOBn

OMe

N

OBn

H

HNBoc

O

SPh

OH

Scheme 3.32. Silyl Enol Ether Formation

308 309

31088% yield, two steps

117

3.1.5. FORMATION OF THE BICYCLO[3.2.1] SYSTEM

It was hoped that the silyl enol ether would be nucleophilic enough to cyclize and

form the desired bicyclo[3.2.1] system under the condition required for N-acyliminium

ion formation. Intramolecular cyclizations are well precedented in the literature15 and

specifically, the N-acyliminium cyclization of a silyl enol ether to a 5-membered ring has

been reported.16

The initial conditions utilized AgBF4 as the thiophile to promote formation of

N-acyliminium 312. Treatment of 310 with AgBF4 in dichloroethane gave a mixture of

unidentified products and only 12% yield of the cyclized product 311 (Scheme 3.33,

Table 3.02, entry 1). Changing the solvent system to THF made a dramatic difference and

the cyclized product 311 was formed in nearly 80% yield (entry 2). Stoichiometric

amounts of AgBF4 were necessary to consume all of the starting thioaminal 310. The

intermediate N-acyliminium ion 312 formed rapidly but cyclization appeared slow and

was complete after stirring for four hours at room temperature. However, at 40 °C the

cyclization was complete after 30 minutes (entry 2).

118

Me

MeOOBn

OMe

N

OBn

H

HNBoc

O

SPh

OTIPS NN

H

O

MeOMe

OBn

H

H

OBn

CHO

Boc

MeO

Table 3.2

Me

MeOOBn

OMe

N

OBn

H

HNBoc

O

OTIPS

-SPh

NN

H

O

MeOMe

OBn

H

H

OBn

Boc

MeO

O

SiF

H

H

15

Scheme 3.33. Cyclization to the Bicyclo[3.2.1] System

entry reagent solvent temp (°C) time yield (%)

1 AgBF4 DCE 25 2 h 12

2 AgBF4 THF 25 40

4 h 30 min.

78 74

3 AgBF4 Et2O 25 30 min. 0

4 AgBF4 PhCl 25 2 h 0

5 BF3·OEt2 CH2Cl2 25 2 h 0

6 Hg(O2CCF3)2 THF 25 30 min. 0

7 Δ PhCl 115 24 h 0

8 1) m-CPBA 2) Δ

CH2Cl2 PhCl

80

2 h 0

9 Bu3SnH, AIBN PhH 80 20 h 0

Table 3.02. Cyclization to the Bicyclo[3.2.1] System

310

312 313

311

119

Further investigation of the reaction proved surprising because under no other

conditions was the cyclized product observed. Changing the solvent to diethyl ether or

chlorobenzene led to the formation of a mixture of unidentified products (entries 3 and

4). Other lewis acids and thiophiles also gave poor results (entries 5 and 6). Heating to

promote elimination of thiophenoxide led to deprotection of the silyl enol ether leaving

the thioaminal untouched (entry 7). Conversion of the sulfide to the sulfoxide resulted in

a better leaving group but upon elimination with heating only unidentified products were

observed (entry 8). Radical cyclization conditions17 gave no reaction (entry 9). The

significance of the solvent selectivity was not determined until our work on renieramycin

G.

Minor impurities were observed during the cyclization in THF and after filtration

of the crude reaction mixture through a plug of silica gel only small amounts of

impurities were present. The cyclized product appeared to be a single diastereomer at the

C15 stereocenter. To determine the relative stereochemistry with certainty a crystalline

derivative was made. Success in obtaining a crystal structure was found with formation of

the oxime derivative 314, from treatment of aldehyde 311 in methanol with

hydroxylamine hydrochloride and potassium acetate (Scheme 3.34). A 1:1 mixture of cis-

and trans-isomers was isolated in 83% yield. Chromatography was employed to obtain

modest separation of the isomers. The more polar fractions gave a 4:1 mixture, cis:trans,

as a white solid which upon recrystallization produced X-ray quality needles. The X-ray

structure (appendix G) shows that the cyclization furnished the aldehyde on the outside

face giving the desired stereochemistry at C15 (Figure 3.03). The selectivity was likely

due to the axial orientation of the C1 side-chain, which provides steric hindrance for

cyclization with the silyl enol ether on the inside face. The X-ray structure shows that the

C1 side-chain remains in the axial orientation even with the bridged ring system present.

120

NN

H

O

MeOMe

OBn

H

H

OBn

CHO

Boc

MeO

HONH2·HCl

KOAc, EtOH25 °C , 2 h

NN

H

O

MeOMe

OBn

H

H

OBn

Boc

MeO

NHOH

83% yield

HH

15

Scheme 3.34. Oxime Formation

Figure 3.03. ORTEP Representation of Oxime 314 X-ray Structure

With an efficient yield of the bicyclo[3.2.1] system we focused our attention on

elaboration of the remainder of lemonomycin. N-Boc deprotection, amide reduction,

debenzylation, C1 glycol incorporation, and a final oxidation to the quinone were

311 314, X-ray

121

required. However, at this stage of the project Stoltz published his total synthesis of

(−)-lemonomycin.1 As described in Chapter 1 his strategy was distinctly different, in fact

if we were to synthesize lemonomycin we would not share a common intermediate. The

lack of overlap between the strategies meant that we would not be able to demonstrate, by

formal synthesis, the completion of lemonomycin. The enantioselective synthesis by

Stoltz was more efficient so with our strategy offering no further improvement

completion of the total synthesis of lemonomycin was abandoned. However, formation of

the lemonomycin aglycone amide (lemonomycinone amide, 317, Scheme 3.36) derivative

was investigated.

Aldehyde 311 was treated under hydrogenolysis conditions to give the diol 315 in

79% yield from the silyl enol ether 310 (Scheme 3.35). The C15 aldehyde did not exist as

the hydrated aldehyde present in (−)-lemonomycin. However, it was observed that when

deuterated methanol was used as an NMR solvent none of the aldehyde signals could be

observed in the proton and carbon NMR. Although the spectra did not show clean

formation of a new compound, it was evident that hydration of the aldehyde did occur.

Evaporation of this sample and then reinvestigation by 1H NMR analysis in deuterated

chloroform indicated reformation of aldehyde 315.

NN

H

O

MeOMe

OBn

H

H

OBn

CHO

Boc

MeO HN

N

H

O

MeOMe

OH

H

H

OH

CHO

Boc

MeO H

Pd(OH)2, MeOH

1 atm H2, 25 °C , 4 h

79% yield from cyclization

1515

Scheme 3.35. Hydrogenolysis

315 311

122

The N-Boc protecting group was removed under acidic conditions to afford amine

316 (Scheme 3.36). Monitoring the reaction by TLC proved difficult and reverse-phase

HPLC conditions were necessary to accurately characterize the deprotected product. 1H

NMR experiments verified the aldehyde existed solely in the hydrated form. In addition,

2D NMR experiments showed epimerization at the C15 stereocenter had not occurred.

An efficient route to quinone 317 involved deprotection of the N-Boc protecting

group of aldehyde 315 and oxidation of the crude product with ammonium cerium (IV)

nitrate to afford the quinone 317 in 35% yield over the two steps. Reverse-phase HPLC

solvents contained 0.1% TFA and hence the TFA salt of the product was isolated after

purification. Characterization of the final compound by 2D NMR showed consistent

stereochemistry at the C15 stereocenter, a distinct doublet for the C16 hydrogen, and

quinone shifts consistent with the reported spectra of (−)-lemonomycin.

3 N HCl

1:1 MeOH:H2O25 °C , 3 h

NN

H

O

MeOMe

OH

H

H

OH

CHO

Boc

MeO HN

NH

H

O

MeOMe

OH

H

H

OH

MeO H

OHHO

· HCl

(NH4)2Ce(NO3)6

H2O, 25 °C , 4 h NNH

H

O

MeO

O

H

H

OH

MeO H

OHHO

· TFA

35% yieldtwo steps

15

16

Scheme 3.36. Formation of Lemonomycinone Amide 317

316315

317

123

3.2. CONCLUSION

The approach to (−)-lemonomycin succeeded in developing new methodology for

the synthesis of tetrahydroisoquinoline alkaloids. The alkylation of isoquinoline with

benzyloxymethyllithium was performed in the presence of (−)-sparteine, showing modest

enantioselectivity. Unfortunately, the analogous reaction with the electron-rich

isoquinoline 262 gave multiple unidentified products. In the absence of (−)-sparteine, the

1,2-dihydroisoquinoline 263 could be formed. Elaboration of the 1,3-cis-substitution

required formation of oxazolidinone derivative 276 but stereoselectivity during ionic

hydrogenation was still maintained. Formation of the eastern fragment required

development of silyl-activated coupling conditions that protected the C3-hydroxyl while

activating the amine to give the amide 287 in excellent yield.

Construction of the bicyclo[3.2.1] system proceeded through the thioaminal 300

which was synthesized as one diastereomer from the corresponding hemi-aminal.

Alkylation via an enolate, generated from the amide, gave the wrong stereochemistry at

the C13 center, but proved completely diastereoselective. Thus, inversion of the center by

diastereoselective reprotonation conditions were very efficient. To close the three carbon

unit to the bicyclo[3.2.1] system, an intramolecular N-acyliminium cyclization with silyl

enol ether 310 was developed. The cyclization was best performed in THF with thiophile

AgBF4, other reaction conditions gave at best only trace amounts of 311. The C15

aldehyde was formed diastereoselectively and the X-ray structure of the oxime derivative

314 proved the desired stereochemistry had been achieved. The unique aldehyde was

eventually converted to the hydrated aldehyde 316 after N-Boc deprotection and a final

oxidation afforded quinone 317. The quinone represented lemonomycinone amide, a

124

317

congener of the initial target. The (±)-lemonomycinone amide 317 was synthesized from

aldehyde 230 in 26 steps with an overall yield of 3% (Scheme 3.37).

OMeMe

MeOCHO

26 steps

3% yield NNH

H

O

MeO

O

H

H

OH

MeO H

OHHO

· TFA

Scheme 3.37. Total Synthesis of (±)-Lemonomycinone Amide 317

Although the total synthesis of lemonomycin was abandoned, success was found

with our approach to this class of tetrahydroisoquinoline alkaloids. Application of this

new methodology to another tetrahydroisoquinoline alkaloid was investigated to

demonstrate its utility in a divergent strategy.

230

125

3.3. REFERENCES 1) Stoltz, B. M.; Cruz, E. G.; Ashley, E. R. J. Am. Chem. Soc. 2003, 125, 15000. 2) Kaufman, T. S. Synlett 1997, 1377. 3) Adams, J.; Plamondon, L.; McCormack, T. A.; Destree, A. T.; Behnke, M. L.; Grenier, L.; Soucy, F. J. Am. Chem. Soc. 1999, 121, 9967. 4) Larock, R. C.; Roesch, K. R. J. Org. Chem. 2002, 67, 86. 5) Williams, R. M.; Metobo, S.; Jin, W. Org. Lett. 2003, 5, 2095. In this publication the reduction of a similar 3,4-olefin of a tetrahydroisoquinoline intermediate could not be performed under high pressure without degradation of the molecule. The failure of reduction led to the termination of this approach. 6) Davies, S. G.; Blagg, J. J. Chem. Soc., Chem. Comm. 1986, 492. 7) Aldrich, J. V.; Murray, T. F.; Berman, F.; Maeda, D. Y. J. Med. Chem. 2000, 43, 5044.

8) Klebe, J. F. In Advances in Organic Chemistry; Taylor, E. C., Ed.; Wiley-Interscience: New York, 1972; Vol. 8, p 97. 9) Fukumoto, K.; Sukegawa, Y.; Shishido, K. Heterocycles 1986, 24, 641. 10) Hu, L.; Yu, C. Tetrahedron Lett. 2001, 42, 5167. 11) Williams, R. M.; Scott, J. D. J. Am. Chem. Soc. 2002, 124, 2951. 12) Jung, M. E.; Blum, R. B. Tetrahedron Lett. 1977, 18, 3794. 13) Davies, S. G.; Bull, S. D.; Epstein, S. W.; Ouzman, J. V. A. Tetrahedron: Asymmetry 1998, 9, 2795. 14) Swern, D.; Mancuso, A. J.; Huang, S. L. J. Org. Chem. 1978, 43, 2480 15) Maryanoff, B. E.; Zhang, H.-C.; Cohen, J. H.; Turchi, I. J.; Maryanoff, C. A. Chem. Rev. 2004, 104, 1431. 16) Speckamp, W. N.; Hiemstra, H.; Vijn, R. J. J. Org. Chem. 1988, 53, 3882. 17) Hart, D. J.; Atarashi, S.; Choi, J.-K.; Ha, D.-C.; Kuzmich, D.; Lee, C.-S.; Maesh, S.; Wu, S. C. J. Am. Chem. Soc. 1997, 119, 6226.

126

Chapter 4: The Total Synthesis of (±)-Renieramycin G

4.0. INTRODUCTION

Continuing our approach to the tetrahydroisoquinoline alkaloids we chose as our

new target renieramycin G (2g, Scheme 4.01). The total synthesis of renieramycin G had

not been previously reported. Furthermore, at the start of the project the only total

synthesis of a renieramycin was by Fukuyama,1 but Danishefsky recently published the

synthesis of cribrostatin IV (renieramycin H).2

We hoped to develop a divergent strategy for the synthesis of

tetrahydroisoquinoline alkaloids, something that has not been previously demonstrated.

To do this we wanted to use an advanced intermediate from our approach to

lemonomycin. The divergence between lemonomycin and renieramycin G occurs from

sulfide 300 (Scheme 4.01). Alkylation with iodide 306 installed the framework for the

bicyclo[3.2.1] system of lemonomycin. However alkylation with a benzyl halide 320

would install the framework for the bicyclo[3.3.1] system of renieramycin G. The choice

of protecting groups was important as stability to the alkylation and the diastereoselective

reprotonation conditions were required. It was anticipated that the reactivity would be

improved if the cyclization precursor 319 was a phenol (R1 = H). This would require a

differential protecting group (R1) to be present on the benzyl halide 320 which could be

readily removed in the presence of the benzyl ether and the N-Boc protecting groups.

After cyclization to 318, removal of the N-Boc protecting group and methylation were

required. The final steps include benzyl ether deprotection, oxidation to the

bis-isoquinolinequinone, and angelate ester formation. The angelate ester is known to

readily isomerize to the more stable cis-isomer.3 Thus, it was important to minimize the

number of steps with the angelate ester incorporated.

127

OMe

MeOO

NN

OOMe

Me

OMe

H

H

H

H

O

OMe

MeO

OMeMe

MeOOBn

NN

HOOMe

Me

OMeBoc

H

H

H

H

OBnO

1

3

NNBoc

OMeO

MeOMe

OBn

H

H

OBn

SPh

1

3

X

MeOMe

OMe

OR1+

1

31

11

13

3

4

21

6 10

8

22

19

17

15

14

26

24

NNBoc

OMeO

MeOMe

OBn

H

H

OBn

SPh OR1

OMe

MeOMe

H

Scheme 4.01. Retrosynthesis of Renieramycin G (2g)

2g 318

319

320

300

128

4.1. RESULTS AND DISCUSSION

4.1.1. BENZYL CHLORIDE FORMATION

Our first objective in the approach to renieramycin G was the synthesis of a

suitable benzyl halide that would fit our reaction profile. For this we chose the benzyl

chloride 321 (Figure 4.01). We hoped that the benzyl chloride would be a reactive

enough electrophile, but formation of the benzyl bromide was possible if this was a

problem. The trityl ether would provide differentiation from the benzyl ethers and N-Boc

(which are more stable to acid than the aryl trityl ether). It was anticipated that the trityl

group would be stable to both the alkylation conditions and the diastereoselective

reprotonation conditions. No literature precedence for this compound existed, but we

utilized known chemistry to synthesis the benzyl halide from commercially available

benzaldehyde 230 (Scheme 4.02). The same benzaldehyde was used to form the

tetrahydroisoquinoline of the western fragment, demonstrating the pseudo-symmetry of

the renieramycins.

MeOMe

MeO

Cl

OTrt

Figure 4.01. Benzyl Chloride 321

321

129

A solution of benzaldehyde 230 in 37% aqueous formaldehyde was treated under

chloromethylation conditions4 (Scheme 4.02), gaseous HCl and zinc (II) chloride. After

chromatography the benzyl chloride 322 was isolated as a white solid in 73% yield.

MeOMe

MeO

O H

MeOMe

MeO

O H

Cl37% aq. formaldehyde

HCl (g), ZnCl2reflux, 90 min.

73% yield

Scheme 4.02. Chloromethylation

A Baeyer-Villiger reaction was performed with benzyl chloride 322 furnishing the

formate ester 323 in 95% yield (Scheme 4.03). This type of Baeyer-Villiger in the

presence of a benzyl chloride had not been previously reported. There was concern that

by-products could form from the addition of m-CPBA at the benzyl chloride rather than

the benzaldehyde; however no by-products from this pathway were observed.

MeOMe

MeO

O H

Cl MeOMe

MeO

Cl

O

O

H

m-CPBA, CH2Cl20 °C to 25 °C, 3 h

95% yield

Scheme 4.03. Formate Ester Formation

322230

322 323

130

In our previous studies we used NaOH in MeOH to hydrolyze the formate ester,

however the analogous procedure with benzyl chloride 323 gave significant amounts of a

by-product that appeared to be from methoxide addition at the benzyl chloride. To avoid

this side-reaction the formate ester was reduced with LiBH4, to furnish the phenol 324 as

an unstable orange oil (Scheme 4.04). The crude phenol was protected as the trityl ether

upon treatment with recrystallized trityl chloride and triethylamine in CH2Cl2. The trityl

ether 321 was unstable and although the reaction appeared clean by TLC analysis

isolation of the pure product proved difficult. Silica gel chromatography was employed to

isolate the semi-pure ether, which upon recrystallization afforded the pure trityl ether 321

in 51% yield from the formate ester 323. The trityl ether was also found to be unstable to

storage at room temperature under argon in the dark.

MeOMe

MeO

Cl

O

O

H

LiBH4, THF

0 °C, 10 min.

MeOMe

MeO

Cl

OH

trityl chloride

Et3N, CH2Cl225 °C, 2 h

MeOMe

MeO

Cl

OTrt51% yieldtwo steps

Scheme 4.04. Trityl Ether 321 Formation

323 324 321

131

4.1.2. FORMATION OF THE BICYCLO[3.3.1] SYSTEM

With benzyl chloride 321 in hand, alkylation of the common intermediate 300

was conducted under identical conditions to those used for the approach to lemonomycin

with iodide 306. Treatment of a degassed solution of thioaminal 300 with KHMDS at

−78 °C followed by addition of benzyl chloride 321 as a solution in THF gave unreacted

starting material (Scheme 4.05). No alkylation product was observed and it is likely that

the benzyl chloride was not reactive enough. O-Alkylation was ruled out because the

starting benzyl chloride was recovered. If O-alkylation had occurred then upon workup

hydrolysis would have given the benzyl alcohol.

NNBoc

OMeO

MeOMe

OBn

H

H

OBn

SPhKHMDS, THF, −78 °C

−78 °C to 25 °C, 3 h

NNBoc

OMeO

MeOMe

OBn

H

H

OBn

SPh OTrtOMe

MeOMe

then 321

Scheme 4.05. Attempted Benzylation of 300

The instability of the benzyl chloride 321 made conversion to a more reactive

benzyl bromide an unattractive route. Instead, the reactivity of the enolate was enhanced.

Conducting the reaction in the presence of 18-crown-6 produced a change in reactivity

and the alkylated product was formed. However, there was noticeable by-product

formation and the alkylated product was formed in approximately 40% yield. It was

noted that deoxygenation of the reaction solution was essential and a dramatic increase in

reaction efficiency was found by starting with thioaminal 300, 18-crown-6, and benzyl

chloride 321 as a solution in THF followed by the addition of KHMDS (Scheme 4.06).

300 325

132

Combining the reagents reduced the time between deprotonation and alkylation,

minimizing by-product formation. The reaction was complete within 20 minutes and near

quantitative formation of the alkylated product 325 was observed. The alkylation

appeared to be diastereoselective.

18-crown-6, KHMDS

NNBoc

OMeO

MeOMe

OBn

H

H

OBn

SPh OTrtOMe

MeOMe

THF, −78 °C, 20 min

98% yield

Scheme 4.06. Optimized Benzylation of Thioaminal 300

It was noted that manipulation of 325 did not lead to degradation or loss of the

trityl protecting group. Whereas the benzyl chloride 321 was much more susceptible to

deprotection and degradation. However, as with most of the compounds in this series,

pronounced carbamate resonance was observed, and the trityl group was not stable to

heat during a variable temperature NMR experiment. We anticipated the trityl group to be

stable to diastereoselective reprotonation.

Initial attempts to invert the C13 stereocenter utilized the conditions employed in

our approach to lemonomycin. The benzylated product 325 was treated with 2.5

equivalents of tert-butyllithium then immediately reprotonated with butylated hydroxyl-

toluene (BHT) (Scheme 4.07). A 6:1 mixture of the desired product 326 and starting

material was isolated in 73% yield. Using in excess of three or four equivalents did not

affect the diastereomeric ratio.

321 300 +

325

133

−78 °C, 30 sec.i) 2.5 equiv. t-BuLi, THF

ii) BHT, THF NNBoc

OMeO

MeOMe

OBn

H

H

OBn

SPh OTrtOMe

MeOMe

Scheme 4.07. Diastereoselective Reprotonation

The isomers were difficult to separate until removal of the trityl group produced a

mixture of the corresponding phenols. The more polar phenols were then separated by

chromatography. A one-pot, two-step procedure was developed that inverted the C13

stereocenter and removed the trityl protecting group. The benzylated product 326 was

treated to the same reprotonation conditions as above (Scheme 4.08). The solution was

then treated with anhydrous HCl in diethyl ether, with warming to 0 °C and careful

monitoring of the reaction; small portions of HCl were added until the trityl ether was not

detected. After separation from the other isomer, phenol 327 was isolated in 73% yield

over the two steps.

−78 °C, 30 sec.ii) BHT, THF

1) i) 2.5 equiv. t-BuLi, THF

2) HCl, Et2O, 0 °C, 1 hN

NBoc

OMeO

MeOMe

OBn

H

H

OBn

SPh OHOMe

MeOMe

73% yield

Scheme 4.08. Optimized Formation of Phenol 327

325 + 325

326

73% combined yield 326:325 = 6:1

325

327

134

The phenol 327 represented the cyclization precursor to be used in the

N-acyliminium cyclization to form the bicyclo[3.3.1] system. The optimized conditions

employed in our approach to lemonomycin were employed first. Treatment of phenol

327, as a solution in THF, with AgBF4 in THF led to immediate starting material

consumption and the formation of more polar products (Scheme 4.09). However upon

aqueous workup and concentration of the organic fractions well over 300% mass

recovery was isolated.

NNBoc

OMeO

MeOMe

OBn

H

H

OBn

SPh OHOMe

MeOMe

OMeMe

MeOOBn

NN

HOOMe

Me

OMeBoc

H

H

H

H

OBnO

AgBF4

THF, 25 °C

Scheme 4.09. Attempted N-Acyliminium Alkylation

The reaction conditions are fairly simple with only AgBF4 being added, and the

extra weight did not appear to be coming from this reagent. A volatile oil was present in

large amounts, which upon 1H NMR analysis appeared to be one of the by-products

observed in our N-acyliminium cyclization of silyl enol ether 310 during our studies on

lemonomycin. This by-product possessed what appeared to be an aldehyde signal and

was originally thought to come from degradation of the starting silyl enol ether 310.

However, observation of the same by-product in the cyclization of phenol 327 suggested

it was unlikely that an aldehyde had formed from the degradation of 327. In addition the

mass recovery could not be explained if degradation of the starting phenol 327 was

assumed.

327 328

135

The answer appeared to be polymerization of the solvent THF. Tests showed that

THF was stable to AgBF4, but polymerization in the presence of a catalyst, our phenol

327, was observed. The polymer could be forming by two possible mechanisms. The first

mechanism could be acid catalyzed polymerization promoted by the HBF4 formed after

cyclization. The strong acid HBF4 is not formed during the cyclization of silyl enol ether

310, which could explain why more polymer was observed during the cyclization of

phenol 327. A second mechanism is needed to rationalize the formation of the same

polymer in the absence of HBF4. We postulate that an N-acyliminium ion 329, formed

from either silyl enol ether 310 or phenol 327, can react with THF to form an oxonium

intermediate 330 (Scheme 4.10). This intermediate can then react with another molecule

of THF leading to polymerization.

NN

OMeO

MeOMe

OBn

H

H

OBn

Boc

NN

OMeO

MeOMe

OBn

H

H

OBn

BocOO

AgBF4

THF, 25 °C

NN

OMeO

MeOMe

OBn

H

H

OBn

BocO

O

O

R

RR

polymerization

Scheme 4.10. Observed Polymerization

327 or 310

329

331 330

136

When the solvent was changed to chlorobenzene the by-product was not observed

(Scheme 4.11). N-Acyliminium formation was rapid and a much more polar compound

was the major product. Isolation of this compound revealed no tert-butyl proton signals

were present in 1H NMR suggesting the absence of the N-Boc protecting group. Further

characterization revealed that the desired cyclization had occurred but loss of N-Boc had

furnished amine 332, presumably due to the presence of HBF4 formed in the reaction.

NNBoc

OMeO

MeOMe

OBn

H

H

OBn

SPh OHOMe

MeOMe

OMeMe

MeOOBn

NNH

HOOMe

Me

OMeH

H

H

H

OBnO

AgBF4, PhCl

25 °C, 10 min.

56% yield

Scheme 4.11. N-Acyliminium Cyclization

The cyclization to 332 was very rapid and none of the N-Boc cyclized

intermediate could be observed or isolated. The amine product 332 was formed within 10

minutes of AgBF4 addition. The cyclization of phenol 327 was significantly more rapid

than the cyclization of silyl enol ether 310, which required four hours at room

temperature after N-acyliminium formation. The relative rates are not necessarily

surprising because formation of the 6-membered bicyclo[3.3.1] system is less strained

than the 5-membered bicyclo[3.2.1] system. There is also a difference of nucleophilicity

between the phenol and the silyl enol ether.

Cyclization of the phenol 327 proceeded rapidly and cleanly in chlorobenzene.

The loss of the N-Boc protecting group was a fortuitous result as it would have been

removed in the next reaction. Under optimized conditions reductive methylation with

327 332

137

sodium cyanoborohydride, acetic acid and formaldehyde (Scheme 4.12) afforded

N-methylamine 333. After chromatography 333 was isolated in 65% yield over the two

steps. The spectroscopic data were consistent for formation of the desired cyclized

product. Attempts to form a crystalline derivative were unsuccessful for this substrate.

1) AgBF4, PhCl25 °C, 10 min. OMe

Me

MeOOBn

NN

HOOMe

Me

OMeMe

H

H

H

H

OBnO

2) NaBH3CN, AcOH, 37% aq. formaldehyde, MeOH, 25 °C, 1h

65% yield

Scheme 4.12. Optimized N-Acyliminium Cyclization

4.1.3. COMPLETION OF THE SYNTHESIS

The final reactions in our approach to renieramycin G required deprotection of the

benzyl ethers, oxidation to the bis-isoquinolinequinone, and angelate ester formation.

Treatment of N-methyl 333 with Pd(OH)2 under a balloon atmosphere of hydrogen led to

debenzylation (Scheme 4.13). The debenzylation was much slower than in our approach

to lemonomycin and under unoptimized conditions the mono-benzylated product 334 was

isolated in 85% yield. After purification by chromatography the mono-benzyl ether 334

was recrystallized from methanol and ethyl acetate yielding small amounts of X-ray

quality needles. An X-ray structure of the mono-benzyl ether 334 (appendix H)

confirmed that the cyclization had occurred as expected with the correct stereochemistry.

327

333

138

OMeMe

MeOOBn

NN

HOOMe

Me

OMeMe

H

H

H

H

OBnO

Pd(OH)2, MeOH

1 atm H2, 1 h

OMeMe

MeOOH

NN

HOOMe

Me

OMeMe

H

H

H

H

OBnO

85% yield

Scheme 4.13. Incomplete Hydrogenolysis

The hydrogenolysis was optimized by the use of at least 40% by weight catalyst

and stirred for six hours under an atmosphere of hydrogen (Scheme 4.14). The triol 335

was isolated in 94% yield. The triol was not very soluble and we were unable to find

appropriate recrystallization conditions.

OMeMe

MeOOBn

NN

HOOMe

Me

OMeMe

H

H

H

H

OBnO

Pd(OH)2, MeOH

1 atm H2, 6 h

OMeMe

MeOOH

NN

HOOMe

Me

OMeMe

H

H

H

H

OHO

94% yield

Scheme 4.14. Optimized Hydrogenolysis

The order in which to do the remaining reactions (phenolic oxidation and angelate

ester formation) was our next concern. Our initial approach was to oxidize to the di-

quinone and then esterify, however, in Danishefsky’s work on cribrostatin IV (78) he

reported that “despite extensive efforts” formation of the angelate ester of the quinone

336 was unsuccessful (Scheme 4.15).2 Their synthetic route was altered in order to go

334, X-ray 333

335 333

139

87

through the phenol 86, which underwent esterification and silyl deprotection to afford 88

in 75% yield over the two steps. Our phenol 335 was slightly different than

Danishefsky’s phenol 86 in that the phenolic hydroxyls present in our substrate were

likely to be more reactive. This could lead to multiple ester products, so we decided to

proceed through oxidation first followed by esterification.

OMe

MeOO

NN

HOOMe

Me

OHMe

H

H

H

OHO

O

OMe

MeOO

NN

HOOMe

Me

OHMe

H

H

H

OO

O

O

Me

Me

H

OTBSMe

MeOOMe

NN

HOOMe

Me

OHMe

H

H

H

OHO

O

OHMe

MeOOMe

NN

HOOMe

Me

OHMe

H

H

H

OO

O

O

Me

Me

H

Me

MeClO

H CH2Cl2

1)

2) AcOH, TBAF, THF

75% yield

Scheme 4.15. Danishefsky’s Angelate Ester Formation

336

86

88

cribrostatin IV (renieramycin H, 78)

140

The triol 335 was oxidized to the diquinone using Stoltz’s conditions5 by

treatment with ammonium cerium (IV) nitrate (Scheme 4.16). The oxidation was

complete within 15 minutes and the diquinone 337 was isolated in 55% yield after silica

gel chromatography.

OMeMe

MeOOH

NN

HOOMe

Me

OMeMe

H

H

H

H

OHO

(NH4)2Ce(NO3)6CH3CN:H2O 6:1

−5 °C, 10 min.

OMe

MeOO

NN

OOMe

Me

OMe

H

H

H

H

OHO

55% yield

Scheme 4.16. Bis-Isoquinolinequinone Formation

For esterification we followed the procedure utilized by Joseph-Nathan and

co-workers in their work on butanoic acid derivatives.6 Excess angeloyl chloride7 (87)

was added to a solution of 337 in CH2Cl2 (Scheme 4.17). After standing in the dark for

24 hours the solution was concentrated under vacuum and purified directly by

reverse-phase HPLC. The initial HPLC conditions chosen were normal-phase, with

elution in isopropanol and hexanes, however the product was not stable in these solvents.

Under reverse-phase conditions the solvent system was H2O and acetonitrile, and in

contrast to our purification of lemonomycinone amide 317 TFA was not present. The

product was very stable to these conditions and trailing was not observed during

separation. After purification (±)-renieramycin G (2g) was isolated in 74% yield.

335 337

141

OMe

MeOO

NN

OOMe

Me

OMe

H

H

H

H

OHO

N

MeO

MeOO

OO

NH

OOMe

Me

OMe

OMe

Me

H

HH

H

Me

MeClO

H

CH2Cl2

74% yield

26

Scheme 4.17. Angelate Ester Formation

We attempted to compare our sample with an authentic one. However, after

discourse with Prof. Davidson, the author responsible for the isolation,7 he informed us

that he no longer had an authentic sample in his possession. Furthermore he was unable

to provide copies of the original spectra. In the isolation paper a list of the observed

proton and carbon signals was published and based on the initial spectra we acquired of

our sample of renieramycin G in CD2Cl2 it appeared we had indeed formed the correct

structure. The key 1H NMR signal was the C26 hydrogen which appears around 5.9 ppm

for the angelate ester but around 6.6 ppm for the tiglate ester, the trans-isomer.8 Thus,

epimerization of the double bond had not occurred. Unfortunately we were not satisfied

with the spectra in CD2Cl2 because trace water, CH2Cl2, and acid led to signal broadening

and overlap. For an improvement in the resolution of our spectra we chose C6D6 as the

NMR solvent. The signals were much sharper, trace C6H6 and H2O appeared out of the

range of renieramycin G signals so there was no overlap. 2D NMR experiments including

COSY, NOESY, HSQC, and HMBC were conducted and all data appeared consistent

with previously published observations of similar structures.

87

337

(±)-renieramycin G (2g)

142

4.2. CONCLUSION

The total synthesis of (±)-renieramycin G successfully demonstrated a divergent

approach to the tetrahydroisoquinoline alkaloids. In chapter three we synthesized a

(±)-lemonomycinone amide (317) from thioaminal 300 and from that same intermediate

we synthesized (±)-renieramycin G (2g). The alkylation of thioaminal 300 with benzyl

chloride 321 proved more efficient than the alkylation with iodide 306. Construction of

the bicyclo[3.3.1] system via an N-acyliminium cyclization was also more efficient than

formation of the bicyclo[3.2.1] system of 317. This can be attributed to the formation of a

less strained 6-membered ring. The incomplete hydrogenolysis provided the mono-benzyl

ether 334 as a crystalline solid. The X-ray structure of which was important to determine

with certainty that the desired cyclization had occurred and the C13 stereocenter had been

inverted. The synthesis of (±)-renieramycin G was completed after oxidation to the

diquinone and angelate ester formation. Although Danishefsky reported problems with

esterification of the C1 side-chain alcohol of his diquinone, difficulties were not observed

for our substrate. The total synthesis of (±)-renieramycin G from commercially available

benzaldehyde 230 was completed in 25 steps with an overall yield of 4% (Scheme 4.18).

OMeMe

MeOCHO

25 steps

4% yield

Scheme 4.18. Total Synthesis of (±)-Renieramycin G (2g)

(±)-renieramycin G (2g)

230

143

4.3. REFERENCES

1) Fukuyama, T.; Linton, S. D.; Tun, M. M. Tetrahedron Lett. 1990, 31, 5989. 2) Danishefsky, S. J.; Chan, C.; Heid, R.; Zheng, S.; Guo, J.; Zhou, B.; Furuuchi, T. J. Am. Chem. Soc. 2005, 127, 4596. 3) Buckles, R. E.; Mock, G. V. J. Org. Chem. 1950, 15, 680. 4) Geissman, T. A.; Schlatter, M. J.; Webb, I. D.; Roberts, J. D. J. Org. Chem. 1946, 11, 741. 5) Stoltz, B. M.; Cruz, E. G.; Ashley, E. R. J. Am. Chem. Soc. 2003, 125, 15000. 6) Joseph-Nathan, P.; Torres-Valencia, J. M.; Cerda-Garcia-Rojas, C. M. Tetrahedron: Asymmetry 1998, 9, 757. 7) Davidson, B. S. Tetrahedron Lett. 1992, 33, 3721. 8) Beeby, P. J. Tetrahedron Lett. 1977, 38, 3379.

144

Chapter 5: Experimentals

5.0. GENERAL INFORMATION

Melting points were taken on a Thomas-Hoover capillary tube apparatus and are

uncorrected. Infrared spectra were recorded on a Nicolet FT-IR spectrophotometer neat

unless otherwise indicated. 1H NMR spectra were recorded on either a General Electric

QE-300 spectrometer at 300 MHz or a Varian spectrometer at 500 MHz in the indicated

solvent and are reported in ppm relative to tetramethylsilane and referenced internally to

the residually protonated solvent. 13C NMR spectra were recorded on a General Electric

QE-300 at 75 MHz or a Varian spectrometer at 125 MHz in the solvent indicated and are

reported in ppm relative to tetramethylsilane and referenced internally to the residually

protonated solvent. Mass spectra were obtained on a VG ZAB2E or a Finnigan TSQ70.

Routine monitoring of reactions was performed using Merck 60 F254 silica gel,

aluminum-backed TLC plates. Flash column chromatography was performed using EMD

silica gel (particle size 0.040-0.063 μm). Reverse-phase HPLC purification was done

with a WATERS 1525 Binary HPLC Pump and WATERS 2996 Photoiodide Array

Detector using C18 packed analytical (VYDAC, 10 μm packing, 4.6 x 250 mm), semi-

preparatory (WATERS, μBondapak, 7.8 x 300 mm), and/or preparatory column

(VYDAC, 10 μm packing, 22 x 250 mm). Optical purities were determined by HPLC

analysis using an analytical chiral column (Daicel, Chiralcel OD; eluent,

hexane/isopropyl alcohol; UV detector, 254 nm) with a Beckman Coulter System Gold

126P solvent module and Beckman Coulter System Gold 168 detector.

Solvents and commercial reagents were purified in accordance with Perrin and

Armarego or used without further purification. All reactions were setup under an

atmosphere of argon and only degassed when specified.

145

5.1. EXPERIMENTAL CONDITIONS

1-Phenylsulfanylmethyl-1H-isoquinoline-2-carboxylic acid methyl ester (157)

1-Phenylsulfanylmethyl-1H-isoquinoline-2,4-dicarboxylic acid dimethyl ester (158)

NCO2Me

PhS

NCO2Me

SPh

CO2Me

To a stirred solution of thioanisole (0.594 mL, 5.06 mmol) and TMEDA (0.764 mL, 5.06

mmol) in THF (8.0 mL) at 0 °C was added n-BuLi (2.30 M in hexanes, 2.20 mL, 5.06

mmol) dropwise within 10 min. The cloudy, yellow solution was allowed to warm to 25

°C and was stirred for 1 h. The solution of phenylthiomethyllithium and TMEDA was

added dropwise within 10 min. to a degassed solution of isoquinoline (0.30 mL, 2.20

mmol) in toluene (18 mL) at –78 °C under an atmosphere of argon. The resulting orange

solution was stirred for 5 min. at –78 °C. The solution was warmed to –20 °C and stirred

for two days. The brown solution was quenched with methyl chloroformate (0.510 mL,

6.60 mmol) and the color of the clear solution changed to pale orange. The solution

stirred for 15 min. and was then cooled to 0 °C, and diluted with sat. aq. NaHCO3 (40

mL). The reaction warmed to room temperature and the aqueous layer was extracted with

EtOAc (2 x 75 mL). The organic extracts were combined, dried (Na2SO4), filtered, and

concentrated under vacuum. The crude yellow oil was purified by flash column

chromatography (SiO2, 5-15% EtOAc in hexanes) to provide 157 (0.267 g, 39% yield) as

a colorless oil and 158 (0.228 g, 28% yield) as a white solid. 157: Rf = 0.37 (20% EtOAc

in hexanes); IR (thin film) 2947, 1716, 1635, 1456, 1439, 1354, 1330, 1232 cm-1; 1H

146

NMR (300 MHz, CDCl3) δ 7.36-7.04 (9 H, m), 6.91 (0.5 H, d, J = 7.7 Hz), 6.69 (0.5 H,

d, J = 7.7 Hz), 5.95 (0.5 H, d, J = 7.8 Hz), 5.85 (0.5 H, d, J = 7.8 Hz), 5.52 (0.5 H, t, J =

7.0 Hz), 5.32 (0.5 H, t, J = 7.0 Hz), 3.79 (2 H, s), 3.71 (1 H, s), 3.27-3.02 (2 H, m) ppm; 1H NMR (500 MHz, DMSO-d6, 120 °C) δ 7.33-7.16 (8 H, m), 7.12 (1 H, d, J = 7.7 Hz),

6.78 (1 H, d, J = 7.7 Hz), 5.97 (1 H, d, J = 7.8 Hz), 5.44 (1 H, t, J = 6.7 Hz), 3.72 (3 H, s),

3.18 (1 H, dd, J = 13.7, 6.8 Hz), 3.13 (1 H, dd, J = 13.7, 6.7 Hz) ppm; 13C NMR (500

MHz, DMSO-d6, 120 °C) δ 152.5, 135.3, 130.0, 129.4, 128.2, 128.1, 127.4, 126.2, 126.0,

125.3, 123.9, 123.8, 108.0, 53.7, 52.4, 36.9 ppm; HRMS calcd. for C18H18NO2S (MH+)

312.1058 Da. Found 312.1049 Da. HPLC conditions for separation of enantiomers using

analytical column Chiracel OD: loaded 15 μl of 0.5 mg/mL sample in 1%

isopropanol/hexanes, gradient 2%-5% over 15 min., enantiomers elute at 11.9 min. and

13.9 min. in a ratio of 1:1. 158: mp = 104-107 °C; Rf = 0.23 (20% EtOAc in hexanes); IR

(thin film) 2953, 1732, 1705, 1614, 1438, 1371, 1243, 1168 cm-1; 1H NMR (300 MHz,

CDCl3) δ 8.23 (1 H, brs), 8.07 (0.5 H, brs), 7.87 (0.5 H, brs), 7.33-7.10 (8 H, m), 5.48

(0.5 H, brs), 5.35 (0.5 H, brs), 3.84 (6 H, s), 3.15 (1 H, brs), 3.05 (1 H, dd, J = 13.7, 7.0

Hz) ppm; 1H NMR (500 MHz, DMSO-d6, 120 °C) δ 8.10 (1 H, d, J = 8.0 Hz), 7.82 (1 H,

s), 7.33- 7.18 (8 H, m), 5.47 (1 H, t, J = 6.5 Hz), 3.81 (3 H, s), 3.80 (3 H, s), 3.20 (2 H, d,

J = 6.5 Hz) ppm; 13C NMR (500 MHz, DMSO-d6, 120 °C) δ 164.5, 152.0, 134.9, 134.2,

129.9, 128.2, 127.4, 126.9, 126.5, 126.4, 125.5, 123.7, 108.9, 54.4, 53.3, 50.6, 37.3 ppm;

HRMS calcd. for C20H20NO4S (MH+) 370.1113 Da. Found 370.1115 Da.

147

Optimized Formation of 1-phenylsulfanylmethyl-1H-isoquinoline-2-carboxylic acid

methyl ester (157)

NCO2Me

PhS

To a stirred solution of thioanisole (60 μL, 0.51 mmol) and (−)-sparteine (117 μL, 0.51

mmol) in THF (1.0 mL) at 0 °C was added n-BuLi (1.45 M in hexanes, 350 μL, 0.51

mmol) dropwise within 10 min. The cloudy, yellow solution was allowed to warm to 25

°C and was stirred for 1 h. The solution of phenylthiomethyllithium and (−)-sparteine was

added dropwise within 10 min. to a degassed solution of isoquinoline (50 μL, 0.43 mmol)

in toluene (3.5 mL) at –78 °C under an atmosphere of argon. The resulting orange

solution was stirred for 5 min. at –78 °C. The solution was warmed to 25 °C and stirred

for 1.5 h. The orange/brown solution was quenched with methyl chloroformate (100 μL,

1.28 mmol) and the color of the clear solution changed to canary yellow. The solution

stirred for 15 min. and was then cooled to 0 °C, and diluted with 1.5 mL of sat. aq.

NaHCO3. The reaction warmed to room temperature and the aqueous layer was extracted

with toluene (3 x 5 mL). The organic extracts were combined, dried (Na2SO4), filtered,

and concentrated under vacuum. The crude yellow oil was purified by flash column

chromatography (SiO2, 3-7% EtOAc in hexanes) to provide 157 (121 mg, 91% yield) as a

colorless oil: characterized as above.

148

1-phenylsulfanylmethyl-3,4-dihydro-1H-isoquinoline-2-carboxylic acid methyl ester

(170)

NCO2Me

PhS

To a solution of 157 (44 mg, 0.14 mmol) in CH2Cl2 (0.20 mL) at 0 °C was added

triethylsilane (112 μL, 0.70 mmol), followed by dropwise addition within 10 min. of TFA

(55 μL, 0.72 mmol). The reaction was allowed to warm to 25 °C and stirred for 1 h. The

reaction was quenched with ice, diluted with sat. aq. NaHCO3 (5 mL) and extracted with

CH2Cl2 (3 x 10 mL). The combined extracts were washed with sat. aq. NaCl (5 mL),

dried (Na2SO4), filtered, and concentrated under vacuum. The crude oil was purified by

flash column chromatography (SiO2, 10% EtOAc in hexanes) to provide 170 (43 mg,

95% yield) as a colorless oil: Rf = 0.31 (20% EtOAc in hexanes). IR (thin film) 2949,

1700, 1446, 1331, 1223, 743 cm-1; 1H NMR (300 MHz, CDCl3) δ 7.43-7.39 (2 H, m),

7.34-7.25 (2 H, m), 7.20-7.16 (5 H, m), 5.43 (0.45 H, t, J = 6.6 Hz), 5.25 (0.55 H, t, J =

6.6 Hz), 4.14-4.09 (0.55 H, m), 3.88-3.82 (0.45 H, m), 3.76 (1.4 H, s), 3.66 (1.6 H, s),

3.50-3.35 (1 H, m), 3.32-3.26 (2 H, m), 2.96-2.76 (2 H, m) ppm; 13C NMR (75 MHz,

CDCl3) δ 156.4, 156.2, 136.6, 136.3, 135.7, 134.6, 134.5, 129.7, 129.3, 129.0, 128.8,

127.9, 127.5, 127.4, 126.4, 126.2, 76.8, 54.5, 54.0, 52.9, 52.7, 40.4, 40.1, 39.3, 38.2, 28.7,

28.3 ppm; HRMS calcd. for C18H20NO2S (MH+) 314.1215 Da. Found 314.1215 Da.

149

3-methyl-1-phenylsulfanylmethyl-1H-isoquinoline-2-carboxylic acid methyl ester

(172)

N

PhS

CO2Me

Me

To a stirred solution of degassed thioanisole (60 μL, 0.51 mmol) and (−)-sparteine (117

μL, 0.51 mmol) in THF (1.0 mL) at 0 °C was added n-butyllithium (1.45 M in hexanes,

350 μL, 0.51 mmol) dropwise within 10 min. The cloudy, yellow solution was allowed to

warm to 25 °C and was stirred for 1 h. The solution of phenylthiomethyllithium and

(−)-sparteine was added dropwise within 10 min. to a degassed solution of 3-methyl

isoquinoline (30 mg, 0.21 mmol) in toluene (3.5 mL) at –78 °C under an atmosphere of

argon. The resulting orange solution was stirred for 5 min. at –78 °C, the reaction was

then warmed to 25 °C and was stirred for 1 h. The orange/brown solution was quenched

with methylchloroformate (100 μL, 1.28 mmol) and the color changed to pale yellow.

The solution was stirred for 15 min., cooled to 0°C, and added sat. aq. NaHCO3 (1.5 mL).

The reaction warmed to 25 °C and was extracted with toluene (3 x 5 mL). The organic

extracts were combined, dried (Na2SO4), filtered, and concentrated under vacuum. The

crude yellow oil was purified by flash column chromatography (SiO2, 3-7% EtOAc in

hexanes) to provide 172 (61 mg, 90% yield) as a colorless oil: Rf = 0.40 (25% EtOAc in

hexanes); IR (thin film) 2950, 1715, 1438, 1340, 1317, 1262, 739 cm-1; 1H NMR (300

MHz, C6D6) δ 7.33 (2 H, m), 7.05-6.99 (3 H, m), 6.92-6.77 (4 H, m), 5.71 (2 H, m), 3.30

(3 H, s), 3.12 (1 H, dd, J = 13.6, 8.9 Hz), 2.84 (1 H, dd, J = 13.6, 7.0 Hz), 2.19 (3 H, brs)

ppm; 13C NMR (75 MHz, CD6) δ 136.9, 132.7, 131.1, 129.4, 128.3, 127.1, 126.6, 126.4,

150

126.1, 125.9, 124.3, 113.9, 55.9, 52.3, 36.4, 21.5 ppm; HRMS calcd. for C19H20NO2S

(MH+) 326.1215 Da. Found 326.1209 Da.

3-methyl-1-phenylsulfanylmethyl-3,4-dihydro-1H-isoquinoline-2-carboxylic acid

methyl ester (173)

NCO2Me

SPh

MeH

H

To a solution of 172 (61 mg, 0.19 mmol) in CH2Cl2 (0.35 mL) at 0 °C was added

triethylsilane (151 μL, 1.0 mmol), followed by dropwise addition within 10 min. of

trifluoroacetic acid (145 μL, 1.88 mmol). The reaction was stirred at 0 °C for 30 min. The

solution was warmed to 0 °C and was stirred 15 min. The reaction appeared complete by

TLC analysis and was quenched with ice, diluted with sat. aq. NaHCO3 (5 mL) until

basic and extracted with CH2Cl2 (3 x 10 mL). The combined extracts were washed with

brine (5 mL), dried (Na2SO4), filtered, and concentrated under vacuum. The crude oil was

purified by flash column chromatography (SiO2, 3-7% EtOAc in hexanes) to afford 173

(52 mg, 87% yield) as a colorless oil: Rf = 0.27 (25% EtOAc in hexanes); IR (thin film)

2951, 1696, 1440, 1387, 1324, 1085, 739 cm-1; 1H NMR (300 MHz, DMSO-d6) δ 7.38-

7.27 (4 H, m), 7.23-7.14 (5 H, m), 5.20 (1 H, br s), 3.98-3.93 (1 H, m), 3.50 (3 H, brs),

2.99 (1 H, dd, J = 16.0, 7.0 Hz), 2.85 (1 H, dd, J = 16.0, 9.9 Hz), 1.31 (3 H, d, J = 6.0

Hz); (300 MHz, DMSO-d6, 100 °C) 7.39-7.37 (2 H, m), 7.32-7.29 (2 H, m), 7.22-7.17 (5

H, m), 5.30 (1 H, dd, J = 7.5, 7.5 Hz), 4.11-4.04 (1 H, m), 3.59 (3 H, s), 3.36 (1 H, dd, J =

151

13.4, 7.5 Hz), 3.31 (1 H, dd, J = 13.4, 7.7 Hz), 3.03 (1 H, dd, J = 6.9, 16.0 Hz), 2.84 (1 H,

dd, J = 16.0, 9.2 Hz), 1.36 (3 H, d, J = 6.3 Hz) ppm; 13C NMR (75 MHz, C6D6) δ 156.5,

136.5, 136.3, 133.8, 129.4, 129.2, 129.1, 128.2, 127.9, 127.6, 127.5, 127.3, 126.5, 126.2,

55.7, 55.4, 52.73 48.1, 40.1, 40.0, 35.4, 23.5, 23.2, 23.0 ppm; HRMS calcd. for

C19H22NO2S (MH+) 328.1371 Da. Found 328.1369 Da.

tert-Butyl-[1-(2-iodo-phenyl)-meth-(E)-ylidene]-amine (179)

NtBu

I

To a solution of 2-iodobenzaldehyde2 (1.0 g, 4.31 mmol) in pentane (22 mL) was added

4Å molecular sieves and tert-butylamine (0.91 mL, 8.62 mmol). The reaction was stirred

at 25 °C for 12 h. The solution was filtered through Celite® and washed with benzene,

then concentrated under vacuum to afford 179 (1.12 g, 92% yield as a pale yellow oil: Rf

= 0.39 (10% EtOAc in hexanes); bp = 78 °C at 0.4 mmHg; IR (neat): 2967, 2928, 2903,

1633, 1464, 1434, 1370, 1271, 1221, 1202, 1012, 752 cm-1; 1H NMR (300 MHz, CDCl3):

δ 8.41 (1 H, s), 7.93 (1 H, d, J = 7.8 Hz), 7.84 (1 H, d, J = 7.9 Hz), 7.37 (1 H, t, J = 7.5

Hz), 7.08 (1 H, t, J = 7.8 Hz), 1.33 (9 H, s) ppm; 13C NMR (75 MHz, CDCl3): δ 159.2,

139.5, 138.0, 131.7, 128.8, 128.6, 100.47, 58.1, 29.9 ppm; HRMS calcd. for C11H15NI

(MH+) 288.0249 Da. Found 288.0247 Da.

152

Dibenzyl-isoquinolin-3-ylmethyl amine (178)

NNBn2

To a stirred solution of imine 179 (0.711 g, 2.48 mmol) and

1-(dibenzylamino)-2-propyne3 (187) (0.70 g, 2.97 mmol) in triethylamine (9.9 mL) was

added PdCl2(PPh3)2 (35 mg, 0.05 mmol) and CuI (5 mg, 0.03 mmol). The mixture was

heated to 55 °C and stirred for 2 h. The mixture was cooled to 25 °C, filtered through

Celite®, and the filtrate was concentrated under vacuum. The resulting residue was

dissolved in anhydrous DMF (25 mL), and CuI (50 mg, 0.30 mmol) was added. The

reaction mixture was flushed with argon, sealed, heated to 100 °C, and stirred for 14 h.

The solution was cooled to 25 °C, diluted with sat. aq. NH4Cl (50 mL), and extracted

with Et2O (2 x 50 mL). The combined extracts were dried (Na2SO4), filtered, and

concentrated under vacuum. The resulting residue was purified by flash column

chromatography (SiO2, 3-20% EtOAc-hexane) to afford 178 (0.705 g, 84% yield) as a

yellow oil: Rf = 0.22 (25% EtOAc in hexanes); IR (thin film): 3059, 3027, 2797, 1629,

1588, 1494, 1454, 1366, 1128, 974, 887, 745 cm-1; 1H NMR (300 MHz, CDCl3): δ 9.22

(1 H, s), 7.96-7.94 (2 H, m), 7.85 (1 H, d, J = 8.2 Hz), 7.68 (1 H, t, J = 7.3 Hz), 7.55 (1 H,

t, J = 7.8 Hz), 7.52-7.48 (4 H, m), 7.38-7.33 (4 H, m), 7.23-7.28 (2 H, m), 3.97 (2 H, s),

3.74 (4 H, s) ppm; 13C NMR (75 MHz, CDCl3): δ 153.5, 152.0, 139.6, 136.6, 130.4,

128.9, 128.4, 127.9, 127.6, 127.1, 126.8, 126.7, 118.6, 59.7, 58.4 ppm; m/z (C.I.) 339

(100%, MH+).

153

[(Dibenzylamino)-methyl]-1-phenylsulfanylmethyl-1H-isoquinoline-2-carboxylic

acid methyl ester (188)

NNBn2

CO2Me

PhS

To a stirred solution of thioanisole (0.26 mL, 2.18 mmol) and (−)-sparteine (0.50 mL,

2.18 mmol) in anhydrous THF (2.6 mL) at 0 °C was added n-BuLi (2.10 M in hexanes,

1.0 mL, 2.18 mmol) dropwise within 10 min. The cloudy, yellow solution was allowed to

warm to 25 °C and was stirred for 1 h. The above solution of phenylthiomethyllithium

and (−)-sparteine was added dropwise within 10 min. to a degassed solution of 178

(0.489 g, 1.45 mmol) in toluene (12 mL) at –78 °C under an atmosphere of argon. The

resulting orange solution was allowed to warm to 25 °C over 15 min. The solution

became black in color and methylchloroformate (1.1 mL, 13.8 mmol) was added. The

pale brown and clear solution was cooled to 0 °C, treated with sat. aq. NaHCO3 (10 mL),

and then extracted with EtOAc (3 x 25 mL). The organic extracts were combined, dried

(Na2SO4), filtered, and concentrated under vacuum. The crude brown oil was purified by

flash column chromatography (SiO2, 5%-10% EtOAc in hexanes) to provide 188 (0.651

g, 86% yield) as a yellow oil: Rf = 0.22 (20% EtOAc in hexanes); IR (thin film): 3060,

3026, 2928, 2797, 1716, 1636, 1439, 1398, 1343, 1320, 1263, 1113, 1065, 1027 971 cm-

1; 1H NMR (300 MHz, CDCl3): δ 7.44-7.09 (19 H, m), 6.62 (1 H, s), 5.45-5.40 (1 H, m),

5.30 (2 H, s), 3.74-3.45 (9 H, m), 3.18 (1 H, dd, J = 13.3, 7.8 Hz), 2.98 (1 H, dd, J = 13.3,

7.1 Hz) ppm; 1H NMR (500 MHz, toluene-d8): δ 7.40-7.39 (4 H, m), 7.25-7.22 (3 H, m),

7.20-7.06 (4 H, m), 7.02-6.97 (4 H, m), 6.93-6.89 (3 H, m), 6.87-6.85 (1 H, m), 6.48 (1

H, s), 5.66 (1 H, brs), 3.89 (1 H, d, J = 15.4 Hz), 3.88 (2 H, d, J = 13.9 Hz), 3.54-3.47 (3

154

H, m), 3.21-3.18 (4 H, m), 2.84 (1 H, dd, J = 13.2, 7.1 Hz) ppm; 13C NMR (125 MHz,

toluene-d8): δ 154.1, 139.3, 136.9, 133.2, 130.7, 129.4, 129.1, 128.6, 128.0, 127.9, 127.7,

127.4, 127.0, 126.8, 126.5, 125.2, 125.0, 124.9, 124.6, 114.6, 58.1, 56.9, 56.2, 52.2, 37.0

ppm; 1H NMR (500 MHz, toluene-d8, 100 °C): δ 7.35-7.34 (4 H, m), 7.24-7.22 (2 H, m),

7.16-7.05 (4 H, m), 7.04-7.02 (1 H, m), 7.00-6.96 (4 H, m), 6.93-6.89 (4 H, m), 6.48 (1

H, s), 5.60 (1 H, t, J = 7.3 Hz), 3.86 (1 H, dd, J = 16.0, 1.1 Hz), 3.76 (2 H, d, J = 13.9

Hz), 3.63 (1 H, d, J = 16.0 Hz), 3.59 (2 H, d, J = 13.9 Hz), 3.29 (3 H, s), 3.18 (1 H, dd, J

= 13.2, 7.5 Hz), 2.91 (1 H, dd, J = 13.3, 7.0 Hz) ppm; m/z (C.I.) 521 (100%, MH+).

3-Chloromethyl-1-phenylsulfanylmethyl-1H-isoquinoline-2-carboxylic acid methyl

ester (194)

Dibenzyl-carbamic acid phenyl ester (195)

NCl

CO2Me

PhS

NCO2Ph

To a stirred solution of 188 (0.651 g, 1.25 mmol) in CH2Cl2 (5 mL) at 25 °C was added

phenyl chloroformate (1.57 mL, 12.5 mmol). The reaction was heated to reflux and

stirred for 11 h. The reaction was cooled to 25 °C, treated with sat. aq. NaHCO3 (10 mL)

and stirred vigorously for 15 min. The reaction mixture was extracted with CH2Cl2 (3 x

20 mL). The combined extracts were dried (Na2SO4), filtered, and concentrated under

vacuum. The resulting residue was purified by flash column chromatography (SiO2, 3%-

10% EtOAc in hexanes) to provide 188 (0.29 g, 45% yield) and an inseparable mixture of

chloride 194 and carbamate 195 as a pale yellow oil (0.495 g, 58% yield): (characterized

155

without separation), Rf = 0.40 (20% EtOAc in hexanes); IR (thin film): 3061, 2953, 1717,

1455, 1439, 1344, 1204, 1068 cm-1; 1H NMR (300 MHz, C6D6): δ 7.29-6.71 (ArH), 5.71

(1 H, s), 5.47 (1 H, brs), 4.41 (1 H, s), 4.28 (1 H, s), 3.84 and 3.80 (carbamate

methylenes), 3.30-3.22 (1 H, m), 3.21 (3 H, s), 2.89 (1 H, dd, J = 13.9, 7.3 Hz) ppm; 13C

NMR (75 MHz, C6D6): δ 154.8, 152.0, 137.5, 136.6, 133.3, 129.9, 129.6, 129.3, 129.1,

129.0, 128.7, 128.4, 127.9, 126.9, 126.6, 126.5, 126.3, 126.2, 126.1, 125.9, 125.4, 125.1,

121.9, 117.1, 115.1, 55.4, 52.6, 49.9, 49.5, 45.5, 36.4 ppm.

3-methyl-1-phenylsulfanylmethyl-3,4-dihydro-1H-isoquinoline-2-carboxylic acid

methyl ester (173)

3-chloromethyl-1-phenylsulfanylmethyl-3,4-dihydro-1H-isoquinoline-2-carboxylic

acid methyl ester (198)

NCO2Me

SPh

MeH

H

NCO2Me

SPh

ClH

H

To a stirred solution of the inseparable mixture of chloride 194 and carbmate 195 (0.50 g)

in CH2Cl2 (0.50 mL) at 25 °C was added Et3SiH (1.5 mL, 9.1 mmol). The solution was

cooled to −15 °C and added trifluoroacetic acid (pre-cooled to −15 °C, 1.1 mL, 13.6

mmol). The solution was warmed to 25 °C and stirred 14 h. The solution was cooled to 0

°C and poured over ice. The aqueous solution was basified with sat. aq. NaHCO3 until

pH = 9, and extracted with CH2Cl2 (2 x 25 mL). The organic extracts were combined,

dried (Na2SO4), filtered, and concentrated under vacuum. The yellow oil was purified by

156

flash column chromatography (SiO2, 5-10% EtOAc in hexanes) to provide an inseparable

mixture of 173 and chloride 198 (0.25 g, 75% yield) as a colorless oil: Rf = 20 (20%

EtOAc in hexanes); recrystallized from toluene as 6:1 ratio of 198 to 173; X-ray (see

Appendix A); mp = 75-80 °C; 198: IR (thin film): 2953, 2925, 1698, 1441, 1387, 1324

cm-1; 1H NMR (300 MHz, CDCl3): δ 7.41-7.15 (9 H, m), 5.52 (0.4 H, brs), 5.25 (0.6 H,

brs), 4.28 (1.2 H, brs), 4.06 (0.8 H, brs), 3.85-3.65 (4 H, m), 3.42-3.36 (1 H, m), 3.25-

3.01 (3 H, m) ppm; 13C NMR (75 MHz, CDCl3): δ 153.2, 136.5, 133.8, 132.9, 129.4,

129.1, 128.3, 128.2, 127.5, 126.7, 126.5, 126.2, 121.9, 55.5, 53.8, 52.7, 48.1, 47.0, 40.1,

35.4, 30.7 ppm; HRMS calcd. for C19H21NO2SCl (MH+) 362.0982 Da. Found 362.0969

Da.

Triisopropyl-prop-2-ynyloxy-silane (200)

OTIPS

To a stirred solution of propargyl alcohol (2.08 mL, 35.7 mmol) and imidazole (2.67 g,

39.3 mmol) in DMF (16 mL) at 0 °C was added TIPS-Cl (8.83 mL, 41.0 mmol). The

reaction was warmed to 25 °C and stirred for 16 h. The reaction mixture was partitioned

between hexanes (125 mL) and sat. aq. NaHCO3 (75 mL). The organic layer was washed

with sat. aq. NaHCO3 (50 mL), sat. aq. NaCl (50 mL), dried (Na2SO4), filtered, and

concentrated under vacuum. The resulting colorless liquid was purified by flash column

chromatography (SiO2, 2-4% EtOAc-hexane) then by distillation (110 °C, 20 mmHg) to

afford 200 (3.8 g, 54% yield) as a colorless liquid: Rf = 0.35 (2% EtOAc in hexanes); IR

(neat): 3313, 2944, 2867, 1464, 1370, 1263, 1101, 1001, 883, 774 cm-1; 1H NMR (300

157

MHz, CDCl3): δ 4.38 (2 H, d, J = 2.2 Hz), 2.39 (1 H, t, J = 2.3 Hz), 1.16-1.05 (21 H, m)

ppm; 13C NMR (75 MHz, CDCl3): δ 82.2, 72.4, 51.5, 17.6, 11.7 ppm; HRMS calcd. for

C12H23OSi (M-H+) 211.1518 Da. Found 211.1515 Da.

3-Triisopropylsilanyloxymethyl-isoquinoline (201)

NOTIPS

To a stirred solution of imine 179 (1.10 g, 3.83 mmol) and alkyne 200 (0.98 g, 4.60

mmol) in THF (15.3 mL) was added PdCl2(PPh3)2 (53 mg, 0.08 mmol) and CuI (7 mg,

0.04 mmol). The reaction was flushed with argon, sealed, heated to 55 °C and stirred for

1 h. The reaction was cooled to 25 °C, filtered through Celite®, and the filtrate was

concentrated under vacuum. The resulting residue was dissolved in anhydrous DMF (38

mL), and added CuI (73 mg, 0.10 mmol). The reaction mixture was flushed with argon,

sealed, heated to 100 °C, and stirred for 4 h. The reaction was cooled to 25 °C, diluted

with Et2O (120 mL), washed with sat. aq. NH4Cl (130 mL), dried (Na2SO4), filtered, and

concentrated under vacuum. The resulting residue was purified by flash column

chromatography (SiO2, 5-10% EtOAc-hexane) to afford isoquinoline 201 (1.13 g, 94%

yield) as a yellow oil: Rf = 0.36 (20% EtOAc in hexanes); IR (thin film): 2954, 2911,

2875, 1630, 1595, 1457, 1373, 1128, 1096, 1015, 879, 816 cm-1; 1H NMR (300 MHz,

CDCl3): δ 9.15 (1 H, s), 7.94 (1 H, d, J = 7.9 Hz), 7.86 (1 H, s), 7.83 (1 H, d, J = 8.3 Hz),

7.66 (1 H, t, J = 8.1 Hz), 7.54 (1 H, t, J = 7.9 Hz) 5.09 (2 H, s), 1.29-1.20 (3 H, m), 1.12

(18 H, d, J = 6.4 Hz) ppm; 13C NMR (75 MHz, CDCl3): δ 136.7, 130.5, 127.9, 127.7,

158

126.8, 126.7, 154.8, 151.9, 115.8, 65.8, 17.7, 12.0 ppm; HRMS calcd. for C19H30NOSi

(MH+) 316.2097 Da. Found 316.2105 Da.

1-Phenylsulfanylmethyl-3-triisopropylsilanyloxymethyl-1H-isoquinoline-2-

carboxylic acid methyl ester (204)

NOTIPS

PhS

CO2Me

To a stirred solution of thioanisole (0.178 mL, 1.52 mmol) and (−)-sparteine (0.340 mL,

1.52 mmol) in anhydrous THF (1.9 mL) at 0 °C was added n-BuLi (2.40 M in hexanes,

0.633 mL, 1.52 mmol) dropwise within 10 min. The cloudy, yellow solution was allowed

to warm to 25 °C and was stirred for 1 h. The above solution of phenylthiomethyllithium

and (−)-sparteine was added dropwise within 10 min. to a degassed solution of

isoquinoline 201 (0.319 g, 1.01 mmol) in toluene (8.2 mL) at −78 °C under an

atmosphere of argon. The resulting orange solution was allowed to warm to 25 °C over

15 min. The solution was stirred for 30 min. and quenched with methylchloroformate

(0.235 mL, 3.03 mmol). The pale yellow solution was cooled to 0 °C, treated with 10 mL

of sat. aq. NaHCO3, and then extracted with EtOAc (3 x 25 mL). The organic extracts

were combined, dried (Na2SO4), filtered, and the solvent was removed under vacuum.

The crude brown oil was purified by flash column chromatography (SiO2, 1-5% EtOAc

in hexanes) to provide 204 (0.501 g, 95% yield) as a pale yellow oil: Rf = 0.35 (20%

EtOAc in hexanes); IR (thin film): 2943, 2865, 1717, 1637, 1456, 1439, 1401, 1321,

1264, 1129, 1109, 1049, 883 cm-1; 1H NMR (300 MHz, C6D6): δ 7.25 (2 H, brs), 7.09-

159

6.84 (6 H, m), 6.78 (1 H, d, J = 7.1 Hz), 6.56 (1 H, brs), 5.60 (1 H, brs), 5.20-4.80 (2 H,

brs), 3.25 (3 H, s), 3.22-3.17 (1 H, m), 2.89 (1 H, dd, J = 13.7, 6.4 Hz), 1.09 (9 H, s) ppm; 13C NMR (75 MHz, C6D6): δ 154.1, 138.8, 136.8, 133.2, 130.8, 129.1, 128.9, 128.5,

128.3, 127.1, 126.8, 126.3, 125.8, 125.2, 112.0, 63.8, 55.8, 52.3, 36.6, 18.1, 12.2 ppm;

HRMS calcd. for C28H40NO3SSi (MH+) 498.2498 Da. Found 498.2514 Da.

Carbonic acid methyl ester 1-phenylsulfanylmethyl-isoquinolin-3-ylmethyl ester

(207)

3-Hydroxymethyl-1-phenylsulfanylmethyl-3,4-dihydro-1H-isoquinoline-2-carboxylic

acid methyl ester (206)

NOCO2Me

PhS

N

PhS

CO2Me

OHH

H

To a stirred solution of 204 (0.508 g, 1.03 mmol) and triethylsilane (0.818 mL, 5.13

mmol) in CH2Cl2 (1.0 mL) at −10 °C was added TFA (pre-cooled to 0 °C, 0.70 mL, 9.0

mmol). TLC analysis showed complete deprotection of silylether but no reduction after

15 min. at −10 °C. The reaction was warmed to 25 °C, stirred for 1 h, and quenched with

sat. aq. NaHCO3 (30 mL). The solution was extracted with CH2Cl2 (3 x 25 mL). The

combined extracts were dried (Na2SO4), filtered, and concentrated under vacuum. The

resulting residue was purified by flash column chromatography (SiO2, 10-70% EtOAc in

hexanes) to provide 207 (0.103 g, 29% yield) as a yellow oil and 206 (0.187 g, 53%

yield) as a colorless oil. 207: Rf = 0.24 (20% EtOAc in hexanes); IR (thin film): 2954,

160

1750, 1699, 1439, 1287, 1268, 741 cm-1; 1H NMR (300 MHz, CDCl3): δ 8.16 (1 H, d, J =

8.0 Hz), 7.81 (1 H, d, J = 8.1 Hz), 7.67 (1 H, t, J = 8.0 Hz), 7.62 (1 H, s), 7.58 (1 H, t, J =

7.9), 7.41-7.37 (2 H, m), 7.26-7.17 (3 H, m), 5.34 (2 H, s), 4.73 (2 H, s), 3.83 (3 H, s)

ppm; m/z (C.I.) 349 (100%, MH+). 206: Rf = 0.19 (50% EtOAc in hexanes); IR (thin

film) 3446, 2923, 2850, 1684, 1445, 1393, 1329, 1264, 1073, 1026, 739, 690 cm-1; 1H

NMR (300 MHz, CDCl3) δ 7.41 (2 H, d, J = 10.2 Hz), 7.37-7.20 (7 H, m), 5.62 (0.4 H,

brs), 5.34 (0.6 H, brs), 4.21-4.05 (2 H, m), 3.66 (4 H, brs), 3.43-3.02 (4 H, m), 2.98-2.90

(1 H, m) ppm; 13C NMR (75 MHz, CDCl3) δ 157.2, 136.1, 135.1, 133.6, 129.3, 128.8,

128.1, 127.8, 126.4, 126.3, 95.1, 65.7, 55.1, 52.8, 40.4, 29.2 ppm; HRMS calcd. for

C19H22NO3S (MH+) 344.1320 Da. Found 344.1317 Da.

3-Benzyloxymethylisoquinoline (203)

NOBn

To a solution of 179 (3.0 g, 10.45 mmol) and propargyl benzyl ether4 (1.83 g, 12.54

mmol) in triethylamine (42 mL) was added PdCl2(Ph3)2 (0.147 g, 2 mol %) followed by

CuI (20 mg, 1 mol %) under an atmosphere of argon at 25 °C. The mixture was warmed

to 55 °C and stirred for 6 hours. The mixture was allowed to cool to 25 °C and filtered

through Celite® to remove the yellow precipitate which was washed with triethylamine.

The filtrate was concentrated under reduced pressure, the resulting residue was dissolved

in DMF (105 mL), and CuI (0.200 g, 10 mol %) was added under an atmosphere of argon

at 25 °C. The mixture was heated to 100 °C and stirred for 9 h. The dark brown solution

was allowed to cool to 25 °C and diluted with Et2O (500 mL), then washed with sat. aq.

161

NH4Cl (400 mL), dried (Na2SO4), and filtered. The solvent was removed under reduced

pressure to give a black oil, which was purified by flash column chromatography (SiO2,

20% to 50% EtOAc in hexanes) to afford 203 as a pale yellow oil (2.2 g, 84% yield) Rf =

0.60 (100% EtOAc); IR (thin film) 3059, 3030, 2857, 1630, 1592, 1496, 1454, 1359,

1127, 1098, 747 cm-1; 1H NMR (300 MHz, CDCl3) δ 9.24 (1 H, s), 7.95 (1 H, d, J = 8.1),

7.70-7.65 (2 H, m), 7.60-7.55 (2 H, m), 7.47-7.30 (5 H, m), 4.88 (2 H, s), 4.75 (2 H, s)

ppm; 13C NMR (75 MHz, CDCl3) δ 152.3, 151.9, 138.3, 136.5, 130.6, 128.6, 128.0,

127.9, 127.7, 127.1, 126.8, 117.6, 73.2, 73.1 ppm; HRMS calcd. for C17H16NO (MH+)

250.1232 Da. Found 250.1239 Da.

3-Benzyloxymethyl-1-phenylsulfanylmethyl-1H-isoquinoline-2-carboxylic acid

methyl ester (205)

N

PhS

CO2Me

OBn

To a stirred, degassed solution thioanisole (0.756 mL, 6.46 mmol) and (−)-sparteine (1.48

mL, 6.46 mmol) in anhydrous THF (8.05 mL) at 0 °C was added n-BuLi (1.95 M in

hexanes, 3.25 mL, 6.46 mmol) dropwise within 10 min. The cloudy, yellow solution was

allowed to warm to 25 °C and was stirred for 1 h. The solution of

phenylthiomethyllithium and (−)-sparteine was added dropwise within 10 min. to a

degassed solution of 203 (1.16 g, 4.61 mmol) in toluene (37 mL) at –78 °C under an

atmosphere of argon. The resulting orange solution was allowed to warm to 25 °C over

15 min. The solution became black in color, stirred 30 min. and quenched with

162

methylchloroformate (1.1 mL, 13.8 mmol). Upon addition the solution became pale

orange. The solution was cooled to 0 °C, treated with 30 mL of sat. aq. NaHCO3, and

then extracted with EtOAc (2 x 75 mL). The organic extracts were combined, washed

with sat. aq. NaCl (30 mL), dried (Na2SO4), filtered, and concentrated under vacuum.

The crude yellow oil was purified by flash column chromatography (SiO2, 10%-20%

EtOAc in hexanes) to provide 205 (1.50 g, 75% yield) as a colorless oil: Rf = 0.23 (20%

EtOAc in hexanes); IR (thin film) 3028, 2928, 2854, 1716, 1638, 1481, 1454, 1439,

1401, 1321, 1264, 1113, 738, 696 cm-1; 1H NMR (300 MHz, CDCl3) δ 7.41-7.11 (14 H,

m), 6.44 (1 H, s), 5.49 (1 H, brs), 4.60 (2 H, s), 4.51 (2 H, brs), 3.70 (3 H, s), 3.25 (1 H,

dd, J = 13.4, 8.1 Hz), 3.04 (1 H, dd, J = 13.7, 6.7 Hz) ppm; 13C NMR (75 MHz, CDCl3) δ

154.0, 138.0, 135.9, 134.7, 132.7, 129.9, 129.0, 128.9, 128.8, 128.2, 128.0, 127.5, 127.0,

126.1, 126.0, 125.0, 113.8, 72.7, 72.3, 69.9, 55.5, 52.9, 36.4 ppm; HRMS calcd. for

C26H26NO3S (MH+) 432.1633 Da. Found 432.1619 Da. HPLC conditions for separation

of enantiomers using ananlytical column Chiracel OD: loaded 15 μL of 0.5 mg/mL

sample in 1% isopropanol/hexanes, gradient 2%-5% over 15 min., enantiomers elute at

11.7 min. and 13.8 min. in a ratio of 1:1.

163

3-Benzyloxymethyl-1-phenylsulfanylmethyl-3,4-dihydro-1H-isoquinoline-2-

carboxylic acid methyl ester (212)

N

PhS

CO2Me

OBnH

H

To a solution of 205 (1.50 g, 3.48 mmol) and triethylsilane (5.60 mL, 34.8 mmol) in

CH2Cl2 (35.0 mL) at –20 °C was added TFA (pre-cooled to –10 °C, 2.70 mL, 34.8 mmol)

in one portion. The solution was allowed to warm to 25 °C and was stirred for 3 h at

which point starting material could not be detected by TLC. The solution was quenched

by pouring over ice, basified with 10% aq. NaOH, and extracted with CH2Cl2 (2 x 50

mL). The combined extracts were washed with sat. aq. NaCl (20 mL), dried (Na2SO4),

filtered, and the solvent was removed under vacuum. The crude oil was purified by flash

column chromatography (SiO2, 10%-20% EtOAc in hexanes) to afford 212 (1.47 g, 97%

yield) as a colorless oil: Rf = 0.18 (20% EtOAc in hexanes); IR (thin film) 3028, 2951,

2855, 1697, 1442, 1390, 1325, 1105, 1026, 738, 697 cm-1; 1H NMR (300 MHz, CDCl3) δ

7.55-7.25 (10 H, m), 7.22-7.18 (4 H, m), 5.55 (0.4 H, brs), 5.25 (0.6 H, brs), 4.57 (2 H, s),

4.38-4.18 (1 H, m), 3.86-3.62 (5 H, m), 3.37 (1 H, m), 3.22-3.13 (2 H, m), 3.01 (1 H, dd,

J = 16.2, 7.2 Hz) ppm; 13C NMR (75 MHz, CDCl3) δ 156.8, 156.0, 138.1, 136.6, 133.4,

128.7, 128.2, 127.5, 126.9, 126.0, 125.7, 73.2, 73.1, 73.0, 72.3, 71.1, 55.1, 54.7, 52.6,

52.4, 51.8, 51.5, 39.3, 39.0, 30.2, 29.6 ppm; HRMS calcd. for C26H28NO3S (MH+)

434.1790 Da. Found 434.1790 Da.

164

12-Phenylsulfanyl-11-oxa-13-aza-tricyclo[7.3.1.02,7]trideca-2,4,6-triene-13-

carboxylic acid methyl ester (213)

O

SPh

N

H

H CO2Me

To a solution of 212 (51 mg, 0.117 mmol) in chlorobenzene (0.5 mL) under an

atmosphere of argon at 0 °C was added N-chlorosuccinimide (16 mg, 0.117 mmol). The

mixture was allowed to warm to 25 °C and was stirred for 2 h, then filtered through a pad

of glass wool into a dry flask under argon and washed with chlorobenzene (0.5 mL). The

solution was cooled to 0 °C and SnCl4 (3.0 M in CH2Cl2, 8 μL, 20 mol%) was added

dropwise. The resulting yellow solution was stirred for 15 min. at 0 °C, quenched with

sat. aq. NaHCO3 (5 mL), and extracted with EtOAc (2 x 10 mL). The combined extracts

were dried (Na2SO4), filtered, and concentrated under vacuum. The crude residue was

purified by flash column chromatography (SiO2, 5%-10% EtOAc in hexanes) to afford

213 (30 mg, 76% yield) as a colorless oil: crystallized from methanol to yield a white

solid; X-ray (see Appendix B); mp = 131-133 °C; Rf = 0.19 (25% EtOAc in hexanes); IR

(thin film) 2953, 1701, 1449, 1409, 1339, 1320, 1310, 1117, 1104, 1065, 742, 692 cm-1; 1H NMR (500 MHz, toluene-d8, 100 °C) δ 8.61 (2 H, d, J = 7.2 Hz), 7.01-6.91 (4 H, m),

6.86 (1 H, t, J = 7.5 Hz), 6.82 (1 H, d, J = 7.6 Hz), 6.70 (1 H, d, J = 7.5 Hz), 5.32 (1 H,

brs), 5.23 (1 H, s), 4.56 (1 H, dd, J = 11.4, 3.1 Hz), 4.25 (1 H, brs), 3.49 (3 H, s), 3.28 (1

H, d, J = 11.4 Hz), 3.05 (1 H, dd, J = 17.3, 7.5 Hz), 2.44 (1 H, d, J = 17.3 Hz) ppm; 13C

NMR (125 MHz, toluene-d8, 100 °C) δ 155.5, 137.8, 136.4, 136.3, 131.7, 129.2, 128.9,

165

128.1, 128.0, 127.1, 126.7, 125.6, 125.4, 88.9, 65.7, 57.5, 52.5, 47.9, 32.0 ppm; HRMS

calcd. for C19H20NO3S (MH+) 342.1164 Da. Found 342.1159 Da.

Improved route to 3-Hydroxymethyl-1-phenylsulfanylmethyl-3,4-dihydro-1H-

isoquinoline-2-carboxylic acid methyl ester (206)

N

PhS

CO2Me

OHH

H

To a stirred solution of 212 (1.05 g, 2.42 mmol) in CH2Cl2 (24.0 mL) at –78 °C was

added BCl3 (1.0 M in CH2Cl2, 4.84 mL, 4.84 mmol) dropwise. Upon complete addition

the solution was allowed to warm to 0 °C and was stirred for 30 min., then treated with

methanol (3 mL), diluted with sat. aq. NaHCO3 (20 mL), and extracted with CH2Cl2 (2 x

50 mL). The organic extracts were combined, dried (Na2SO4), filtered, and the solvent

was removed under vacuum. The crude residue was purified by flash column

chromatography (SiO2, 20%-40% EtOAc in hexanes) to afford 206 (0.806 g, 95% yield)

as a colorless oil: characterized as above.

166

1-Phenylsulfanylmethyl-3-(toluene-4-sulfonyloxymethyl)-3,4-dihydro-1H-

isoquinoline-2-carboxylic acid methyl ester (222)

N

PhS

CO2Me

OTsH

H

To a stirred solution of alcohol 206 (0.215 g, 0.627 mmol) in pyridine (0.50 mL) and

CH2Cl2 (0.50 mL) at 0 °C was added p-toluenesulfonyl chloride (0.144 g, 0.752 mmol).

The reaction was warmed to 25 °C and stirred for 4 h. The solution was diluted with

EtOAc (25 mL) and washed with H2O (15 mL), 10% aq. CuSO4 (4 x 15 mL), H2O (15

mL), and sat. aq. NaCl (15 mL). The organic layer was dried (Na2SO4), filtered, and

concentrated under vacuum. The resulting residue was purified by flash column

chromatography (SiO2, 10-50% EtOAc in hexanes) to provide 222 (0.286 g, 86% yield)

as a colorless oil: Rf = 0.36 (40% EtOAc in hexanes); IR (thin film): 3030, 2953, 1698,

1443, 1390, 1363, 1328, 1189, 1177, 1096, 919 cm-1; 1H NMR (300 MHz, CDCl3): δ

7.83 (2 H, d, J = 7.9 Hz), 7.38-7.14 (11 H, m), 5.47 (0.5 H, brs), 5.18 (0.5 H, brs), 4.54

(0.5 H, brs), 4.24-4.10 (2.5 H, m), 3.57 (3 H, s), 3.28 (1 H, dd, J = 13.4, 6.7 Hz), 3.14-

2.95 (3 H, m), 2.45 (3 H, s) ppm; 13C NMR (75 MHz, CDCl3): δ 156.1, 145.1, 136.2,

136.0, 132.9, 132.7, 130.1, 129.2, 129.1, 128.2, 127.5, 126.7, 126.5, 126.2, 71.1, 70.1,

60.5, 55.5, 52.9, 51.9, 51.1, 39.3, 30.0, 29.4, 21.8, 14.4 ppm; HRMS calcd. for

C26H28NO5S2 (MH+) 498.1409 Da. Found 498.1418 Da.

167

3-Azidomethyl-1-phenylsulfanylmethyl-3,4-dihydro-1H-isoquinoline-2-carboxylic

acid methyl ester (223)

Phenylsulfanylmethyl-1,5,10,10a-tetrahydro-oxazolo[3,4-b]isoquinolin-3-one (224)

N

PhS

CO2Me

N3H

H

N

PhS

H

H

O

O

To a stirred solution of 222 (0.131 g, 0.262 mmol) in DMF (0.5 mL) at 25 °C was added

NaN3 (51 mg, 0.789 mmol). The reaction was heated to 55 °C for 12 h and then cooled to

25 °C, diluted with sat. aq NaHCO3 (5 mL) and extracted with EtOAc (3 x 15 mL). The

combined extracts were washed with sat. aq. NaCl (15 mL), dried (Na2SO4), filtered, and

concentrated under vacuum. The resulting residue was purified by flash column

chromatography (SiO2, 5-40% EtOAc in hexanes) to afford azide 223 as a colorless oil

63 mg, 65% yield) and oxazolidinone 224 (25 mg, 31% yield) as a white solid. 223: Rf =

0.18 (20% EtOAc in hexanes); IR (thin film) 2952, 2103, 1698, 1481, 1441, 1388, 1326,

1289, 1248, 739, 690 cm-1; 1H NMR (300 MHz, CDCl3) δ 7.39 (2 H, d, J = 7.6 Hz), 7.37-

7.17 (7 H, m), 5.53 (0.4 H, brs), 5.24 (0.6 H, brs), 4.21-3.95 (2 H, m), 3.85-3.56 (4 H, m),

3.41 (1 H, dd, J = 13.4, 6.8 Hz), 3.20 (1 H, dd, J = 13.4, 8.0 Hz), 3.15-2.91 (2 H, m) ppm; 13C NMR (75 MHz, CDCl3) δ 156.6, 136.3, 133.0, 129.4, 129.1, 128.2, 127.9, 127.5,

126.7, 126.5, 126.3, 55.6, 54.1, 53.9, 53.3, 53.2, 53.0, 52.7, 48.1, 39.7, 30.6 ppm; HRMS

calcd. For C19H21N4O2S (MH+) 369.1385 Da. Found 369.1380 Da. 224: Rf = 0.17 (30%

EtOAc in hexanes); mp = 130-132 °C; IR (thin film): 2901, 1748, 1404, 1346, 1280,

1227, 1063, 1025, 743 cm-1; 1H NMR (300 MHz, CDCl3): δ 7.28-7.09 (9 H, m), 5.10 (1

H, dd, J = 4.9, 2.4 Hz), 4.44 (1 H, t, J = 7.6 Hz), 4.08 (1 H, dd, J = 13.6, 4.9 Hz), 3.97-

168

3.90 (1 H, m), 3.76 (1 H, dd, J = 11.2, 8.0 Hz), 3.44 (1 H, dd, J = 13.9, 2.3 Hz), 3.10 (1

H, dd, J = 14.3, 11.3 Hz), 2.90 (1 H, dd, J = 14.4, 3.0 Hz) ppm; 13C NMR (75 MHz,

CDCl3): δ 157.0, 136.3, 134.6, 133.9, 129.2, 128.9, 127.8, 127.4, 126.1, 68.7, 54.6, 54.3,

39.7, 34.1 ppm; HRMS calcd. for C18H18NO2S (MH+) 312.1058 Da. Found 312.1065 Da.

3-Azidomethyl-1-phenylsulfanylmethyl-3,4-dihydro-1H-isoquinoline-2-carboxylic

acid methyl ester (223)

N

PhS

CO2Me

N3H

H

To a solution of 206 (0.790 g, 2.30 mmol) in THF (7.7 mL) at 0 °C was added

triphenylphosphine (1.33 g, 5.06 mmol) and diethyl azodicarboxylate (0.797 mL, 5.06

mmol). After 2 min. a white precipitate formed and diphenylphosphoryl azide (1.09 mL,

5.06 mmol) was added. The mixture was allowed to warm to 25 °C and was stirred for 2

h, then concentrated under reduced pressure. The residue was purified by flash column

chromatography (SiO2, 5% to 7% to 10% EtOAc in hexanes) to afford azide 223 (0.694

g, 82% yield) as a colorless oil: characterized as above.

169

12-Phenylsulfanyl-11,13-diaza-tricyclo[7.3.1.02,7]trideca-2,4,6,11-tetraene-13-

carboxylic acid methyl ester (226)

N

H

H CO2Me

N

SPh

To a solution of 223 (45 mg, 0.122 mmol) in chlorobenzene (0.61 mL) under an

atmosphere of argon at 0 °C was added N-chlorosuccinimide (16 mg, 0.122 mmol). The

mixture was allowed to warm to 25 °C and was stirred for 2 h, then filtered through a pad

of glass wool into a dry flask under argon and washed with chlorobenzene (0.6 mL). The

solution was cooled to 0 °C and SnCl4 (3.0 M in CH2Cl2, 41 μL, 0.122 mmol) was added

dropwise. The solution immediately became yellow, a precipitate formed, and the

evolution of gas was observed. Upon complete addition no starting material could be

detected by TLC. The mixture was diluted with sat. aq. NaHCO3 until basic (5 mL) and

extracted with CH2Cl2 (2 x 10 mL). The combined extracts were dried (Na2SO4), filtered,

and the solvent was removed under vacuum. The crude residue was purified by flash

column chromatography (SiO2, 10%-40% EtOAc in hexanes) to afford 226 as a yellow

oil (22 mg, 49% yield). Rf = 0.16 (30% EtOAc in hexanes); IR (thin film) 2922, 1705,

1637, 1449, 1320, 1121, 741, 688 cm-1; 1H NMR (300 MHz, CDCl3) δ 7.42-7.30 (5 H,

m), 7.25-7.15 (4 H, m), 5.49 (0.4 H, brs), 5.33 (0.6 H, brs), 4.79 (0.4 H, m), 4.67 (0.6 H,

m), 4.24-4.12 (1 H, m), 3.72 (3 H, s), 3.67 (1 H, d, J = 18.1 Hz), 3.41-3.33 (1 H, m), 2.71

(1 H, d, J = 17.5 Hz) ppm; 13C NMR (75 MHz, CDCl3) δ 145.6, 134.8, 133.3, 129.5,

129.3, 129.2, 128.9, 127.8, 127.4, 126.1, 105.0, 56.6, 54.6, 53.1, 43.8, 33.9 ppm; HRMS

calcd. for C19H19N2O2S (MH+) 339.1167 Da. Found 339.1163 Da.

170

12-Oxo-11,13-diaza-tricyclo[7.3.1.02,7]trideca-2,4,6-triene-13-carboxylic acid methyl

ester (229)

N

H

H CO2Me

NH

O

To a solution of 226 (22 mg, 0.060 mmol) in Et2O (1.0 mL) was added 5% aq. sodium

hydrogen sulfate (2.0 mL) with vigorous stirring. After 5 min. the layers were separated

and the aqueous layer was extracted with Et2O (2 x 3.0 mL). The organic extracts were

combined, washed with sat. aq. NaCl (1.0 mL), dried (Na2SO4), filtered, and the solvent

was removed under vacuum. The crude oil was purified by flash column chromatography

(SiO2, 100% EtOAc) to afford 229 (15 mg, >95% yield) as a colorless oil: crystallized

from EtOAc to give a white solid; X-ray (appendix C); mp = 211-213 °C; Rf = 0.17

(100% EtOAc); IR (thin film) 3266, 2955, 1701, 1685, 1489, 1452, 1411, 1323, 1251,

1119, 746 cm-1; 1H NMR (300 MHz, CDCl3) δ 7.39 (1 H, d, J = 6.0 Hz), 7.25-7.12 (3 H,

m), 6.16-5.99 (1 H, m), 5.53-5.41 (1 H, m), 5.02-4.85 (1 H, m), 3.91-3.85 (1 H, m), 3.73

(3 H, s), 3.47 (1 H, dd, J = 17.6, 8.0 Hz), 3.23 (1 H, dd, J = 11.9, 2.6 Hz), 2.81 (1 H, d, J

= 17.7 Hz) ppm; 13C NMR (75 MHz, CDCl3) δ 169.0, 154.8, 132.2, 128.7, 128.0, 126.9,

126.4, 56.9, 52.9, 47.1, 42.4, 32.6 ppm; HRMS calcd. for C13H15N2O3 (MH+) 247.1083

Da. Found 247.1082 Da.

171

2-Benzyloxy-3,5-dimethoxy-4-methyl benzaldehyde (233)

OMe

Me

MeOOBn

CHO

To a stirred solution of phenol 2315 (1.12 g, 6.67 mmol) in acetic acid (33 mL) was added

hexamethylenetetramine (5.61 g, 40.0 mmol) in one portion. The solution was heated to

100 °C and stirred 2 h. The brown/red solution was cooled to 25 °C and poured into H2O

(75 mL). The aqeous solution was extracted with CH2Cl2 (2 x 75 mL). The organic

extracts were combined, washed with 5% aq. Na2CO3 (2 x 75 mL), sat. aq. NaCl (75

mL), dried (Na2SO4), filtered, and concentrated under vacuum. The brown oil was

purified by flash column chromatography (SiO2, 15% to 25% EtOAc in hexanes) to

afford the aldehyde (0.676 g, 52% yield) as an impure yellow solid. The aldehyde (3.45

mmol) was then dissolved in DMF (35 mL) and to the stirred solution was added

tetrabutylammonium iodide (25 mg, 0.070 mmol), Na2CO3 (0.731 g, 6.90 mmol), and

benzyl bromide (0.492 mL, 4.14 mmol). The yellow solution was stirred 14 h at 25 °C.

The reaction was diluted with sat. aq. NH4Cl (60 mL) and extracted with Et2O (2 x 100

mL). The combined extracts were washed with sat. aq. NaHCO3 (75 mL), sat. aq. NaCl

(75 mL), dried (Na2SO4), filtered, and concentrated under vacuum. The residue was

purified by flash column chromatography (SiO2, 10% EtOAc in hexanes) to afford

benzaldehyde 233 (0.910 g, 48% yield over the two steps) as a colorless oil: Rf = 0.36

(20% EtOAc in hexanes); IR (thin film) 2939, 2859, 1683, 1598, 1464, 1412, 1372, 1332,

1283, 1205, 1133, 1081, 1023 cm-1; 1H NMR (300 MHz, CDCl3) δ 10.15 (1 H, s), 7.38 (5

H,s), 7.01 (1 H, s), 5.12 (2 H, s), 3.90 (3 H, s), 3.84 (3 H, s), 2.24 (3 H, s) ppm; 13C NMR

172

(75 MHz, CDCl3) δ 189.3, 154.4, 152.0, 149.3, 136.1, 129.5, 128.5, 128.4, 128.3, 127.4,

101.8, 76.7, 60.5, 55.6, 9.5 ppm; HRMS calcd. for C17H19O4 (MH+) 287.1283 Da. Found

287.1281 Da.

(2-Benzyloxy-3,5-dimethoxy-4-methyl-phenyl)-methanol (236)

OMe

MeOOBn

Me

OH

To a stirred solution of phenol 2315 (7.31g, 43.5 mmol) and paraformaldehyde (1.96 g) in

CH2Cl2 (89 mL) at 0 °C under an atmosphere of argon was added neat Et2AlCl (6.27 mL,

50.0 mmol) dropwise within 15 min. The solution was stirred for 30 min. at 0 °C and 25

°C for 21 h. The solution was quenched with aq. 2 M HCl (120 mL) and quickly

extracted with EtOAc (2 x 200 mL). The organic extracts were combined, washed with

sat. aq. NaCl (100 mL), dried (Na2SO4), filtered through Celite® to remove trace

precipitate, and concentrated under vacuum to afford 235 as an acid sensitive yellow

solid (8.6 g, 99% yield) which was shown to be free of impurities by spectrographic

analysis and used immediately without further purification.

To a solution of 235 (8.6 g, 43.4 mmol) in acetone (217 mL) at 25 °C was added

anhydrous K2CO3 (25.0 g, 178 mmol) and benzyl bromide (25.8 mL, 217 mmol). The

mixture was heated at reflux for 4 h and then cooled to 25 °C, diluted with saturate

aqueous NH4Cl (400 mL), and extracted with EtOAc (2 x 400 mL). The combined

extracts were dried (Na2SO4), filtered, and concentrated under vacuum. The resulting

173

brown oil was purified by flash column chromatography (SiO2, 20% to 50% EtOAc in

hexanes) to afford 236 (11.2 g, 89% yield) as a burnt orange oil: Rf = 0.31 (40% EtOAc

in hexanes); IR (thin film) 3416, 2936, 2873, 2837, 1606, 1586, 1484, 1455, 1410, 1375,

1328, 1223, 1129, 1089, 1026, 845, 755, 698 cm-1; 1H NMR (300 MHz, CDCl3) δ 7.46-

7.35 (5 H, m), 6.63 (1 H, s), 5.01 (2 H, s), 4.55 (2 H, brs), 3.87 (3 H, s), 3.79 (3 H, s),

2.60 (1 H, brs), 2.20 (3 H, s) ppm; 13C NMR (75 MHz, CDCl3) δ 154.2, 151.6, 143.0,

137.3, 131.8, 128.4, 128.3, 128.2, 128.0, 120.0, 105.2, 75.2, 60.8, 60.3, 55.5, 8.7 ppm;

HRMS calcd. for C17H21O4 (MH+) 289.1440 Da. Found 289.1432 Da.

(2-Benzyloxy-6-iodo-3,5-dimethoxy-4-methyl-phenyl)-methanol (237)

OMe

MeOOBn

Me

OH

I

To a stirred suspension of 236 (3.60 g, 12.5 mmol) and silver trifluoroacetate (5.52 g,

25.0 mmol) at 0 °C in CHCl3 (55 mL) was added I2 (4.76 g, 18.8 mmol) as a solution in

CHCl3 (82 mL). The mixture was stirred for 1 h at 0 °C, filtered through Celite® and

washed with CHCl3. The organic solution was washed with 10% aq. sodium thiosulfate

(50 mL) and sat. aq. NaCl (50 mL), dried (Na2SO4), filtered, and concentrated under

vacuum. The resulting yellow oil was purified by flash column chromatography (SiO2,

10% to 25% EtOAc in hexanes) to afford 237 (4.20 g, 82% yield) as a pale yellow oil: Rf

= 0.46 (20% EtOAc in hexanes); IR (thin film) 3474, 2934, 2888, 2835, 1447, 1404,

1371, 1314, 1228, 1104, 1088, 1012, 965, 736, 698 cm-1; 1H NMR (300 MHz, CDCl3) δ

174

7.50-7.36 (5 H, m), 5.04 (2 H, s), 4.78 (2 H, d, J = 5.6 Hz), 3.86 (3 H, s), 3.76 (3 H, s),

2.31 (3 H, s) ppm; 13C NMR (75 MHz, CDCl3) δ 154.5, 152.5, 147.1, 136.8, 135.2,

128.5, 128.2, 126.1, 91.8, 76.0, 64.3, 60.3, 60.2, 10.6 ppm; HRMS calcd. for C17H19O4I

(M) 414.0328 Da. Found 414.0323 Da.

2-Benzyloxy-6-iodo-3,5-dimethoxy-4-methyl benzaldehyde (234)

OMe

MeOOBn

Me

O

I

To a stirred solution of 237 (12.9 g, 31.1 mmol) at 0 °C in CH2Cl2 (78.0 mL) was added

pyridinium chlorochromate (20.1 g, 93.2 mmol) in small portions. The solution was

allowed to warm to 25 °C and was stirred for 12 h. Silica gel was added to the mixture

such that upon removal of the solvent under vacuum a fine brown powder was obtained,

which was purifed by flash column chromatography (SiO2, 10% to 25% EtOAc in

hexanes) to afford 234 (12.8 g, 98% yield) as a pale yellow oil: Rf = 0.29 (20% EtOAc in

hexanes); IR (thin film) 3031, 2935, 2870, 1699, 1445, 1392, 1362, 1304, 1255, 1238,

1105, 1017, 962, 748, 698 cm-1; 1H NMR (300 MHz, CDCl3) δ 10.05 (1 H, s), 7.49-7.35

(5 H, m), 5.05 (2 H, s), 3.87 (3 H, s), 3.78 (3 H, s), 2.36 (3 H, s) ppm; 13C NMR (75

MHz, CDCl3) δ 191.9, 155.2, 153.1, 150.6, 136.1, 131.9, 129.1, 128.5, 128.3, 87.4, 76.7,

60.5, 60.3, 11.1 ppm; HRMS calcd. for C17H17O4I (M) 412.0171 Da. Found 412.0165 Da.

175

[1-(2-Benzyloxy-6-iodo-3,5-dimethoxy-4-methyl-phenyl)-meth-(E)-ylidene]-tert-butyl

amine (238)

2-Benzyloxy-6-(3-benzyloxy-prop-1-ynyl)-3,5-dimethoxy-4-methyl benzaldehyde

(239)

8-Benzyloxy-3-benzyloxymethyl-5,7-dimethoxy-6-methyl isoquinoline (240)

OMe

Me

MeOOBn

N

I

MeOMe

MeOOBn

CHO

OBn

NOBn

OMeMe

MeOOBn

To a stirred solution of 234 (1.28 g, 3.10 mmol) in toluene (8 mL) was added 4 Å

molecular sieves and t-butyl amine (0.98 mL, 9.31 mmole) at 25 °C. The mixture was

stirred for 3 h and filtered through Celite®, washed with toluene, and the solvent

evaporated under vacuum to afford imine 238, which was used without further

purification.

To a degassed solution of 238 (1.45 g, 3.10 mmol) and progargyl benzyl ether (0.543 g,

3.72 mmol) in triethylamine (12.4 mL) at 25 °C under argon was added PdCl2(PPh3)2 (44

mg, 0.062 mmol) and CuI (6 mg, 0.031 mmol). The reaction flask was sealed under

argon, heated to 55 °C and stirred for 3 h. The solution became black in color and a

precipitate formed. The mixture was cooled to 25 °C, filtered through Celite®, and

concentrated filtrate under vacuum. The resulting residue was dissolved in anhydrous

DMF (30 mL), and to this solution was added CuI (50 mg, 0.30 mmole). The reaction

flask was flushed with argon, sealed, heated to 100 °C, and stirred for 5 h. The reaction

was cooled to 25 °C, diluted with sat. aq. NH4Cl (30 mL) and extracted with Et2O (2 x 50

mL). The combined extracts were dried (Na2SO4), filtered, and concentrated under

176

vacuum. The resulting residue was purified by flash column chromatography (SiO2,

10-40% EtOAc-hexane) to afford starting material 234 (0.524 g, 41% yield), coupled

acetylene 239 (0.240, 18% yield) and product 240 (0.440, 33% yield). 239: Rf = 0.45

(40% EtOAc in hexanes); IR (thin film): 2937, 2852, 1695, 1456, 1395, 1371, 1332,

1281, 1100, 1075, 1011 cm-1; 1H NMR (300 MHz, CDCl3): δ 10.45 (1 H, s), 7.53-7.32

(10 H, m), 5.07 (2 H, s), 4.79 (2 H, s), 4.53 (2 H, s), 3.91 (6 H, s), 2.30 (3 H, s) ppm; 13C

NMR (75 MHz, CDCl3): δ 189.3, 157.4, 152.9, 149.8, 137.4, 136.2, 132.9, 129.0, 128.6,

128.4, 128.3, 128.2, 128.0, 127.7, 112.9, 94.7, 79.5, 76.4, 71.3, 60.7, 60.6, 57.8, 10.0

ppm; HRMS calcd. for C27H27O5 (MH+) 431.1858 Da. Found 431.1846 Da. 240: Rf =

0.43 (40% EtOAc in hexanes); IR (thin film) 2936, 2857, 1589, 1454, 1394, 1359, 1317,

1079, 1004, 735, 698 cm-1; 1H NMR (300 MHz, CDCl3) δ 9.45 (1 H, s), 7.91 (1 H, s),

7.54 (2 H, d, J = 8.01 Hz), 7.46-7.31 (8 H, m), 5.18 (2 H, s), 4.84 (2 H, s), 4.73 (2 H, s),

4.01 (3 H, s), 3.90 (3 H, s), 2.43 (3 H, s) ppm; 13C NMR (75 MHz, CDCl3) δ 150.8,

149.2, 148.9, 147.0, 143.0, 138.0, 136.7, 128.8, 128.4, 128.3, 128.2, 127.7, 127.3, 126.7,

122.3, 111.6, 75.8, 73.0, 72.6, 61.4, 60.6, 10.0 ppm; HRMS calcd. for C27H28NO4 (MH+)

430.2018 Da. Found 430.2027 Da.

177

Optimized formation of 8-Benzyloxy-3-benzyloxymethyl-5,7-dimethoxy-6-methyl

isoquinoline (240)

NOBn

OMeMe

MeOOBn

To a solution of 234 (0.490 g, 1.19 mmol) in toluene (12.5 mL) was added 4 Å molecular

sieves (ca. 2 g) and t-butyl amine (0.375 mL, 3.57 mmole) at 25 °C. The mixture was

stirred for 3 h, filtered through Celite®, washed with toluene, and concentrated under

vacuum to afford imine 238, which was used without further purification.

To a degassed solution of 238 (0.58 g, 1.19 mmol) and progargyl benzyl ether4 (0.183 g,

1.25 mmol) in triethylamine (6.0 mL) at 25 °C under argon was added CuI (0.283 g, 1.49

mmol) in small portions. The mixture was stirred for 24 h and a yellow precipitate

formed. The mixture was diluted with triethylamine (8.0 mL), degassed, warmed to 85

°C, and stirred for 1 h. The mixture turned dark brown and the precipitate dissolved. The

solution was cooled to 25 °C, a black precipitate formed, and the mixture was filtered,

washed with triethylamine, and the solvent was removed under reduced pressure to give a

black oil, which was purified by flash column chromatography (SiO2, 10% to 20%

EtOAc in hexanes with 1% triethylamine) to afford 240 (0.465 g, 91% yield) as a yellow

oil: characterized as above.

178

8-Benzyloxy-3-benzyloxymethyl-5,7-dimethoxy-6-methyl-1-phenylsulfanylmethyl-

1H-isoquinoline-2-carboxylic acid methyl ester (241)

NOBn

OMeMe

MeOOBn

SPh

CO2Me

To a stirred, degassed solution of thioanisole (0.136 mL, 1.16 mmol) and (−)-sparteine

(0.266 mL, 1.16 mmol) in anhydrous THF (1.45 mL) at 0 °C was added n-BuLi (2.10 M

in hexanes, 0.551 mL, 1.16 mmol) dropwise within 10 min. The yellow solution was

allowed to warm to 25 °C and was stirred for 1 h. The solution of

phenylthiomethyllithium and (−)-sparteine was added dropwise within 10 min. to a

degassed solution of 240 (0.166 g, 0.386 mmol) in toluene (3.10 mL) at –78 °C under an

atmosphere of argon. The resulting orange solution was allowed to warm to 25 °C over

15 min. at which point the solution became black in color. To the reaction was added

methylchloroformate (0.149 mL, 1.93 mmol) at 25 °C and the solution became pale

orange. The solution was cooled to 0 °C, quenched with sat. aq. NaHCO3 (10 mL), and

then extracted with EtOAc (2 x 20 mL). The organic extracts were combined, washed

with sat. aq. NaCl (10 mL), dried (Na2SO4), filtered, and the solvent was removed under

vacuum. The crude yellow oil was purified by flash column chromatography (SiO2, 10%-

20% EtOAc in hexanes) to provide 241 (0.195 g, 83% yield) as a colorless oil: Rf = 0.26

(20% EtOAc in hexanes); IR (thin film) 2951, 2936, 2857, 1715, 1463, 1454, 1439, 1414,

1327, 1295, 1122, 1070, 1027, 1005 737, 698 cm-1; 1H NMR (300 MHz, CDCl3) δ 7.41-

7.29 (12 H, m), 7.10-7.06 (3 H, m), 6.67 (1 H, s), 5.91 (1 H, brs), 5.15 (1 H, d, J = 10.8

Hz), 4.90 (1 H, d, J = 10.8 Hz), 4.63-4.48 (4 H, m), 3.86 (3 H, s), 3.71 (3 H, s), 3.58 (3 H,

179

brs), 3.12-2.98 (2 H, m), 2.22 (3 H, s) ppm; 13C NMR (75 MHz, CDCl3) δ 150.1, 143.4,

138.1, 137.0, 136.4, 128.6, 128.4, 128.2, 128.0, 127.9, 127.5, 127.4, 125.3, 125.1, 119.7,

119.7, 75.3, 72.3, 70.0, 61.2, 60.3, 52.6, 50.1, 34.9, 9.1 ppm; HRMS calcd. for

C36H38NO6S (MH+) 612.2120 Da. Found 612.2413 Da. HPLC conditions for separation

of enantiomers using ananlytical column Chiracel OD: loaded 12 μL of 0.53 mg/mL

sample in 1% isopropanol/hexanes, gradient 2-8% over 15 min., enantiomers elute at 12.4

min. and 13.1 min. in a ratio of 1:1.

8-Benzyloxy-5,7-dimethoxy-3,6-dimethyl-1-phenylsulfanylmethyl-3,4-dihydro-1H-

isoquinoline-2-carboxylic acid methyl ester (242)

8-Benzyloxy-3-benzyloxymethyl-5,7-dimethoxy-6-methyl-1-phenylsulfanylmethyl-

3,4-dihydro-1H-isoquinoline-2-carboxylic acid methyl ester (243)

N

OMeMe

MeOOBn

SPh

CO2Me

NOBn

OMeMe

MeOOBn

SPh

CO2Me

To a stirred solution of 241 (53 mg, 0.087 mmol) and triethylsilane (0.138 mL, 0.867

mmol) in CH2Cl2 (0.50 mL) at –10 °C was added TFA (pre-cooled to –20 °C, 34 µL,

0.435 mmol) in one portion. The solution was allowed to warm to 25 °C and was stirred

20 min. The reaction was then quenched with sat. aq. NaHCO3 (5 mL) and extracted with

CH2Cl2 (2 x 10 mL). The combined extracts were dried (Na2SO4), filtered, and

concentrated under vacuum. The resulting crude oil was purified by flash column

chromatography (SiO2, 5% to 7% to 10% EtOAc in hexanes) to afford 242 (27 mg, 61%

180

yield) as a colorless oil and 243 (17 mg, 31% yield) as a colorless oil. 242: Rf = 0.44

(40% EtOAc in hexanes); IR (thin film) 2988, 2952, 2935, 1697, 1452, 1412, 1313, 1254,

1116, 1083, 1061, 738, 698 cm-1; 1H NMR (300 MHz, CDCl3) δ 7.47-7.28 (7 H, m),

7.13-7.08 (3 H, m), 6.07 (0.4 H, brs), 5.80 (0.6 H, brs), 5.07 (1 H, brs), 4.86 (1 H, brs),

4.13-4.01 (1 H, m), 3.85 (3 H, s), 3.69 (3 H, s), 3.67-3.43 (3 H, m), 3.31 (1 H, dd, J =

16.1, 7.3 Hz), 3.26-3.06 (2 H, m), 2.51 (1 H, dd, J = 16.1, 10.4 Hz), 2.23 (3 H, s), 1.47 (3

H, d, J = 6.2 Hz) ppm; 13C NMR (75 MHz, CDCl3) δ 151.6, 150.0, 144.0, 137.2, 128.8,

128.7, 128.4, 128.2, 128.1, 127.9, 127.7, 127.6, 125.5, 125.4, 124.6, 122.5, 95.1, 75.2,

60.6, 60.3, 52.2, 48.0, 28.1, 9.2 ppm; HRMS calcd. for C29H34NO5S (MH+) 508.2158 Da.

Found 508.2165 Da. 243: Rf = 0.21 (20% EtOAc in hexanes); IR (thin film), 2950, 2935,

2856, 1697, 1453, 1442, 1412, 1314, 1256, 1115, 1082, 1061, 1008, 738, 697 cm-1; 1H

NMR (300 MHz, CDCl3) δ 7.47-7.24 (12 H, m), 7.04 (3 H, brs), 6.08 (0.4 H, brs), 5.78

(0.6 H, brs), 5.12-4.98 (1 H, s), 4.91-4.78 (1 H, m), 4.66 (1 H, d, J = 12.1 Hz), 4.57 (1 H,

d, J = 12.0 Hz), 4.23-4.08 (1 H, m), 3.85 (4 H, brs), 3.77 (1 H, dd, J = 9.3, 3.0 Hz), 3.69

(3 H, s), 3.55 (3 H, brs), 3.35-3.11 (3 H, m), 2.91-2.85 (1 H, m), 2.23 (3 H, s) ppm;

HRMS calcd. for C36H40NO6S (MH+) 614.2576 Da. Found 614.2565 Da.

181

Improved Synthesis of 8-Benzyloxy-3-benzyloxymethyl-5,7-dimethoxy-6-methyl-1-

phenylsulfanylmethyl-3,4-dihydro-1H-isoquinoline-2-carboxylic acid methyl ester

(243)

NOBn

OMeMe

MeOOBn

SPh

CO2Me

To a solution of 241 (107 mg, 0.175 mmol), triethylsilane (0.280 mL, 1.75 mmol), and

benzyl alcohol (0.271 mL, 2.63 mmol) at –20 °C was added TFA (pre-cooled to –20 °C,

0.338 mL, 4.37 mmol) in one portion. The solution was allowed to warm to 0 °C and was

stirred for 16 h. The reaction was then quenched with sat. aq. NaHCO3 (10 mL) and

extracted with CH2Cl2 (2 x 10 mL). The combined extracts were dried (Na2SO4), filtered,

and concentrated under vacuum. The resulting crude oil was purified by flash column

chromatography (SiO2, 5% to 7% to 10% EtOAc in hexanes) to afford 242 (20 mg, 22%

yield) as a colorless oil and 243 (76 mg, 71% yield) as a colorless oil: characterized as

above.

182

8-Benzyloxy-3-hydroxymethyl-5,7-dimethoxy-6-methyl-1-phenylsulfanylmethyl-3,4-

dihydro-1H-isoquinoline-2-carboxylic acid methyl ester (248)

NOH

OMeMe

MeOOBn

SPh

CO2Me

To a solution of 243 (0.133 g, 0.217 mmol) in CH2Cl2 (1.0 mL) at –40 °C was added

BCl3 (1.0 M in CH2Cl2, 0.434 mL, 0.434 mmol) dropwise. Upon complete addition the

solution was stirred for 2 h and treated with methanol (0.5 mL), diluted with sat. aq.

NaHCO3 (5 mL), and extracted with CH2Cl2 (2 x 10 mL). The combined extracts were

dried (Na2SO4), filtered, and concentrated under vacuum. The crude residue was purified

by flash column chromatography (SiO2, 20%-50% EtOAc in hexanes) to afford 248

(0.100 g, 88% yield) as a colorless oil: Rf = 0.27 (40% EtOAc in hexanes); IR (thin film)

3448, 2936, 1697, 1412, 1396, 1349, 1315, 1257, 1115, 1056, 1006, 738, 697 cm-1; 1H

NMR (300 MHz, CDCl3) δ 7.52-7.28 (7 H, m), 7.12 (3 H, brs), 6.14 (0.4 H, brs), 5.86

(0.6 H, brs), 5.15-5.08 (1 H, m), 4.86-4.79 (1 H, m), 4.20 (1 H, dd, J = 11.8, 3.0 Hz),

4.06-3.98 (1 H, m), 3.86 (4 H, s), 3.75-3.70 (4 H, m), 3.68-3.46 (3 H, m), 3.27-2.82 (4 H,

m), 2.24 (3 H, s) ppm; 13C NMR (75 MHz, CDCl3) δ 157.4, 151.9, 150.0, 143.7, 137.1,

135.0, 128.8, 128.6, 128.4, 128.1, 128.0, 125.7, 125.0, 124.9, 122.6, 95.0, 75.3, 65.3,

60.7, 60.3, 55.4, 52.3, 48.5, 38.3, 22.2, 9.2 ppm; HRMS calcd. for C29H34NO6S (MH+)

524.2107 Da. Found 524.2109 Da.

183

3-Azidomethyl-8-benzyloxy-5,7-dimethoxy-6-methyl-1-phenylsulfanylmethyl-3,4-

dihydro-1H-isoquinoline-2-carboxylic acid methyl ester (250)

NN3

OMeMe

MeOOBn

SPh

CO2Me

To a solution of 248 (0.10 g, 0.191 mmol) in THF (0.640 mL) at 0 °C was added

triphenylphosphine (0.11 g, 0.420 mmol) and diisopropyl azodicarboxylate (83 μL, 0.420

mmol). After 2 min. a white precipitate formed and diphenylphosphoryl azide (91 μL,

0.42 mmol) was added. The mixture was allowed to warm to 25 °C and was stirred for

1.5 h. The mixture was treated with H2O (0.4 mL) and concentrated to a crude residue

that was purified by flash column chromatography (SiO2, 5% to 10% EtOAc in hexanes)

to afford 250 (89 mg, 85% yield) as a colorless oil: Rf = 0.46 (20% EtOAc in hexanes);

IR (thin film) 3060, 2951, 2104, 1698, 1443, 1412, 1392, 1340, 1314, 1256, 1192, 1116,

1062, 1006, 737, 698 cm-1; 1H NMR (300 MHz, CDCl3) δ 7.52-7.28 (7 H, m), 7.09 (3 H,

brs), 6.10 (0.4 H, brs), 5.82 (0.6 H, brs), 5.15-4.98 (1 H, m), 4.78-4.91 (1 H, m), 4.04 (1

H, m), 3.86 (4 H, s), 3.82-3.70 (4 H, m), 3.50 (3 H, brs), 3.38-3.09 (3 H, m), 2.76-2.67 (1

H, m), 2.24 (3 H, s) ppm; 13C NMR (75 MHz, CDCl3) δ 156.4, 151.8, 150.1, 144.0,

137.1, 136.3, 129.2, 129.1, 129.7, 128.9, 128.7, 128.5, 128.3, 128.0, 127.9, 125.2, 124.9,

121.6, 75.2, 60.8, 60.4, 54.2, 52.5, 52.2, 48.8, 38.2, 23.3, 9.3 ppm; HRMS calcd. for

C29H33N4O5S (MH+) 549.2172 Da. Found 549.2478 Da.

184

3-Benzyloxy-4,6-dimethoxy-5-methyl-12-oxo-11,13-diaza-tricyclo[7.3.1.02,7]trideca-

2(7),3,5-triene-13-carboxylic acid methyl ester (251)

N

H

H CO2Me

NH

OBnO

MeO

MeOMe

To a solution of 250 (33 mg, 0.060 mmol) in chlorobenzene (0.300 mL) at 0 °C was

added N-chlorosuccinimide (8 mg, 0.060 mmol). The mixture was allowed to warm to 25

°C and was stirred for 2 h, then filtered through a pad of glass wool into a dry flask under

argon and washed with chlorobenzene (0.700 mL). The solution was cooled to 0 °C and

SnCl4 (3.0 M in CH2Cl2, 71 μL, 0.060 mmole) was added dropwise. The solution

immediately became orange and a precipitate formed. Upon complete addition no starting

material could be detected by TLC. The mixture was diluted with sat. aq. NaHCO3 until

basic (1.0 mL) and extracted with EtOAc (2 x 10 mL). The combined extracts were dried

(Na2SO4), filtered, and the solvent was removed under vacuum. The crude residue was

purified by flash column chromatography (SiO2, 20%-80% EtOAc in hexanes) to afford

251 (13 mg, 51% yield) as a colorless oil: Rf = 0.38 (100% EtOAc); IR (thin film) 2955,

1716, 1674, 1455, 1413, 1378, 1291, 1250, 1110, 1065, 1042, 973, 699 cm-1; 1H NMR

(500 MHz, CDCl3) δ 8.18 (1 H, s), 7.47 (2 H, d, J = 7.0 Hz), 7.43-7.39 (2 H, m), 7.37-

7.34 (1 H, m), 6.60 (1 H, brs), 5.15 (1 H, 11.0 Hz), 4.97 (1 H, 11.0 Hz), 4.87-4.85 (1 H,

m), 3.83-3.80 (4 H, m), 3.70 (3 H, s), 3.66 (3 H, s), 3.26 (1 H, ddd, J = 17.3, 5.3, 1.4 Hz),

3.17 (1 H, d, J = 11.5 Hz), 2.69 (1 H, dd, J = 7.3, 1.0 Hz), 2.18 (3 H, s) ppm; 13C NMR

(125 MHz, CDCl3) δ 158.6, 153.7, 143.8, 137.25, 128.7, 128.3, 127.9, 126.3, 120.0,

185

75.64, 64.4, 60.5, 59.9, 53.1, 51.6, 30.1, 29.6, 9.46 ppm; HRMS calcd. for C23H27N2O6

(MH+) 427.1869 Da. Found 427.1867 Da.

1-Benzyloxymethyl-1H-isoquinoline-2-carboxylic acid methyl ester (261)

NCO2Me

OBn

To a stirred solution of BnOCH2SnBu36 (0.35 g, 0.85 mmol) and (−)-sparteine (0.32 mL,

1.41 mmol) in toluene (6.0 mL) at −78 °C was added n-butyllithium (2.1 M in hexanes,

0.40 mL, 0.85 mmol) dropwise within 5 min. The brown solution was stirred for 10 min.

at −78 °C, and isoquinoline (0.10 mL, 0.71 mmol) was added as a solution in THF (0.10

mL). The brown solution was allowed to warm to 25 °C and stirred for 15 min. The

reaction was quenched with ClCO2Me (0.16 mL, 2.12 mmol) and the solution became

colorless. The solution was cooled to 0 °C, diluted with sat. aq. NaHCO3 (5 mL), and

extracted with EtOAc (3 x 15 mL). The extracts were combined and washed with sat. aq.

NaCl (15 mL), dried (Na2SO4), filtered, and concentrated under vacuum. The resulting

residue was purified by flash column chromatography (SiO2, 10-20% EtOAc in hexanes)

to provide 261 (144 mg, 66% yield) as a colorless oil: Rf = 0.30 (20% EtOAc in

hexanes); IR (thin film): 3063, 3028, 2953, 2856, 1716, 1632, 1456, 1441, 1416, 1354,

1330, 1292, 1235, 1196, 1125 cm-1; 1H NMR (300 MHz, C6D6): δ 7.21-6.68 (10 H, m),

5.79 (0.6 H, t, J = 6.6 Hz), 5.54 (0.4 H, d, J = 7.8 Hz), 5.49 (0.6 H, d, J = 7.8 Hz), 5.40

(0.4 H, t, J = 6.8 Hz), 4.34 (0.6 H, d, J = 12.1 Hz), 4.19 (0.6 H, d, 12.2 Hz), 4.15 (0.4 H,

d, J = 12.4 Hz), 4.07 (0.4 H, d, J = 12.3 Hz), 3.54 (0.6 H, dd, J = 9.9, 7.5 Hz), 3.45-3.29

186

(4 H, m), 3.14 (0.4 H, dd, J = 9.7, 5.9 Hz) ppm; 13C NMR (75 MHz, C6D6): δ 153.6,

152.8, 138.4, 138.2, 130.9, 130.6, 129.8, 129.5, 127.9, 127.0, 126.9, 126.8, 126.7, 126.4,

126.0, 125.4, 124.4, 124.2, 107.8, 72.2, 70.2, 55.2, 54.3, 52.3 ppm; 1H NMR (500 MHz,

toluene-d8, 100 °C): δ 7.07-6.85 (9 H, m), 6.79 (1 H, d, J = 7.5 Hz), 5.55 (1 H, d, J = 7.7

Hz), 5.54 (1 H, brs), 4.29 (1 H, d, J = 12.2 Hz), 4.22 (1 H, d, J = 12.1 Hz), 4.30-4.20 (2

H, brs), 3.53 (1 H, dd, J = 9.9, 7.1 Hz), 3.49 (3 H, s), 3.36 (1 H, dd, J = 9.7, 6.0 Hz) ppm; 13C NMR (125 MHz, toluene-d8, 100 °C): δ 154.0, 139.3, 131.7, 130.9, 129.2, 129.0,

128.8, 128.3, 128.1, 127.9, 127.6, 126.9, 125.8, 125.5, 125.3, 108.4, 73.5, 71.1, 56.1, 52.8

ppm; HRMS calcd. for C19H20NO3 (MH+) 310.1443 Da. Found 310.1444 Da. HPLC

conditions: loaded 15 µL of 0.50 mg/mL sample solution in 1% isopropanol in hexanes,

gradient = 2% to 5% isopropanol in hexanes, enantiomers elute at 12.85 min. and 16.47

min. in a ratio of 2:1, 33% ee.

Benzyloxy-5,7-dimethoxy-6-methyl-3-triisopropylsilanyloxymethyl-isoquinoline

(262)

OMe

Me

MeOOBn

NOTIPS

To a stirred solution of o-iodoimine 238 (12.1 g, 26.0 mmol) and acetylene 200 (5.61 g,

28.6 mmol) in triethylamine (130 mL) was added CuI (6.09 g, 31.2 mmol) in small

portions over 20 min. The reaction was stirred under argon at 25 °C for 24 h, and treated

with sat. aq. NaHCO3 (200 mL). The solution was extracted with EtOAc (3 x 200 mL).

187

The combined extracts were washed with sat. aq. NaHCO3 (75 mL) until aqueous layer

was no longer blue, washed with sat. aq. NaCl (100 mL), dried (Na2SO4), filtered, and

concentrated under vacuum to a pale orange solid. The coupled product was dissolved in

dry DMF (260 mL) and degassed (argon bubbling for 3 h). CuI (0.989 g, 5.19 mmol) was

added. The reaction flask was sealed and heated to 100 °C, and the clear solution was

stirred for 2 h. The reaction was cooled to 25 °C, treated with sat. aq. NH4Cl (300 mL),

and extracted with EtOAc (3 x 300 mL). The combined extracts were washed with sat.

aq. NH4Cl (200 mL), sat. aq. NaCl (200 mL), dried (Na2SO4), filtered, and concentrated

under vacuum. The resulting brown oil was purified by flash column chromatography

(SiO2, 5-10% EtOAc-hexane) to afford isoquinoline 262 (10.8 g, 76% yield) as a yellow

oil: Rf = 0.23 (10% EtOAc in hexanes); IR (thin film): 2942, 2865, 1623, 1591, 1456,

1394, 1363, 1315, 1121, 1106, 1082, 1014, 883 cm-1; 1H NMR (300 MHz, CDCl3): δ

9.38 (1 H, s), 8.05 (1 H, s), 7.54 (2 H, d, J = 7.0 Hz), 7.41-7.37 (3 H, m), 5.18 (2 H, s),

5.10 (2 H, s), 4.0 (3 H, s), 3.90 (3 H, s), 2.43 (3 H, s), 1.29-1.20 (3 H, m), 1.15 (18 H, d, J

= 5.5 Hz) ppm; 13C NMR (75 MHz, CDCl3): δ 154.4, 149.3, 148.6, 146.6, 146.4, 143.2,

137.0, 128.8, 128.6, 128.4, 128.3, 122.2, 109.5, 75.9, 66.1, 61.3, 60.7, 18.0, 12.0, 10.1

ppm; HRMS calcd. for C29H42NO4Si (MH+) 496.2883 Da. Found 496.2887 Da.

188

8-Benzyloxy-1-benzyloxymethyl-5,7-dimethoxy-6-methyl-3-triisopropylsilanyloxy-

methyl-1H-isoquinoline-2,4-dicarboxylic acid dimethyl ester (264)

OMe

Me

MeOOBn

NOTIPS

OBn

CO2Me

CO2Me

To a stirred solution of BnOCH2SnBu36 (0.193 g, 0.47 mmol) in THF (0.71 mL) at −78

°C was added n-BuLi (1.90 M in hexanes, 83 µL, 0.157 mmol) dropwise. The solution

became yellow in color and was stirred for 15 min. at −78 °C, then a solution of

isoquinoline 262 (78 mg, 0.157 mmol) in THF (0.20 mL) was added dropwise. The

yellow solution was allowed to warm to 25 °C and stirred 15 min. The brown/orange

solution was quenched with ClCO2Me (61 µL, 0.785 mmol). The pale yellow solution

was diluted with sat. aq. NaHCO3 (5 mL) and extracted with EtOAc (2 x 10 mL). The

combined extracts were washed with sat. aq. NaCl (10 mL), dried (Na2SO4), filtered, and

concentrated under vacuum. The resulting yellow oil was purified by flash column

chromatography (SiO2, 5-20% EtOAc-hexane) to afford starting isoquinoline 262 (68 mg,

43% yield) and diacylated product 264 (37 mg, 32% yield) as a colorless oil: Rf = 0.27

(20% EtOAc in hexanes); IR (thin film): 2943, 2866, 1750, 1716, 1441, 1268, 1126, 1069

cm-1; 1H NMR (300 MHz, CDCl3): δ 7.41-7.25 (5 H, m), 7.17-7.03 (3 H, m), 6.85-6.81 (2

H, m), 5.74 (1 H, brs), 5.04-4.93 (4 H, m), 4.75 (1 H, d, J = 10.9 Hz), 4.53 (1 H, d, J =

14.7 Hz), 3.74 (6 H, s), 3.67 (3 H, s), 3.51 (3 H, brs), 2.93-2.87 (2 H, m), 2.22 (3 H, s),

1.29-1.01 (21 H, m) ppm; 13C NMR (75 MHz, CDCl3): δ 155.3, 149.6, 143.5, 137.4,

135.7, 134.2, 131.0, 128.6, 128.1, 127.6, 126.4, 124.3, 119.9, 106.7, 74.7, 66.7, 63.3,

189

61.0, 60.1, 54.5, 52.5, 52.0, 35.3, 17.9, 11.9, 9.02 ppm; HRMS calcd. for C41H56NO9Si

(MH+) 734.3724 Da. Found 734.3732 Da.

8-Benzyloxy-1-benzyloxymethyl-5,7-dimethoxy-6-methyl-3-triisopropylsilanyloxy-

methyl-1H-isoquinoline-2-carboxylic acid methyl ester (263)

8-Benzyloxy-1-butyl-5,7-dimethoxy-6-methyl-3-triisopropylsilanyloxymethyl-1H-

isoquinoline-2-carboxylic acid methyl ester (265)

OMe

Me

MeOOBn

NOTIPS

OBn

CO2Me

Me

MeOOBn

OMe

NOTIPS

CO2Me

To a stirred solution of BnOCH2SnBu36 (0.348 g, 0.847 mmol) in THF (1.4 mL) at −78

°C was added n-BuLi (2.10 M in hexanes, 0.403 mL, 0.847 mmol) dropwise. The

solution became yellow in color and was stirred for 15 min. at −78 °C, then a solution of

isoquinoline 262 (140 mg, 0.282 mmol) in THF (0.5 mL) was added dropwise. The

red/brown solution was stirred at −78 °C for 1.5 h and quenched with ClCO2Me (109 µL,

1.41 mmol). The pale yellow solution was diluted with sat. aq. NaHCO3 (10 mL) and

extracted with EtOAc (2 x 15 mL). The combined extracts were washed with sat. aq.

NaCl (10 mL), dried (Na2SO4), filtered, and concentrated under vacuum. The resulting

yellow oil was purified by flash column chromatography (SiO2, 5-10% EtOAc-hexane) to

afford n-butyl adduct 265 (57 mg, 33% yield) as a colorless oil and 1,2-

dihydroisoquinoline 263 (86 mg, 45% yield) as a colorless oil. 265: Rf = 0.26 (10%

EtOAc in hexanes); 1H NMR (300 MHz, CDCl3): δ 7.56-7.52 (2 H, m), 7.46-7.34 (3 H,

190

m), 6.68 (1 H, s), 5.70 (1 H, brs), 5.07 (1 H, d, J = 10.7 Hz), 4.97 (1 H, d, J = 10.7 Hz),

4.95 (1 H, brs), 4.57 (1 H, d, J = 14.0 Hz), 3.85 (3 H, s), 3.73 (3 H, s), 3.72 (3 H, s), 2.22

(3 H, s), 1.71-1.63 (2 H, m), 1.32-0.77 (28 H, m) ppm; 1H NMR (500 MHz, toluene-d8,

100 °C): δ 7.46-7.44 (2 H, m), 7.20-7.17 (3 H, m), 6.88 (1 H, t, J = 1.5 Hz), 5.92 (1 H,

dd, J = 9.4, 4.7 Hz), 5.05 (1 H, dd, J = 14.0, 1.3 Hz), 5.05 (1 H, d, J = 11.3 Hz), 4.93 (1

H, d, J = 11.2 Hz), 4.79 (1 H, dd, J = 13.9, 1.5 Hz), 3.64 (3 H, s), 3.55 (3 H, s), 3.46 (3 H,

s), 2.20 (3 H, s), 1.85-1.80 (1 H, m), 1.55-1.49 (1 H, m), 1.47-1.40 (2 H, m), 1.32-1.23 (2

H, m), 1.18-1.14 (21 H, m), 0.81 (3 H, t, J = 3.9 Hz) ppm; 13C NMR (125 MHz, toluene-

d8, 100 °C): δ 155.1, 152.0, 151.5, 144.4, 138.9, 138.1, 128.8, 128.7, 128.2, 124.3, 121.0,

109.1, 76.0, 65.0, 60.9, 60.2, 52.9, 52.4, 33.3, 28.4, 22.9, 18.5, 14.1, 13.1, 9.5 ppm. 263:

Rf = 0.21 (10% EtOAc in hexanes); IR (thin film): 2943, 2865, 1714, 1456, 1321, 1128,

1090, 1067 cm-1; 1H NMR (300 MHz, CDCl3): δ 7.54-7.51 (2 H, m), 7.45-7.29 (6 H, m),

7.21-7.17 (2 H, m), 6.74 (1 H, s), 6.05 (1 H, brs), 5.10 (1 H, brs), 5.09 (1 H, d, J = 10.8

Hz), 4.98 (1 H, d, J = 10.8 Hz), 4.50 (2 H, m), 4.38 (1 H, d, J = 12.2 Hz), 3.82 (3 H, s),

3.73 (3 H, s), 3.71 (3 H, s), 3.56 (1 H, t, J = 10.3 Hz), 3.27 (1 H, dd, J = 10.6, 4.3 Hz),

2.21 (3 H, s), 1.21-1.08 (21 H, m) ppm; 13C NMR (75 MHz, CDCl3): δ 150.0, 144.2,

138.6, 137.7, 128.6, 128.2, 127.4, 125.0, 121.2, 107.1, 95.1, 75.4, 73.5, 72.4, 68.4, 67.2,

63.4, 61.3, 60.5, 53.1, 51.0, 18.2, 12.2, 9.4 ppm; 1H NMR (500 MHz, toluene-d8, 100

°C): 7.46-7.44 (2 H, m), 7.25-6.96 (8 H, m), 6.91 (1 H, t, J = 1.5 Hz), 6.28 (1 H, dd, J =

8.8, 4.7 Hz), 5.20 (1 H, dd, J = 14.5, 1.5 Hz), 5.05 (1 H, d, J = 11.1 Hz), 4.95 (1 H, d, J =

11.2 Hz), 4.76 (1 H, dd, J = 14.5, 1.6 Hz), 4.42 (1 H, d, J = 12.2 Hz), 4.31 (1 H, d, J =

12.2 Hz), 3.71 (1 H, dd, J = 10.6, 8.8 Hz), 3.61 (3 H, s), 3.56 (3 H, s), 3.45 (3 H, s), 3.41

(1 H, dd, J = 10.6, 4.8 Hz), 2.20 (3 H, s), 1.16-1.14 (21 H, m) ppm; 13C NMR (125 MHz,

toluene-d8, 100 °C): 155.0, 151.9, 151.4, 145.0, 138.8, 137.9, 137.5, 129.3, 128.7, 128.4,

191

128.3, 127.8, 127.5, 122.0, 108.1, 76.0, 73.7, 73.1, 69.9, 67.9, 64.6, 61.0, 60.2, 52.3, 18.5,

13.1, 9.5 ppm; HRMS calcd. for C39H54NO7Si (MH+) 676.3591 Da. Found 676.3601 Da.

Optimized formation of 8-Benzyloxy-1-benzyloxymethyl-5,7-dimethoxy-6-methyl-3-

triisopropylsilanyloxy-methyl-1H-isoquinoline-2-carboxylic acid methyl ester (263)

OMe

Me

MeOOBn

NOTIPS

OBn

CO2Me

To a degassed (argon bubbling) and stirred solution of BnOCH2SnBu36 (9.0 g, 21.9

mmol) in THF (109 mL) at −78 °C was added n-BuLi (2.10 M in hexanes, 7.81 mL, 16.4

mmol) within 8 min. The solution became yellow in color and was stirred for 5 min, then

a degassed (argon bubbling) solution of isoquinoline 262 (2.71 g, 5.47 mmol) in THF

(5.0 mL) was added dropwise within 10 min. The reaction solution became deep

red/brown in color. The solution was stirred for 2 h at −78 °C, then quenched rapidly with

ClCO2Me (2.1 mL, 27.4 mmol). The dark color of the solution faded to pale yellow. The

reaction was diluted with sat. aq. NaHCO3 (150 mL) and extracted with EtOAc (2 x 250

mL). The combined extracts were washed with sat. aq. NaCl (100 mL), dried (Na2SO4),

filtered, and concentrated under vacuum. The resulting yellow oil was purified by flash

column chromatography (SiO2, 5-10% EtOAc-hexane) to afford 1,2-dihydroisoquinoline

263 (3.22 g, 79% yield) as a colorless oil: characterized as above.

192

8-Hydroxy-1-hydroxymethyl-5,7-dimethoxy-6-methyl-3-triisopropylsilanyloxy-

methyl-1H-isoquinoline-2-carboxylic acid methyl ester (273)

NOTIPS

CO2Me

OMeMe

MeOOH

OH

To a solution of 1,2-dihydroisoquinoline 263 (38 mg, 0.056 mmol) in EtOH (1.0 mL) was

added PtO2 (48 mg, 0.211 mmol). The solution was charged with balloon pressure of H2

and stirred for 2 h. The solution was filtered through Celite® and concentrated under

vacuum. The residue was purified by flash column chromatography (SiO2, 20-40%

EtOAc in hexanes) to provide 273 (24 mg, 87% yield) as a colorless oil: Rf = 0.27 (40%

EtOAc in hexanes); IR (thin film): 3387, 2943, 2866, 1713, 1693, 1465, 1444, 1414,

1337, 1310, 1258, 1128, 1102, 1063, 1012, 882 cm-1; 1H NMR (300 MHz, CDCl3): δ

6.51 (1 H, s), 5.80 (1 H, t, J = 7.6 Hz), 5.59 (1 H, brs), 5.07 (1 H, brs), 4.46 (1 H, d, J =

11.9 Hz), 3.78 (6 H, s), 3.68 (3 H, s), 3.63-3.50 (2 H, m), 2.21 (3 H, s), 1.21-1.09 (21 H,

m) ppm; 13C NMR (75 MHz, CDCl3): δ 162.5, 147.5, 145.0, 140.8, 135.6, 123.0, 120.4,

117.4, 110.9, 95.1, 64.2, 61.4, 60.7, 52.9, 52.4, 17.7, 11.8, 9.2 ppm; HRMS calcd. for

C25H41NO7Si (M+) 495.2652 Da. Found 495.2652 Da.

193

8-Benzyloxy-1-benzyloxymethyl-3-hydroxymethyl-5,7-dimethoxy-6-methyl-1H-

isoquinoline-2-carboxylic acid methyl ester (274)

OMe

Me

MeOOBn

NOH

OBn

CO2Me

To a stirred solution of 263 (22 mg, 0.033 mmol) in CH2Cl2 (0.33 mL) at 0 °C was added

TFA dropwise with reaction monitoring until all starting material was consumed. The

reaction was diluted with sat. aq. NaHCO3 (15 mL) and extracted with EtOAc (2 x 20

mL). The combined extracts were washed with sat. aq. NaCl (10 mL), dried (Na2SO4),

filtered, and concentrated under vacuum. The residue was purified by flash column

chromatography (SiO2, 25-40% EtOAc-hexane) to afford 274 (15 mg, 88% yield) as a

colorless oil: Rf = 0.28 (50% EtOAc in hexanes); IR (thin film): 3462, 2938, 2862, 1699,

1456, 1414, 1395, 1346, 1331, 1290, 1251, 1114, 1064, 1008 cm-1; 1H NMR (300 MHz,

CDCl3): δ 7.51-7.19 (10 H, m), 6.44 (1 H, s), 5.98 (1 H, s), 5.10 (1 H, d, J = 11.1 Hz),

4.99 (1 H, d, J = 11.1 Hz), 4.54 (2 H, m), 4.31 (2 H, m), 3.83 (3 H, s), 3.76 (3 H, s), 3.71

(3 H, s), 3.47 (1 H, t , J = 10.2 Hz), 3.20 (1 H, m), 2.21 (3 H, s) ppm; 13C NMR (75 MHz,

CDCl3): δ 154.1, 143.8, 137.2, 128.2, 128.0, 127.3, 127.2, 125.1, 120.3, 111.2, 95.1, 75.2,

72.1, 67.6, 63.8, 61.3, 60.2, 53.1, 20.6, 14.0, 9.1 ppm; HRMS calcd. for C30H33NO7

(MH+) 519.2257 Da. Found 519.2253 Da.

194

1-Benzyloxymethyl-8-hydroxy-3-hydroxymethyl-5,7-dimethoxy-6-methyl-1H-

isoquinoline-2-carboxylic acid methyl ester (275)

NOH

CO2Me

OMeMe

MeOOH

OBn

To a solution of 1,2-dihydroisoquinoline 274 (13 mg, 0.025 mmol) in EtOH (0.30 mL)

was added 5% platinum on carbon (3 mg). The solution was charged with balloon

pressure of H2 and stirred for 16 h. The solution was filtered through Celite® and

concentrated under vacuum. The residue was purified by flash column chromatography

(SiO2, 10-30% EtOAc in hexanes) to provide 275 (10 mg, 94% yield) as a colorless oil:

Rf = 0.10 (40% EtOAc in hexanes); IR (thin film): 3392, 2959, 2861, 1686, 1445, 1445,

1414, 1335, 1261, 1101, 1061, 1011 cm-1; 1H NMR (300 MHz, CDCl3): δ 7.35-7.27 (5 H,

m), 6.43 (1 H, s), 6.01 (1 H, brs), 5.74 (1 H, s), 4.63 (1 H, d, J = 12.0 Hz), 4.52 (1 H, brs),

4.50 (1 H, d, 12.0 Hz), 4.29 (1 H, dd, J = 12.8, 4.5 Hz), 3.83 (3 H, s), 3.80 (3 H, s), 3.70

(3 H, s), 3.56 (1 H, dd, J = 10.3, 8.8 Hz), 3.43 (1 H, dd, J = 10.1, 5.0 Hz), 2.22 (3 H, s)

ppm; 13C NMR (75 MHz, CDCl3): δ 155.1, 147.6, 145.6, 141.1, 137.9, 136.5, 128.4,

127.7, 127.5, 123.7, 120.4, 111.1, 72.8, 68.4, 64.1, 61.6, 60.9, 53.4, 50.6, 9.5 ppm; m/z

(C.I.) 412 (100%, MH+−H2O).

195

6-Benzyloxy-5-benzyloxymethyl-7,9-dimethoxy-8-methyl-1,5-dihydro-oxazolo[3,4-

b]isoquinolin-3-one (276)

OMe

Me

MeOOBn

N

OBn

O

O

To a stirred solution of 275 (15 mg, 0.029 mmol) in MeOH (0.33 mL) at 25 °C was

added NaBH4 (31 mg, 0.813 mmol). The reaction was stirred for 15 h at 25 °C, quenched

with H2O (0.5 mL), diluted with sat. aq. NaHCO3 (5 mL), and extracted with EtOAc (2 x

10 mL). The combined extracts were washed with sat. aq. NaCl (10 mL), dried (Na2SO4),

filtered, and concentrated under vacuum. The residue was purified by flash column

chromatography (SiO2, 10-30% EtOAc-hexane) to afford 276 (12 mg, 86% yield) as a

colorless oil: Rf = 0.23 (30% EtOAc in hexanes); IR (thin film): 2937, 2862, 1773, 1675,

1457, 1364, 1228, 1119, 1049, 1008 cm-1; 1H NMR (300 MHz, CDCl3): δ 7.45-7.35 (5 H,

m), 7.24-7.21 (3 H, m), 7.18-7.15 (2 H, m), 5.77 (1 H, t, J = 2.0 Hz), 5.57 (1 H, t, J = 3.9

Hz), 5.03-4.99 (4 H, m), 4.60 (1 H, d, J = 12.0 Hz), 4.24 (1 H, d, J = 11.9 Hz), 3.79 (3 H,

s), 3.62 (3 H, s), 3.62-3.58 (2 H, m), 2.23 (3 H, s) ppm; HRMS calcd. for C29H30NO6

(MH+) 488.2073 Da. Found 488.2051 Da.

196

Optimized formation of 6-Benzyloxy-5-benzyloxymethyl-7,9-dimethoxy-8-methyl-

1,5-dihydro-oxazolo[3,4-b]isoquinolin-3-one (276)

OMe

Me

MeOOBn

N

OBn

O

O

To a stirred solution of 1,2-dihydroisoquinoline 275 (11.2 g, 16.6 mmol) in THF (110

mL) at −10 °C was added TBAF (1.0 M in THF, 16.6 mL, 16.6 mmol) dropwise within 5

min. The yellow solution was stirred for 15 min. at −10 °C, warmed to 0 °C and stirred

30 min., warmed further to 25 °C and stirred for 10 min. The reaction was quenched with

sat. aq. NaHCO3 (200 mL) and extracted with EtOAc (3 x 200 mL). The combined

extracts were washed with sat. aq. NaCl (200 mL), dried (Na2SO4), filtered, and

concentrated under vacuum. The resulting yellow oil was purified by flash column

chromatography (SiO2, 20-30% EtOAc-hexane) to afford oxazolidinone 276 (7.3 g, 91%

yield) as a yellow oil: characterized as above.

197

6-Benzyloxy-5-benzyloxymethyl-7,9-dimethoxy-8-methyl-1,5,10,10a-tetrahydro-

oxazolo[3,4-b]isoquinolin-3-one (279)

MeO

Me

MeOO

N

OPhPh

1

11

7

5

3

9

10

15

1416

Ha Hb

O

O

HaHb

H

To a stirred solution of oxazolidinone 276 (1.09 g, 2.24 mmol) and Et3SiH (5.40 mL,

33.6 mmol) in CH2Cl2 (11 mL) at −10 °C was added TFA (pre-cooled to −10 °C, 2.60

mL, 33.6 mmol). The yellow solution was stirred for 15 min. at −10 °C to 0 °C, then

warmed to 25 °C and stirred for 3 h. The reaction was cooled to 0 °C and slowly

quenched with 10% aq. Na2CO3 until the aqueous layer was basic, and extracted with

CH2Cl2 (3 x 30 mL). The combined extracts were washed with sat. aq. NaCl (30 mL),

dried (Na2SO4), filtered, and concentrated under vacuum to a yellow oil. The yellow oil

was purified by flash column chromatography (SiO2, 20-40% EtOAc-hexane) to afford

the tetrahydroisoquinoline 279 (1.04 g, 95% yield) as a yellow oil: Rf = 0.16 (40%

EtOAc in hexanes); IR (thin film): 2936, 1751, 1456, 1411, 1117, 1030 cm-1; 1H NMR

(300 MHz, CDCl3): δ 7.36-7.28 (5 H, m), 7.19-7.16 (3 H, m), 7.00-6.96 (2 H, m), 5.01

(C1, 1 H, t, J = 2.2 Hz), 4.96 (C16, 1 H, d, J = 11.3 Hz), 4.92 (C16, 1 H, d, J = 11.3 Hz),

4.37 (C11-Hb, 1 H, t, J = 7.6 Hz), 4.40 (C15, 1 H, d, J = 12.2 Hz), 4.29 (C14, 1 H, dd, J =

9.8, 2.8 Hz), 4.22 (C15, 1 H, d, J = 12.2 Hz), 3.94 (C11-Ha, 1 H, dd, J = 10.8, 8.2 Hz),

3.78 (C7-OMe, 3 H, s), 3.77-3.71 (C3, 1 H, m), 3.64 (C5-OMe, 3 H, s), 3.52 (C14, 1 H,

dd, J = 9.8, 2.0 Hz), 3.14 (C4-Hb, 1 H, dd, J = 14.1, 3.0 Hz), 2.61 (C4-Ha, 1 H, dd, J =

14.0, 1.6 Hz), 2.24 (C6-Me, 3 H, s) ppm; 13C NMR (75 MHz, CDCl3): δ 156.5, 151.6,

198

150.5, 144.8, 138.7, 137.1, 128.5, 128.1, 128.0, 127.3, 127.1, 126.8, 124.6, 124.3, 74.4

(C16), 72.9 (C15), 70.0 (C14), 68.6 (C11), 60.7 (C5), 60.3 (C7), 53.9 (C3), 50.9 (C1),

27.7 (C4), 9.5 (C6) ppm; HRMS calcd. for C29H32NO6 (MH+) 490.2230 Da. Found

490.2216 Da.

(8-Benzyloxy-1-benzyloxymethyl-5,7-dimethoxy-6-methyl-1,2,3,4-tetrahydro-

isoquinolin-3-yl)-methanol (283)

MeO

Me

MeOO

NH

OPhPh

1

11

7

5

3

9

10

141315

Ha Hb

OH

To a stirred solution of oxazolidinone 276 (7.0 g, 14.4 mmol) and Et3SiH (34 mL, 215

mmol) in CH2Cl2 (72 mL) at −10 °C was added TFA (pre-cooled to −10 °C, 16.7 mL,

215 mmol). The yellow solution was stirred for 45 min. at −10 °C to 0 °C, then warmed

to 25 °C and stirred for 2 h. The reaction was cooled to 0 °C and slowly quenched with

10% aq. Na2CO3 until the aqueous layer was basic, and extracted with CH2Cl2 (3 x 150

mL). The combined extracts were washed with sat. aq. NaCl (100 mL), dried (Na2SO4),

filtered, and concentrated under vacuum to a yellow oil. The crude tetrahydroisoquinoline

279 was dissolved in ethylene glycol (144 mL) then KOH (21 g, 373 mmol) and

hydrazine monohydrate (3.48 mL, 72.0 mmol) were added at 25 °C. The reaction mixture

was warmed to 150 °C and became homogeneous, then stirred for 3 h. The heat was

removed and solution cooled to 25 °C. The mixture was made homogeneous by the

199

addition of CH2Cl2 and minimal amounts of MeOH, then purified by flash column

chromatography (SiO2 pre-treated with TEA, 100% CH2Cl2) to afford amino alcohol 283

(5.7 g, 86% yield) as a brown solid: recrystallized from EtOAc and hexanes; X-ray (see

Appendix D); mp = 84-89 °C; Rf = 0.19 (100% EtOAc); IR (thin film): 3333, 2935, 2864,

1455, 1410, 1336, 1113, 1060, 1026 cm-1; 1H NMR (300 MHz, CDCl3): δ 7.38-7.29 (5 H,

m), 7.27-7.19 (5 H, m), 5.02 (C15, 1 H, d, J = 11.3 Hz), 4.85 (C15, 1 H, d, J = 11.2 Hz),

4.43 (C14, 1 H, d, J = 12.1 Hz), 4.38 (C14, 1 H, d, J = 12.0 Hz), 4.35 (C1, 1 H, dd, J =

8.0, 2.6 Hz), 4.03 (C12, 1 H, dd, J = 9.3, 3.2 Hz), 3.80 (C7-OMe, 3 H, s), 3.72 (C11, 1 H,

dd, J = 10.9, 3.4 Hz), 3.67 (C5-OMe, 3 H, s), 3.61 (C12, 1 H, dd, J = 9.2, 7.4 Hz), 3.52

(C11, 1 H, dd, J = 11.0, 6.6 Hz), 2.83-2.78 (C3, C4-Hb, 2 H, m), 2.75 (NH, OH, 2 H, brs),

2.32 (C4-Ha, 1 H, dd, J = 15.9, 11.6 Hz), 2.21 (C6-Me, 3 H, s) ppm; 13C NMR (75 MHz,

CDCl3): δ 152.3 (C5), 149.9 (C7), 145.4 (C8), 138.5 (C14Ph-ipso, 137.7 (C15Ph-ipso),

128.4, 128.2, 128.0, 127.9, 127.6 (C9), 127.5, 127.4, 125.8 (C10), 123.5 (C6), 74.5

(C15), 74.1 (C12), 72.9 (C14), 65.9 (C11), 60.3 (C7-OMe), 60.2 (C5-OMe), 53.6 (C3),

53.5 (C1), 26.5 (C4), 9.3 (C6) ppm; HRMS calcd. for C28H34NO5 (MH+) 464.2437 Da.

Found 464.2423 Da.

200

tert-Butoxycarbonylamino-acetic acid 8-benzyloxy-1-benzyloxymethyl-5,7-

dimethoxy-6-methyl-1,2,3,4-tetrahydro-isoquinolin-3-ylmethyl ester (286)

MeOMe

MeOO

NH

O

O

O

HN

PhPh

1

11

7

5

3

9

10

13 14

212022

O

O16

Ha Hb

18

19

To a stirred solution of 283 (100 mg, 0.216 mmol) and Boc-Gly-OH (40 mg, 0.227

mmol) in CH2Cl2 (1.1 mL) at 0 °C was added Py-BOP® (118 mg, 0.227 mmol), and after

5 min. Hünig’s base (0.12 mL, 0.681 mmol) was added. After stirring at 0 °C for 1 h the

solution was warmed to 25 °C and stirred 3 h. The reaction was treated with H2O (5 mL)

and extracted with EtOAc (2 x 15 mL). The combined extracts were washed with 5% aq.

NaHSO4 (15 mL), sat. aq. NaHCO3 (15 mL), sat. aq. NaCl (15 mL), dried (Na2SO4),

filtered, and concentrated under vacuum. The residue was purified by flash column

chromatography (SiO2, 20-40% EtOAc-hexane) to afford 286 (110 mg, 82% yield) as a

pale yellow oil: Rf = 0.26 (30% EtOAc in hexanes); IR (thin film): 3421, 3357, 1754,

1716, 1455, 1366, 1165, 1058 cm-1; 1H NMR (500 MHz, CDCl3): δ 7.38-7.30 (5 H, m),

7.29-7.19 (5 H, m), 5.03 (C22, 1 H, d, J = 11.1 Hz), 5.00 (BocNH, 1 H, brs), 4.82 (C22, 1

H, d, J = 11.1 Hz), 4.43 (C21, 1 H, d, J = 12.3 Hz), 4.40 (C21, 1 H, d, J = 12.1), 4.38

(C1, 1 H, m), 4.30 (C11, 1 H, dd, J = 10.9, 4.4 Hz), 4.14 (C11, 1 H, dd, J = 11.0, 7.2 Hz),

4.12 (C20, 1 H, dd, J = 9.2, 3.0 Hz), 3.93 (C14, 2 H, brs), 3.79 (C7-OMe, 3 H, s), 3.67

(C5-OMe, 3 H, s), 3.54 (C20, 1 H, dd, J = 9.0, 7.4 Hz), 2.98-2.93 (C3, 1 H, m), 2.85 (C4-

Hb, dd, J = 15.4, 2.2 Hz), 2.30 (C4-Ha, dd, J = 15.1, 11.0 Hz), 2.20 (C6-Me, 3 H, s), 1.43

201

(Boc, 9 H, s) ppm; 13C NMR (125 MHz, CDCl3): δ 170.4 (C13), 155.7 (C16), 152.3 (C5),

150.2 (C7), 145.6 (C8), 138.6 (C21Ph-ipso), 137.7 (C22Ph-ipso), 128.4, 128.3, 128.0,

127.9, 127.5, 127.4, 127.2 (C9), 125.1 (C10), 123.6 (C6), 80.0 (C18), 74.6 (C22), 74.4

(C20), 73.0 (C21), 68.9 (C11), 60.4 (C7-OMe), 60.2 (C5-OMe), 53.9 (C1), 51.0 (C3),

42.4 (C14), 28.3 (C19), 27.4 (C4), 9.3 (C6-Me) ppm; HRMS calcd. for C35H45N2O8

(MH+) 621.3176 Da. Found 621.3186 Da.

tert-Butoxycarbonylamino-acetic acid 8-benzyloxy-1-benzyloxymethyl-2-(2-tert-

butoxycarbonylamino-acetyl)-5,7-dimethoxy-6-methyl-1,2,3,4-tetrahydro-

isoquinolin-3-ylmethyl ester (288)

OMeMe

MeOOBn

NNHBoc

OOBn

O

ONHBoc

To a stirred solution of amino alcohol 283 (75 mg, 0.162 mmol) and Boc-Gly-OH (85

mg, 0.485 mmol) in CH2Cl2 (0.81 mL) at 0 °C was added Py-BOP® (252 mg, 0.485

mmol), and after 5 min. Hünig’s base (0.260 mL, 1.45 mmol) was added. After 1 hour

stirring at 0 °C the solution was warmed to 25 °C and stirred 15 h. The reaction was

treated with H2O (5 mL) and extracted with EtOAc (2 x 15 mL) The combined extracts

were washed with aq. 1 N HCl (15 mL), sat. aq. NaHCO3 (15 mL), sat. aq. NaCl (15

mL), dried (Na2SO4), filtered, and concentrated under vacuum. The residue was purified

by flash column chromatography (SiO2, 20-40% EtOAc-hexane) to afford 288 (107 mg,

202

85% yield) as a pale yellow oil: Rf = 0.33 (50% EtOAc in hexanes); IR (thin film): 3419,

3362, 2977, 2936, 1716, 1652, 1498, 155, 1417, 1366, 1249, 1166, 1055 cm-1; 1H NMR

(300 MHz, CDCl3): δ 7.51-7.12 (10 H, m), 5.36 (1 H, brs), 5.23 (1 H, dd, J = 9.1, 4.1

Hz), 5.12-5.04 (3 H, m), 4.56 (1 H, brs), 4.45-4.34 (3 H, m), 4.29-4.13 (3 H, m), 3.90-

3.82 (2 H, m), 3.81 (3 H, s), 3.67 (3 H, s), 3.46 (1 H, t, J = 9.6 Hz), 3.32 (1 H, brs), 3.21

(1 H, dd, J = 16.4, 7.5 Hz), 2.61 (1 H, dd, J = 16.2, 9.7 Hz), 2.23 (3 H, s), 1.46 (9 H, s),

1.44 (9 H, s) ppm; 13C NMR (75 MHz, CDCl3): δ 169.8, 169.5, 155.5, 151.7, 149.8,

144.0, 137.3, 137.1, 128.5, 128.3, 128.2, 128.1, 128.0, 127.6, 127.5, 127.3, 125.5, 125.3,

121.6, 79.7, 79.0, 74.8, 72.8, 70.8, 65.6, 60.6, 60.2, 60.0, 50.5, 49.1, 42.7, 42.2, 28.1,

22.6, 9.3 ppm; HRMS calcd. for C42H56N3O11 (MH+) 778.3915 Da. Found 778.3924 Da.

[2-(8-Benzyloxy-1-benzyloxymethyl-3-hydroxymethyl-5,7-dimethoxy-6-methyl-3,4-

dihydro-1H-isoquinolin-2-yl)-2-oxo-ethyl]-carbamic acid tert-butyl ester (287)

OMe

Me

MeOOBn

NNHBoc

OOBn

OH

To a stirred solution of dicoupled product 288 (75 mg, 0.096 mmol) in DMSO (1.0 mL)

was added 1 N aq. NaOH (1.0 mL). The solution was stirred for 5 min., diluted with H2O

(10 mL), and extracted with EtOAc (3 x 20 mL). The combined extracts were dried

(Na2SO4), filtered, and concentrated under vacuum. The yellow residue was purified by

flash column chromatography (SiO2, 20-50% EtOAc-hexane) to afford amide 287 (36

mg, 60% yield) as a colorless oil: Rf = 0.42 (75% EtOAc in hexanes); IR (thin film):

203

3420, 2975, 2935, 1714, 1656, 1455, 1415, 1366, 1251, 1168, 1054 cm-1; 1H NMR (300

MHz, CDCl3, signals from minor, 20%, carbamate resonance isomer not reported): δ

7.53-7.17 (10 H, m), 6.38-6.34 (0.3 H, m), 5.56 (0.3 H, brs), 5.37 (0.7 H, brs), 5.24 (0.7

H, dd, J = 10.3, 4.6 Hz), 5.17-5.04 (1.7 H, m), 4.88 (0.3 H, d, J = 10.4 Hz), 4.66 (0.3 H,

d, J = 11.8 Hz), 4.42 (0.7 H, d, J = 11.6 Hz), 4.37-3.91 (5 H, m), 3.82 (3 H, s), 3.68 (3 H,

s), 3.61-3.39 (2 H, m), 3.21-2.97 (3 H, m), 2.83-2.73 (1 H, m), 2.06 (3 H, s), 1.47 (9 H, s)

ppm; 13C NMR (75 MHz, CDCl3): δ 170.0, 155.5, 151.8, 149.6, 143.8, 137.1, 136.8,

128.6, 128.4, 128.3, 128.2, 128.0, 127.8, 127.6, 125.6, 125.0, 124.8, 122.8, 79.1, 74.9,

73.0, 70.1, 64.4, 60.6, 60.0, 54.0, 53.0, 42.8, 28.2, 22.2, 9.2 ppm; 1H NMR (500 MHz,

toluene-d8, 100 °C): δ 7.40 (2 H, d, J = 7.6 Hz), 7.25 (2 H, d, J = 7.6 Hz), 7.13-7.96 (6 H,

m), 5.60 (1 H, brs), 5.43 (1 H, brs), 4.98 (1 H, d, J = 11.1 Hz), 4.91 (1 H, d, J = 11.2 Hz),

4.28-4.20 (4 H, m), 3.99 (1 H, d, J = 14.4 Hz), 3.80 (1 H, brs), 3.59 (3 H, s), 3.53-3.43 (3

H, m), 3.39 (3 H, s), 2.98 (1 H, brs), 2.66-2.61 (2 H, m), 2.19 (3 H, s), 1.41 (9 H, s) ppm;

HRMS calcd. for C35H45N2O8 (MH+) 621.3176 Da. Found 621.3170 Da.

204

Optimized Formation of [2-(8-Benzyloxy-1-benzyloxymethyl-3-hydroxymethyl-5,7-

dimethoxy-6-methyl-3,4-dihydro-1H-isoquinolin-2-yl)-2-oxo-ethyl]-carbamic acid

tert-butyl ester (287)

OMe

Me

MeOOBn

NNHBoc

OOBn

OH

To a stirred solution of amino alcohol 283 (6.6 g, 14.2 mmol) in THF (71 mL) at 0 °C

was added triethylamine (5.95 mL, 42.7 mmol) then freshly distilled TMS-Cl (3.70 mL,

29.2 mmol) dropwise within 10 min. A white precipitate formed. The reaction was

warmed to 25 °C and stirred for 3 h.

To a stirred solution of Boc-Gly-OH (2.74 g, 15.7 mmol) and triethylamine (6.0 mL, 42.7

mmol) in THF (78 mL) at −20 °C was added pivalyl chloride (1.93 mL, 15.7 mmol)

dropwise within 10 min. The reaction was stirred for 2.5 h, then cooled to −65 °C. The

above solution of disilylated 290 was added with filtration (cotton plug) within 30 min.

The reaction was stirred at −65 °C to −10 °C for 3 h, warmed to 25 °C and stirred for 10

h. The reaction was treated with aq. 1 N HCl (100 mL), stirred vigorously for 1 min. and

extracted with EtOAc (3x 150 mL). The combined extracts were washed with aq. 1 N

HCl (100 mL), H2O (150 mL), aq. sat. NaHCO3 (150 mL), sat. aq. NaCl (150 mL), dried

(Na2SO4), filtered, and concentrated under vacuum. The resulting pale yellow oil was

purified by flash column chromatography (SiO2, 30-60% EtOAc-hexane) to afford amide

287 (7.2 g, 82% yield) as a colorless oil: characterized as above.

205

1-(8-Benzyloxy-1-benzyloxymethyl-3-hydroxymethyl-5,7-dimethoxy-6-methyl-3,4-

dihydro-1H-isoquinolin-2-yl)-2,2-dimethyl-propan-1-one (292)

Me

MeOOBn

OMe

N

OBn

H

H

O

OH

The reaction was conducted as above but scaled for 277 mg of amino alcohol 283 and the

addition to mixed anhydride 289 was done at −20 °C, which afforded 287 (212 mg, 57%

yield, characterized above) and tert-butyl amide 292 (69 mg, 21% yield) as a colorless

oil: 292: Rf = 0.19 (50% EtOAc in hexanes); IR (thin film): 3434, 2935, 2860, 1625,

1455, 1412, 1366, 1115, 1080 cm-1; 1H NMR (300 MHz, CDCl3): δ 7.46-7.36 (5 H, m),

7.28-7.24 (3 H, m), 7.19-7.15 (2 H, m), 6.03 (1 H, t, J = 7.2 Hz), 5.22 (1 H, d, J = 11.1

Hz), 4.92 (1 H, d, J = 11.1 Hz), 4.43 (1 H, d, J = 11.9 Hz), 4.37 (1 H, d, J = 11.8 Hz),

4.35 (1 H, brs), 3.87 (1 H, dd, J = 11.6, 3.3 Hz), 3.79 (3 H, s), 3.69 (3 H, s), 3.67-3.57 (3

H, m), 3.49 (1 H, brs), 3.23 (1 H, dd, J = 16.2, 7.6 Hz), 2.65 (1 H, dd, J = 16.4, 9.6 Hz),

2.25 (3 H, s), 1.30 (9 H, s) ppm; 13C NMR (75 MHz, CDCl3): δ 179.4, 151.9, 149.8,

144.1, 137.8, 137.1, 128.2, 128.1, 127.6, 127.5, 126.9, 124.9, 122.6, 74.6, 73.0, 70.7,

66.7, 60.4, 60.1, 54.2, 50.2, 39.5, 28.5, 25.8, 9.2 ppm; HRMS calcd. for C33H42NO6

(MH+) 548.3012 Da. Found 548.3019 Da.

206

Benzyloxy-6-benzyloxymethyl-1-hydroxy-8,10-dimethoxy-9-methyl-4-oxo-

1,3,4,6,11,11a-hexahydro-pyrazino[1,2-b]isoquinoline-2-carboxylic acid tert-butyl

ester (293)

OMe

Me

MeOOBn

NNBoc

OOBn

OHH

H

To a stirred solution of oxalyl chloride (73 μL, 0.834 mmol) in CH2Cl2 (3.6 mL) at −78

°C was added DMSO (119 μL, 1.67 mmol). The reaction was stirred for 10 min. and

amide 287 (0.42 g, 0.642 mmol) was added as a solution in CH2Cl2 (0.5 mL) within 10

min. After 8 min. triethylamine (0.447 mL, 3.21 mmol) was added dropwise. The

reaction was stirred at −78 °C for 10 min. and the solution allowed to warm for 8 min.

(when the reaction mixture was allowed to completely warm to 25 °C and stir for more

than 15 to 30 minutes, degradation was observed). The reaction mixture was diluted with

H2O (15 mL) and extracted with CH2Cl2 (3 x 25 mL). The combined extracts were

washed with sat. aq. NaCl (15 mL), dried (Na2SO4), filtered, and concentrated under

vacuum. The crude oil was purified by flash column chromatography (SiO2, 20-40%

EtOAc-hexane) to afford hemi-aminal 293 (0.364 g, 87% yield) as a pale yellow oil and a

3:2 mixture of diastereomers: Rf = 0.23/0.18 (40% EtOAc in hexanes); IR (thin film):

3400, 2976, 2936, 2869, 1700, 1653. 1455, 1413, 1393, 1367, 1339, 1164, 1115, 1061

cm-1; 1H NMR (300 MHz, CDCl3, only listed signals for dominant isomer): δ 7.44-7.28

(5 H, m), 7.25-7.19 (3 H, m), 7.15-7.05 (2 H, m), 5.94 (1 H, t, J = 4.9 Hz), 5.59 (0.6 H,

brs), 5.47 (0.4 H, brs), 4.98 (2 H, s), 4.52-4.38 (2 H, m), 4.23-4.18 (1 H, m), 3.95 (1 H, d,

J = 6.6 Hz), 3.88 (1 H, d, J = 17.5 Hz), 3.81 (3 H, s), 3.80- 3.70 (1 H, m), 3.69 (3 H, s),

207

3.59-3.38 (2 H, m), 3.19-3.01 (2 H, m), 2.26 (3 H, s), 1.50 (9 H, s) ppm; 13C NMR (75

MHz, CDCl3): δ 171.0, 165.9, 153.8, 151.3, 150.3, 144.5, 137.6, 137.0, 128.4, 128.3,

128.2, 128.0, 127.8, 127.5, 127.3, 127.2, 126.4, 124.8, 122.9, 81.2, 75.2, 74.6, 72.6, 71.7,

64.2, 60.7, 60.1, 55.5, 47.7, 46.0, 28.1, 23.7, 9.2 ppm; HRMS calcd. for C35H43N2O8

(MH+) 619.3019 Da. Found 619.3016 Da.

7-Benzyloxy-6-benzyloxymethyl-1,8,10-trimethoxy-9-methyl-4-oxo-1,3,4,6,11,11a-

hexahydro-pyrazino[1,2-b]isoquinoline-2-carboxylic acid tert-butyl ester (297)

OMe

Me

MeOOBn

NNBoc

OOBn

OMeH

H

To a stirred solution of 293 (73 mg, 0.118 mmole) and trimethylorthoformate (0.30 mL)

in MeOH (1.2 mL) at 0 °C was added (1S)-(+)-10-camphorsulfonic acid (55 mg, 0.236

mmole). The solution was stirred 30 min at 0 °C, then quenched with sat. aq. NaHCO3

(10 mL) and extracted with EtOAc (2 x 15 mL). The combined extracts were washed

with sat. aq. NaCl (15 mL), dried (Na2SO4), filtered, and concentrated under vacuum to

afford 297 (71 mg, 95% yield) as a pale yellow oil: Rf = 0.20 (30% EtOAc in hexanes);

IR (thin film): 2971, 2936, 2862, 1704, 1653, 1455, 1393, 1367, 1328, 1165, 1118, 1069,

907 cm-1; 1H NMR (300 MHz, CDCl3): δ 7.39-7.27 (5 H, m), 7.21-7.12 (5 H, m), 5.77-

5.71 (1 H, m), 5.39 (0.6 H, s), 5.19 (0.4 H, s), 5.06 (1 H, d, J = 11.0 Hz), 5.00-4.97 (1 H,

m), 4.52-4.46 (1 H, m), 4.41 (1 H, d, J = 12.1 Hz), 4.30-4.17 (1 H, m), 3.91-3.80 (4 H,

m), 3.71-3.66 (5 H, m), 3.37-3.24 (4 H, m), 3.02-2.85 (2 H, m), 2.25 (3 H, s), 1.51 (9 H,

208

s) ppm; 13C NMR (75 MHz, CDCl3): δ 164.5, 153.9, 153.0, 151.1, 150.9, 150.2, 144.8,

138.6, 138.5, 136.9, 128.4, 128.2, 128.1, 128.0, 127.8, 127.6, 127.5, 127.1, 124.3, 124.1,

81.8, 81.1, 80.5, 75.1, 74.5, 72.5, 71.6, 69.4, 60.9, 60.2, 56.4, 55.3, 54.9, 49.2, 45.0, 44.1,

28.1, 25.0, 9.2 ppm; HRMS calcd. for C36H45N2O8 (MH+) 633.3176 Da. Found 633.3173

Da.

7-Benzyloxy-6-benzyloxymethyl-8,10-dimethoxy-9-methyl-4-oxo-1-phenylsulfanyl-

1,3,4,6,11,11a-hexahydro-pyrazino[1,2-b]isoquinoline-2-carboxylic acid tert-butyl

ester (300)

OMe

Me

MeOOBn

NNBoc

OOBn

SPhH

H

To a stirred solution of oxalyl chloride (1.32 mL, 15.1 mmol) in CH2Cl2 (66 mL) at −78

°C was added DMSO (2.14 mL, 30.2 mmol). The reaction was stirred for 10 min. and

amide 187 (7.2 g, 11.6 mmol) was added as a solution in CH2Cl2 (15 mL) within 10 min.

After 8 min. triethylamine (4.85 mL, 34.8 mmol) was added dropwise. The reaction was

stirred at −78 °C for 10 min. and the solution was allowed the solution to warm for 8 min.

(when the reaction mixture was allowed to completely warm to 25 °C and stir for more

than 15 to 30 minutes, degradation was observed). The reaction mixture was diluted with

H2O (100 mL) and extracted with CH2Cl2 (3 x 150 mL). The combined extracts were

washed with sat. aq. NaCl (100 mL), dried (Na2SO4), filtered, and concentrated under

vacuum. The product was used directly for thioaminal formation.

209

To a stirred solution of the crude product mixture of hemi-aminal 293 (11.6 mmol) in

CH2Cl2 (60 mL) at 0 °C was added thiophenol (6.0 mL, 58.0 mmol) and

p-toluenesulfonic acid monohydrate (2.65 g, 13.9 mmol). The reaction was stirred for 1.5

h at 0 °C to 5 °C, quenched with sat. aq. NaHCO3 (100 mL), and extracted with CH2Cl2

(3 x 150 mL). The combined extracts were washed with sat. aq. NaCl (150 mL), dried

(Na2SO4), filtered, and concentrated under vacuum. The resulting yellow oil was purified

by flash column chromatography (SiO2, 10-30% EtOAc-hexane) to afford thioaminal 300

(7.1 g, 86% yield over two steps) as a white solid: recrystallized from EtOAc and

hexanes; X-ray (see Appendix E); mp = 162-164 °C; Rf = 0.26 (30% EtOAc in hexanes);

IR (thin film): 2977, 2936, 2864, 1702, 1658, 1455, 1415, 1391, 1368, 1305, 1163, 1118,

cm-1; 1H NMR (300 MHz, CDCl3): δ 7.57-7.52 (2 H, m), 7.42-7.24 (8 H, m), 7.20-7.12 (5

H, m), 5.98 (0.5 H, d, J = 2.5 Hz), 5.81-5.75 (1 H, m), 5.65 (0.5 H, d, J = 2.5 Hz), 5.06-

4.96 (2 H, m), 4.53-4.40 (2 H, m), 4.20-4.05 (2 H, m), 3.84 (1.5 H, s), 3.83 (1.5 H, s),

3.82-3.77 (1 H, m), 3.72 (1.5 H, s), 3.67 (1.5 H, s), 3.70-3.55 (2 H, m), 3.14-3.06 (2 H,

m), 2.28 (1.5 H, s), 2.26 (1.5 H, s), 1.26 (4.5 H, s), 1.15 (4.5 H, s) ppm; 1H NMR (500

MHz, DMSO-d6, 130 °C, signals were still significantly broad at 90 °C): δ 7.56-7.54 (2

H, m), 7.39-7.27 (8 H, m), 7.22-7.18 (3 H, m), 7.13-7.12 (2 H, m), 5.87 (1 H, brs), 5.76

(1 H, t, J = 4.4 Hz), 5.02 (1 H, d, J = 11.4 Hz), 4.95 (1 H, d, J = 11.2 Hz), 4.41 (1 H, d, J

= 12.0 Hz), 4.36 (1 H, d, J = 12.2 Hz), 4.13 (1 H, d, J = 17.8 Hz), 3.91 (1 H, d, J = 17.8

Hz), 3.77 (3 H, s), 3.73 (1 H, d, J = 12.2 Hz), 3.69-3.63 (2 H, m), 3.65 (3 H, s), 3.15 (1 H,

dd, J = 14.8, 3.3 Hz), 2.99 (1 H, t, J = 14.5 Hz), 2.20 (3 H, s), 1.25 (9 H, s) ppm; 13C

NMR (125 MHz, DMSO-d6, 130 °C): δ 163.7, 150.7, 149.5, 143.8, 137.9, 136.8, 133.2,

128.3, 127.5, 127.3, 127.2, 127.0, 126.4, 126.3, 125.8, 123.2, 122.9, 79.9, 73.8, 71.7,

59.9, 59.3, 56.3, 48.2, 27.1, 25.4, 8.5 ppm; HRMS calcd. For C41H47N2O7S (MH+)

711.3104 Da. Found 711.3113 Da.

210

3-Allyl-7-benzyloxy-6-benzyloxymethyl-8,10-dimethoxy-9-methyl-4-oxo-1-

phenylsulfanyl-1,3,4,6,11,11a-hexahydro-pyrazino[1,2-b]isoquinoline-2-carboxylic

acid tert-butyl ester (301)

MeO

Me

MeOO

N

O

SPh

PhPh

17

5

3

9

10

1820

Ha Hb

NH

O

O

O11

1314

15

17

16

19

24

23

Hb

Ha

21

H

To a stirred solution of thioaminal 300 (30 mg, 0.042 mmol) in THF (0.33 mL) at −78 °C

was added LiHMDS (0.57 M in THF, 86 µL, 0.049 mmol) dropwise within 5 min. The

yellow solution was stirred for 15 min. at −78 °C, and allyl bromide (4.1 μL, 0.049

mmol) was added. After 20 min. the reaction was quenched with sat. aq. NH4Cl (5 mL)

and extracted with EtOAc (2 x 10 mL). The combined extracts were washed with sat. aq.

NaCl (5 mL), dried (Na2SO4), filtered, and concentrated under vacuum. The resulting

pale yellow oil was purified by flash column chromatography (SiO2, 10-30% EtOAc-

hexane) to afford product 301 (22 mg, 70% yield) as a white solid: recrystallized from

benzene and hexanes; X-ray (see Appendix F); mp = 119-122 °C; Rf = 0.38 (30% EtOAc

in hexanes); IR (thin film): 2976, 2935, 2865, 1703, 1661, 1455, 1414, 1368, 1338, 1289,

1163, 1115 cm-1; 1H NMR (300 MHz, CDCl3): δ 7.57-7.15 (15 H, m), 5.92 (1 H, t, J =

5.2 Hz), 5.73-5.54 (2 H, m), 5.06-4.92 (4 H, m), 4.53-4.36 (3 H, m), 3.85-3.77 (6 H, m),

3.71-3.52 (4 H, m), 3.26-3.10 (1 H, m), 3.05-2.85 (1 H, m), 2.66 (1 H, brs), 2.25 (3 H, s),

1.41 (4.5 H, s), 1.14 (4.5 H, s) ppm; 1H NMR (500 MHz, toluene-d8, 100 °C): δ 7.55

(PhS-ortho, 2 H, d, J = 6.7 Hz), 7.10-6.94 (13 H, m), 6.23 (C1, 1 H, t, J = 5.2 Hz), 5.79-

5.71 (C11, C16, 2 H, m), 5.07 (C20, 1 H, d, J = 11.0 Hz), 4.99 (C20, 1 H, d, J = 11.0 Hz),

211

4.98-4.95 (C17-Ha, 1 H, m), 4.83-4.80 (C17-Hb, 1 H, m), 4.59 (C13, 1 H, brs), 4.41 (C19,

1 H, d, J = 12.0 Hz), 4.34 (C19, 1 H, d, J = 12.1 Hz), 4.03 (C18, 2 H, d, J = 5.0 Hz),

3.85-3.80 (C3, 1 H, m), 3.61 (C7-OMe, 3 H, s), 3.40-3.34 (C4-Ha, 1 H, m), 3.36 (C5-

OMe, 3 H, s), 2.98 (C4-Hb, 1 H, dd, J = 14.7, 3.4 Hz), 2.80-2.73 (C15, 2 H, m), 2.23 (C6-

Me, 3 H, s), 1.24 (C24, 9 H, s) ppm; 13C NMR (125 MHz, toluene-d8, 100 °C): δ 168.2,

153.5, 152.5, 151.5, 146.1, 139.8, 138.5, 137.9, 137.5, 135.2, 134.8, 134.4 (C16), 129.3,

128.9, 128.6, 128.4, 128.0, 127.7, 127.3, 125.5, 124.8, 124.1, 118.1 (C17), 81.2 (C23),

75.8 (C20), 73.7 (C18), 73.4 (C19), 69.5 (C11), 60.5 (C5-OMe), 60.1 (C7-OMe), 59.1

(C13), 56.9 (C3), 50.3 (C1), 38.9 (C15), 28.4 (C24), 27.0 (C4), 9.6 (C6-Me) ppm; HRMS

calcd. for C44H51N2O7S (MH+) 751.3417 Da. Found 751.3410 Da.

3-Allyl-7-benzyloxy-6-benzyloxymethyl-8,10-dimethoxy-9-methyl-4-oxo-1-

phenylsulfanyl-1,3,4,6,11,11a-hexahydro-pyrazino[1,2-b]isoquinoline-2-carboxylic

acid tert-butyl ester (302)

MeO

Me

MeOO

N

O

SPh

PhPh

17

5

3

9

10

1820

Ha Hb

NH

O

O

O11

1314

15

17

16

19

24

23

Hb

Ha

21

H

To a solution of 301 (75 mg, 0.10 mmol) in THF (1.2 mL) at −78 °C was added t-BuLi

(0.9 M in pentane, 0.28 mL, 0.25 mmol). Upon addition of t-BuLi the solution became

orange in color. Within 10 seconds of complete t-BuLi addition the reaction was

quenched with a solution of butylated hydroxyltoluene (0.110 g, 0.50 mmol) in THF (0.5

212

mL) and the solution became pale yellow in color. The solution was diluted with H2O (10

mL) and extracted with EtOAc (3 x 10 mL). The extracts were combined, washed with

sat. aq. NaCl (15 mL), dried (Na2SO4), filtered, and concentrated under vacuum. The

residue was purified by flash column chromatography (SiO2, 10-20% EtOAc-hexane) to

yield 302 (0.145 g, 73% yield) as a colorless oil: Rf = 0.41 (30% EtOAc in hexanes); IR

(thin film): 2977, 2935, 2863, 1696, 1653, 1455, 1413, 1382, 1316, 1162, 1117 cm-1; 1H

NMR (300 MHz, CDCl3): δ 7.47-7.45 (2 H, m), 7.35-7.23 (8 H, m), 7.22-7.15 (5 H, m),

6.17 (0.5 H, s), 6.08-5.94 (0.5 H, m), 5.93-5.73 (2 H, m), 5.01-4.93 (3 H, m), 4.91-4.79 (1

H, m), 4.56-4.51 (0.5 H, m), 4.47 (2 H, s), 4.44-4.40 (0.5 H, m), 3.81 (3 H, s), 3.76 (1.0

H, d, J = 5.2 Hz), 3.68 (1.0 H, d, J = 3.9 Hz), 3.65-3.45 (4 H, m), 3.20-2.91 (3 H, m),

2.80-2.61 (1 H, m), 2.22 (3 H, s), 1.40 (6 H, s), 1.27 (3 H, s) ppm; 1H NMR (500 MHz,

toluene-d8, 100 °C): δ 7.52 (PhS-ortho, 2 H, d, J = 7.7 Hz), 7.09-6.95 (13 H, m), 6.26

(C16, 1 H, brs), 6.12 (C1, 1 H, t, J = 4.8 Hz), 6.08 (C11, 1 H, brs), 5.08 (C17-Ha, 1 H, d,

J = 16.5 Hz), 5.03 (C20, 2 H, s), 4.99 (C17-Hb, 1 H, d, J = 9.4 Hz), 4.50 (C13, 1 H, brs),

4.40 (C19, 1 H, d, J = 12.0 Hz), 4.30 (C19, 1 H, d, J = 12.1 Hz), 3.98 (C18, 1 H, dd, J =

9.9, 5.3 Hz), 3.83 (C18, 1 H, dd, J = 9.8, 4.0 Hz), 3.66 (C7-OMe, 3 H, s), 3.36 (C5-OMe,

3 H, s), 3.35-3.29 (C15, C3, C4-Ha, 3 H, m), 2.97-2.87 (C15, C4-Hb, 2 H, m), 2.25 (C6-

Me, 3 H, s), 1.32 (C24, 9 H, s) ppm; 13C NMR (125 MHz, toluene-d8, 100 °C): δ 167.8,

153.6, 152.4, 151.5, 145.7, 139.7, 138.3, 137.9, 137.5, 135.3, 132.7 (C16), 129.3, 128.9,

128.6, 128.4, 128.0, 127.7, 127.4, 125.5, 124.8, 124.7, 116.1 (C17), 81.3 (C23), 75.5

(C20), 73.9 (C18), 73.6 (C19), 60.6 (C5-OMe), 60.2 (C7-OMe), 58.6 (C3), 57.8 (C13),

50.6 (C1), 41.2 (C15), 28.5 (C24), 28.2 (C4), 9.6 (C6-Me) ppm; HRMS calcd. for

C44H51N2O7S (MH+) 751.3417 Da. Found 751.3419 Da.

213

(3-Bromo-propoxy)-triisopropyl-silane (304)

Br OTIPS

To a stirred solution of 3-bromopropanol (2.6 mL, 28.8 mmol), triethylamine (8.0 mL,

57.7 mmol), and dimethylaminopyridine (0.70 g, 5.76 mmol) in CH2Cl2 (144 mL) at 0 °C

was added TIPS-Cl (7.3 mL, 34.5 mmol) dropwise. The reaction was warmed to 25 °C

and stirred for 16 h, then treated with aq. 1 N HCl (100 mL), and extracted with CH2Cl2

(3 x 50 mL). The combined extracts were washed with washed with sat. aq. NaCl (50

mL), dried (Na2SO4), filtered, and concentrated under vacuum. The resulting pale yellow

oil was purified by flash column chromatography (SiO2, 1-2% EtOAc-hexane) then by

distillation (96 °C, 1 mmHg) to afford the TIPS ether 304 as a colorless liquid (4.1 g,

48% yield): Rf = 0.34 (2% EtOAc in hexanes); IR (neat): 2943, 1463, 1382, 1261, 1148,

1106, 1062, 952, 882, 733, 681 cm-1; 1H NMR (300 MHz, CDCl3): δ 3.82 (2 H, t, J = 5.5

Hz), 3.56 (2 H, t, J = 6.5 Hz), 2.06 (2 H, quintet, J = 6.1 Hz), 1.09-1.03 (21 H, m) ppm; 13C NMR (75 MHz, CDCl3): δ 60.5, 35.6, 30.6, 17.8, 11.7 ppm; HRMS calcd. for

C12H28BrOSi (MH+) 297.1072 Da. Found 297.1075 Da.

(3-Iodo-propoxy)-triisopropyl-silane (306)

I OTIPS

To a stirred solution of TIPS ether 304 (4.1 g, 13.9 mmol) in acetone (35 mL) was added

NaI (20.9 g, 139.4 mmol). The reaction was heated at reflux for 3 h and cooled to 25 °C

214

then stirred for another 12 h. The solution was diluted with H2O (75 mL) and extracted

with Et2O (3 x 100 mL). The combined extracts were washed with sat. aq. NaCl (75 mL),

dried (Na2SO4), filtered, and concentrated under vacuum to afford pure iodide 3067 (4.0

g, 84% yield) as a pale yellow oil: Rf = 0.40 (2% EtOAc in hexanes); IR (neat): 2938,

1462, 1385, 1261, 1150, 1108, 955, 884, 736, 678 cm-1; 1H NMR (300 MHz, CDCl3): δ

3.75 (2 H, t, J = 5.4 Hz), 3.32 (2 H, t, J = 6.6 Hz), 2.01 (2 H, quintet, J = 6.2 Hz), 1.10-

1.03 (21 H, m) ppm; 13C NMR (75 MHz, CDCl3): δ 62.4, 36.3, 17.8, 11.7 ppm; HRMS

calcd. for C12H28IOSi (MH+) 342.0876 Da. Found 342.0892 Da.

7-Benzyloxy-6-benzyloxymethyl-8,10-dimethoxy-9-methyl-4-oxo-1-phenylsulfanyl-3-

(3-triisopropylsilanyloxy-propyl)-1,3,4,6,11,11a-hexahydro-pyrazino[1,2-

b]isoquinoline-2-carboxylic acid tert-butyl ester (305)

OMe

Me

MeOOBn

NNBoc

OOBn

SPhH

HOTIPS

To a stirred solution of thioaminal 300 (52 mg, 0.075 mmol) in THF (0.40 mL) at −78 °C

was added KHMDS (0.5 M in toluene, 180 µL, 0.090 mmol) dropwise within 5 min. The

yellow, clear solution was stirred for 5 min. and bromide 304 (20 µL, 0.090 mmol) was

added dropwise. The reaction was allowed to warm to 25 °C and stirred 30 min. The

reaction was quenched with sat. aq. NaHCO3 (5 mL) and extracted with EtOAc (3 x 10

mL). The combined extracts were washed with sat. aq. NaCl (10 mL), dried (Na2SO4),

filtered, and concentrated under vacuum. The resulting residue was purified by flash

215

column chromatography (SiO2, 5-10% EtOAc-hexane) to afford alkylated product 305

(33 mg, 48% yield) as a colorless oil and starting material 300 (16 mg, 30% yield). 305:

Rf = 0.34 (20% EtOAc in hexanes); IR (thin film): 2940, 2865, 1701, 1663, 1457, 1414,

1367, 1337, 1304, 1164, 1106 cm-1; 1H NMR (300 MHz, CDCl3): δ 7.60-7.15 (15 H, m),

5.98 (1 H, t, J = 5.3 Hz), 5.71 (1 H, brs), 5.03 (2 H, s), 4.51 (2.5 H, brs), 4.30 (0.5 H, brs),

3.95-3.84 (2 H, m), 3.82 (3 H, s), 3.64-3.54 (6 H, m), 3.23-3.14 (1.5 H, m), 3.02-2.90 (0.5

H, m), 2.27 (3 H, s), 2.05-1.95 (1 H, m), 1.80-1.48 (3 H, m), 1.42 (6 H, brs), 1.16 (3 H,

brs), 1.06-0.99 (21 H, m) ppm; 13C NMR (75 MHz, CDCl3): δ 168.3, 151.1, 150.2, 145.3,

138.3, 137.4, 134.6, 133.1, 128.9, 128.8, 128.4, 128.2, 128.1, 128.0, 127.8, 127.7, 127.3,

127.2, 126.9, 124.6, 122.8, 81.1, 74.7, 72.4, 62.9, 60.7, 60.0, 55.7, 48.4, 28.1, 27.9, 17.8,

17.5, 13.5, 11.7, 9.2 ppm; 1H NMR (500 MHz, DMSO-d6, 90 °C): δ 7.52 (2 H, d, J = 7.6

Hz), 7.43 (2 H, d, J = 7.2 Hz), 7.39-7.27 (6 H, m), 7.19-7.16 (3 H, m), 7.11-7.09 (2 H,

m), 5.83 (1 H, t, J = 5.4 Hz), 5.79 (1 H, s), 4.99 (1 H, d, J = 10.9 Hz), 4.91 (1 H, d, J =

10.9 Hz), 4.42 (1 H, d, J = 12.3 Hz), 4.39 (1 H, d, J = 12.1 Hz), 4.14 (1 H, dd, J = 7.5, 4.2

Hz), 3.95 (1 H, ddd, J = 11.9, 4.0, 1.6 Hz), 3.78-3.74 (1 H, m), 3.76 (3 H, s), 3.64 (1 H,

dd, J = 9.4, 6.9 Hz), 3.61-3.59 (2 H, m), 3.60 (3 H, s), 3.19 (1 H, brs), 3.05-2.99 (1 H, m),

2.18 (3 H, s), 1.90-1.78 (2 H, m), 1.61-1.54 (1 H, m), 1.48-1.39 (1 H, m), 1.27 (9 H, s),

1.01-0.91 (21 H, m) ppm; HRMS calcd. for C53H73N2O8SiS (MH+) 925.4857 Da. Found

925.4833 Da.

216

Optimized formation of 7-Benzyloxy-6-benzyloxymethyl-8,10-dimethoxy-9-methyl-

4-oxo-1-phenylsulfanyl-3-(3-triisopropylsilanyloxy-propyl)-1,3,4,6,11,11a-

hexahydro-pyrazino[1,2-b]isoquinoline-2-carboxylic acid tert-butyl ester (305)

OMe

Me

MeOOBn

NNBoc

OOBn

SPhH

HOTIPS

To a stirred solution of thioaminal 300 (3.04 g, 4.28 mmol) in THF (21 mL) at −78 °C

was added KHMDS (0.5 M in toluene, 10.3 mL, 5.14 mmol) dropwise within 5 min. The

pale orange, clear solution was stirred for 5 min. and iodide 306 (1.9 g, 5.56 mmol) was

added dropwise. No color change was observed. The reaction appeared complete by TLC

analysis after 10 min. The reaction was quenched with water (10 mL), diluted with sat.

aq. NH4Cl (75 mL), and extracted with EtOAc (3 x 150 mL). The combined extracts were

washed with sat. aq. NaCl (100 mL), dried (Na2SO4), filtered, and concentrated under

vacuum. The resulting pale yellow oil was purified by flash column chromatography

(SiO2, 5-10% EtOAc-hexane) to afford alkylated product 305 (3.10 g, 78% yield) as a

colorless oil and starting material 300 (0.31 g, 10% yield).

217

7-Benzyloxy-6-benzyloxymethyl-8,10-dimethoxy-9-methyl-4-oxo-1-phenylsulfanyl-3-

(3-triisopropylsilanyloxy-propyl)-1,3,4,6,11,11a-hexahydro-pyrazino[1,2-

b]isoquinoline-2-carboxylic acid tert-butyl ester (307)

OMe

Me

MeOOBn

NNBoc

OOBn

SPhH

HOTIPS

To a stirred, degassed solution of 305 (0.20 g, 0.22 mmol) in THF (2.2 mL) at −78 °C

was added t-BuLi (1.2 M in pentane, 0.40 mL, 0.48 mmol). Upon addition of t-BuLi the

solution became brown/orange in color. Within 10 seconds of complete t-BuLi addition

the reaction was quenched with a solution of butylated hydroxyltoluene (0.19 g, 0.86

mmol) in THF (0.5 mL) and the solution became pale yellow in color. The solution was

diluted with H2O (15 mL) and extracted with EtOAc (3 x 15 mL). The extracts were

combined, washed with sat. aq. NaCl (15 mL), dried (Na2SO4), filtered, and concentrated

under vacuum. The residue was purified by flash column chromatography (SiO2, very

slow gradient of 5-10-20% EtOAc-hexane) to yield 307 (0.145 g, 73% yield) as a

colorless oil: Rf = 0.38 (20% EtOAc in hexanes); IR (thin film): 2941, 2865, 1698, 1653,

1456, 1414, 1384, 1367, 1310, 1163, 1116 cm-1; 1H NMR (300 MHz, CDCl3): δ 7.52-

7.47 (2 H, m), 7.37-7.19 (13 H, m), 6.17 (0.5 H, s), 5.85-5.79 (1.5 H, m), 5.06-4.96 (2 H,

m), 4.50-4.45 (2.5 H, m), 4.34-4.30 (0.5 H, m), 3.83 (3 H, s), 3.79-3.48 (8 H, m), 3.20-

2.97 (2 H, m), 2.38-2.10 (2 H, m), 2.25 (3 H, s), 2.05-1.84 (2 H, m), 1.43 (6 H, brs), 1.27

(3 H, brs), 1.07-0.94 (21 H, m) ppm; 13C NMR (75 MHz, CDCl3): δ 168.3, 153.3, 151.0,

150.2, 144.6, 138.5, 137.1, 133.3, 131.2, 128.8, 128.7, 128.5, 128.3, 128.1, 127.8, 127.7,

127.2, 126.9, 126.8, 124.4, 123.8, 81.3, 74.4, 72.7, 72.3, 63.3, 62.2, 60.8, 60.2, 57.7, 56.7,

218

49.4, 33.1, 31.8, 31.4, 27.9, 27.7, 26.8, 17.9, 11.8, 9.2 ppm; 1H NMR (500 MHz, DMSO-

d6, 120 °C): δ 7.53-7.51 (2 H, m), 7.38-7.27 (8 H, m), 7.23-7.18 (3 H, m), 7.16-7.14 (2 H,

m), 6.01 (1 H, s), 5.78 (1 H, t, J = 5.2 Hz), 4.99 (1 H, d, J = 11.1 Hz), 4.95 (1 H, d, J =

11.0 Hz), 4.42 (1 H, d, J = 12.2 Hz), 4.37 (1 H, d, J = 12.3 Hz), 4.27 (1 H, t, J = 5.4 Hz),

3.77 (3 H, s), 3.69-3.59 (5 H, m), 3.61 (3 H, s), 3.03 (2 H, d, J = 7.5 Hz), 2.28-2.22 (1 H,

m), 2.19 (3 H, s), 1.98-1.88 (2 H, m), 1.67-1.62 (1 H, m), 1.34 (9 H, s), 1.11-1.01 (21 H,

m) ppm; 13C NMR (125 MHz, DMSO-d6, 120 °C): δ 167.2, 150.7, 149.5, 143.8, 138.0,

136.8, 131.3, 128.3, 127.6, 127.4, 127.2, 127.1, 126.6, 126.5, 126.4, 125.9, 123.2, 123.1,

80.1, 73.8, 72.0, 71.8, 62.7, 60.0, 59.4, 56.8, 48.3, 32.0, 30.7, 27.2, 26.1, 17.2, 17.1, 11.1,

8.6 ppm; HRMS calcd. for C53H73N2O8SiS (MH+) 925.4857 Da. Found 925.4824 Da.

7-Benzyloxy-6-benzyloxymethyl-3-(3-hydroxy-propyl)-8,10-dimethoxy-9-methyl-4-

oxo-1-phenylsulfanyl-1,3,4,6,11,11a-hexahydro-pyrazino[1,2-b]isoquinoline-2-

carboxylic acid tert-butyl ester (308)

OMe

Me

MeOOBn

NNBoc

OOBn

SPhH

HOH

To a stirred, degassed solution of 305 (1.37 g, 1.48 mmol) in THF (15 mL) at −78 °C was

added t-BuLi (1.2 M in pentane, 2.71 mL, 3.26 mmol) within 30 sec. After 10 seconds

the orange solution was quenched with a solution of butylated hydroxyltoluene (1.31 g,

5.92 mmole) in THF (3.0 mL). The pale yellow solution was diluted with H2O (75 mL)

and extracted with EtOAc (3 x 100 mL). The combined extracts were washed with sat.

219

aq. NaCl (75 mL), dried (Na2SO4), filtered, and concentrated under vacuum. The residue

was dissolved in CH3CN (15 mL), cooled to 0 °C, and 48% aq. HF (1.5 mL) added

dropwise. The solution became dark orange in color. After stirring for 30 min. at 0 °C the

reaction was quenched with sat. aq. NaHCO3 (100 mL) and extracted with EtOAc (3 x

100 mL). The combined extracts were washed with sat. aq. NaCl (75 mL), dried

(Na2SO4), filtered, and concentrated under vacuum. The resulting yellow residue was

purified by flash column chromatography (SiO2, 30-50% EtOAc-hexane) to afford

alcohol 308 (0.829 g, 83% yield) as a colorless oil: Rf = 0.28 (50% EtOAc in hexanes);

IR (thin film): 3472, 2936, 2866, 1696, 1652, 1454, 1414, 1388, 1368, 1336, 1262, 1162,

1117, 1059 cm-1; 1H NMR (300 MHz, CDCl3): δ 7.53-7.42 (2 H, m), 7.33-7.17 (13 H,

m), 6.19 (0.5 H, s), 5.76-5.73 (1.5 H, s), 5.05-4.98 (2 H, m), 4.49 (2 H, s), 4.43-4.35 (1 H,

s), 3.84 (3 H, s), 3.81-3.45 (8 H, m), 3.25-2.97 (2 H, m), 2.61-2.57 (0.5 H, m), 2.41-2.37

(0.5 H, m), 2.25 (3 H, s), 1.95-1.85 (2.5 H, m), 1.64-1.61 (0.5 H, m), 1.43 (3 H, brs), 1.24

(6 H, brs) ppm; 13C NMR (75 MHz, CDCl3): δ 168.9, 152.3, 150.8, 150.3, 144.6, 144.5,

138.4, 136.8, 133.7, 132.4, 130.9, 129.6, 128.9, 128.7, 128.5, 128.1, 127.8, 127.7, 127.6,

128.2, 126.8, 126.7, 124.4, 123.4, 81.5, 74.5, 72.6, 72.1, 65.6, 62.1, 61.0, 60.7, 60.2, 57.5,

56.2, 54.1, 49.6, 33.3, 32.6, 31.4, 30.6, 28.1, 27.7, 26.8, 26.5, 9.2 ppm; 1H NMR (500

MHz, DMSO-d6, 120 °C): δ 7.54-7.53 (2 H, m), 7.39-7.27 (8 H, m), 7.27-7.18 (3 H, m),

7.16-7.14 (2 H, m), 6.01 (1 H, s), 5.78 (1 H, t, J = 5.2 Hz), 5.01 (1 H, d, J = 11.2 Hz),

4.95 (1 H, d, J = 11.1 Hz), 4.42 (1 H, d, J = 12.3 Hz), 4.38 (1 H, d, J = 12.3 Hz), 4.27 (1

H, t, J = 6.2 Hz), 3.90 (1 H, brs), 3.77 (3 H, s), 3.67-3.60 (3 H, m), 3.61 (3 H, s), 3.43-

3.40 (2 H, m), 3.04-3.00 (2 H, m), 2.28-2.20 (1 H, m), 2.19 (3 H, s), 1.92-1.75 (2 H, m),

1.59-1.54 (1 H, m), 1.35 (9 H, s) ppm; HRMS calcd. for C44H53N2O8S (MH+) 769.3523

Da. Found 769.3525 Da.

220

7-Benzyloxy-6-benzyloxymethyl-8,10-dimethoxy-9-methyl-4-oxo-3-(3-oxo-propyl)-1-

phenylsulfanyl-1,3,4,6,11,11a-hexahydro-pyrazino[1,2-b]isoquinoline-2-carboxylic

acid tert-butyl ester (309)

OMe

Me

MeOOBn

NNBoc

OOBn

SPhH

H O

To a solution of oxalyl chloride (0.139 mL, 1.60 mmol) in CH2Cl2 (7.0 mL) at −78 °C

was added DMSO (0.227 mL, 3.20 mmol). The reaction was stirred 10 min. and added

alcohol 308 (0.95 g, 1.23 mmol) as a solution in CH2Cl2 (1.5 mL) within 5 min. The

solution became dark orange and a precipitate formed. After 10 min. triethylamine (0.514

mL, 3.69 mmol) was added dropwise. The reaction was stirred at −78 °C for 5 min. and

allowed to warm for 9 min. The reaction was quenched with H2O (25 mL) and extracted

with CH2Cl2 (3 x 40 mL). The combined extracts were washed with sat. aq. NaCl (50

mL), dried (Na2SO4), filtered, and concentrated under vacuum. A small portion of

product was purified, for the purposes of characterization, by flash column

chromatography (SiO2, 20-30% EtOAc-hexane) to afford 309 as a colorless oil: Rf = 0.19

(30% EtOAc in hexanes); IR (thin film): 2976, 2937, 2863, 2721, 1722, 1694, 1651,

1454, 1415, 1385, 1368, 1336, 1291, 1162, 1117, 1078 cm-1; 1H NMR (300 MHz, CDCl3,

pronounced carbamate resonance led to two aldehyde signals for CHO): δ 9.74 (0.5 H, s),

9.65 (0.5 H, s), 7.50-7.42 (2 H, m), 7.34-7.10 (13 H, m), 6.21 (0.5 H, s), 5.82-5.75 (1.5 H,

m), 5.01 (2 H, s), 4.48 (2 H, s), 4.40-4.25 (1 H, m), 3.83 (3 H, s), 3.78-3.45 (6 H, m),

3.18-2.98 (2 H, m), 2.96-2.64 (2 H, m), 2.60-2.21 (2 H, m), 2.26 (3 H, s), 1.28 (4 H, s),

1.09 (5 H, s) ppm; 13C NMR (75 MHz, CDCl3): δ 201.9, 201.4, 167.8, 167.5, 153.1,

221

152.3, 150.8, 150.3, 144.6, 138.4, 137.0, 136.8, 133.1, 132.6, 132.5, 131.1, 129.6, 128.9,

128.5, 128.3, 128.2, 128.1, 128.0, 127.8, 127.7, 127.6, 127.2, 127.0, 126.6, 126.2, 124.4,

123.6, 81.8, 81.4, 74.4, 72.2, 65.0, 61.7, 61.0, 60.8, 60.2, 57.5, 55.1, 53.2, 49.5, 42.3,

28.0, 27.8, 26.6, 14.0, 9.2 ppm; 1H NMR (500 MHz, DMSO-d6, 100 °C): δ 9.64 (1 H, s),

7.53 (2 H, d, J = 7.0 Hz), 7.38-7.28 (8 H, m), 7.23-7.19 (3 H, m), 7.16-7.14 (2 H, m),

6.04 (1 H, s), 5.76 (1 H, t, J = 5.1 Hz), 5.00 (1 H, d, J = 11.1 Hz), 4.95 (1 H, d, J = 11.0

Hz), 4.43 (1 H, d, J = 12.2 Hz), 4.38 (1 H, d, J = 12.2 Hz), 4.29 (1 H, t, J = 6.8 Hz), 3.77

(3 H, s), 3.70-3.59 (3 H, m), 3.63 (3 H, s), 3.12-3.08 (1 H, m), 3.05-3.01 (1 H, m), 2.72-

2.66 (1 H, m), 2.55-2.40 (2 H, m), 2.19 (3 H, s), 2.14-2.08 (1 H, m), 1.32 (9 H, s) ppm;

13C NMR (125 MHz, DMSO-d6, 100 °C): δ 201.6, 166.9, 150.7, 150.0, 143.9, 138.0,

136.8, 132.5, 131.6, 128.5, 127.7, 127.4, 127.2, 126.9, 126.6, 126.5, 125.9, 123.4, 123.1,

80.5, 73.8, 71.9, 71.8, 60.1, 59.5, 56.7, 54.9, 48.4, 41.0, 27.4, 27.3, 25.9, 8.7 ppm; HRMS

calcd. for C44H51N2O8S (MH+) 767.3366 Da. Found 767.3366 Da.

222

7-Benzyloxy-6-benzyloxymethyl-8,10-dimethoxy-9-methyl-4-oxo-1-phenylsulfanyl-3-

((E)-3-triisopropylsilanyloxy-allyl)-1,3,4,6,11,11a-hexahydro-pyrazino[1,2-

b]isoquinoline-2-carboxylic acid tert-butyl ester (310)

OMe

Me

MeOOBn

NNBoc

OOBn

SPhH

HOTIPS

The crude aldehyde product 309 was dissolved in Et2O (12.3 mL) and triethylamine

(0.343 mL, 2.46 mmol), cooled to 0 °C, and freshly distilled TIPSOTf (0.495 mL, 1.85

mmol) added dropwise. The solution was warmed to 25 °C and stirred for 16 h. The

reaction was quenched with sat. aq. NaHCO3 (50 mL) and extracted with EtOAc (3 x 75

mL). The combined extracts were washed with sat. aq. NaCl (75 mL), dried (Na2SO4),

filtered, and concentrated under vacuum. The resulting yellow residue was purified by

flash column chromatography (SiO2, 5-10-20% EtOAc-hexane) to afford silyl ether 310

(1.01 g, 88% yield) as a colorless foam: Rf = 0.33 (20% EtOAc in hexanes); IR (thin

film): 2943, 2866, 1695, 1653, 1456, 1414, 1384, 1330, 1166, 1118 cm-1; 1H NMR (300

MHz, CDCl3): δ 7.51-7.48 (2 H, m), 7.39-7.16 (13 H, m), 6.39-6.11 (2 H, m), 5.85-5.76

(1 H, m), 5.06-4.84 (3 H, m), 4.50 (2 H, s), 4.46-4.42 (1 H, m), 3.83 (3 H, s), 3.77-3.48 (6

H, m), 3.21-2.82 (3 H, m), 2.76-2.58 (1 H, m), 2.23 (3 H, s), 1.46 (6 H, s), 1.39-1.26 (3

H), 1.05-0.85 (21 H, m) ppm; 13C NMR (75 MHz, CDCl3): δ 167.6, 153.4, 151.1, 150.3,

144.5, 141.6, 138.5, 137.0, 134.0, 132.6, 131.9, 130.5, 128.9, 128.7, 128.5, 128.3, 128.1,

127.8, 127.6, 127.2, 126.9, 126.5, 124.3, 123.8, 107.9, 81.3, 74.5, 72.7, 72.3, 62.2, 60.7,

60.1, 58.1, 57.5, 49.5, 33.5, 27.9, 26.8, 17.6, 11.7, 9.2 ppm; HRMS calcd. for

C53H71N2O8SiS (MH+) 923.4700 Da. Found 923.4702 Da.

223

aldehyde 311

OMeMe

MeOOBn

N

OOBn

N

HO

BocH

H

To a stirred solution of silyl ether 310 (23 mg, 0.025 mmol) in THF (1.0 mL) at 15 °C

was added AgBF4 as a solution in THF (0.3 mL from 44 mg AgBF4 in 1.0 mL THF). A

precipitate immediately formed. After 15 min. at 15 °C the reaction was warmed to 25 °C

and stirred for another 4 h. The reaction was quenched with sat. aq. NaHCO3 (2 mL) and

extracted with EtOAc (3 x 5 mL). The combined extracts were washed with sat. aq. NaCl

(5 mL), dried (Na2SO4), and concentrated under vacuum. The residue could be purified

by flash column chromatography (SiO2, 20-60% EtOAc-hexane) to afford aldehyde 311

(13 mg, 78% yield) as a colorless oil: Rf = 0.46 (60% EtOAc in hexanes); IR (thin film):

2975, 2936, 2868, 1726, 1700, 1663, 1456, 1412, 1368, 1317, 1161, 1114 cm-1; 1H NMR

(300 MHz, CDCl3): δ 9.56 (1 H, s), 7.38-7.27 (5 H, m), 7.22-7.19 (3 H, m), 7.05-7.02 (2

H, m), 5.43 (1 H, s), 5.02-4.95 (2 H, m), 4.63 (1 H, brs), 4.57 (1 H, brs), 4.29 (1 H, d, J =

11.8 Hz), 4.24 (1 H, d, J = 11.7 Hz), 4.15-4.09 (1 H, m), 3.81 (3 H, s), 3.69 (3 H, s), 3.66-

3.57 (1 H, m), 3.34 (1 H, dd, J = 9.4, 1.9 Hz), 3.08 (1 H, t, J = 7.9 Hz), 2.92 (1 H, dd, J =

14.3, 2.5 Hz), 2.71 (1 H, t, J = 12.9 Hz), 2.42-2.32 (1 H, m), 2.27 (3 H, s), 2.07-1.99 (1

H, m), 1.43 (9 H, s) ppm; 13C NMR (75 MHz, CDCl3): δ 198.9, 167.9, 150.9, 150.3,

144.2, 138.1, 136.9, 128.4, 128.2, 128.1, 127.8, 127.4, 126.4, 124.6, 124.3, 81.0, 74.4,

72.9, 71.4, 60.7, 60.1, 57.5, 56.8, 49.9, 48.5, 32.2, 28.0, 25.2, 9.3 ppm; HRMS calcd. for

C38H45N2O8 (MH+) 657.3176 Da. Found 657.3201 Da.

224

oxime 314

OMeMe

MeOOBn

N

OOBn

N

HN

BocH

H

HO

To a solution of hydroxylamine hydrochloride (12 mg, 0.165 mmol) in H2O (0.10 mL) at

0 °C was added KOAc (40 mg, 0.411 mmol) then aldehyde 311 (90 mg, 0.137 mmol) as

a solution in EtOH (1.0 mL). The reaction was warmed to 25 °C and stirred for 2 h. The

reaction mixture was extracted with Et2O (3 x 15 mL). The combined extracts were

washed with sat. aq. NaCl (15 mL), dried (Na2SO4), filtered, and concentrated under

vacuum. The resulting residue was purified by flash column chromatography (SiO2, 20-

40% EtOAc-hexane) to afford two fractions, each with predominantly one isomer: the

trans-oxime (50 mg, 45% yield) with Rf = 0.32 (50% EtOAc in hexanes) as a 1:4 mixture

of cis:trans oxime and cis-oxime (44 mg, 38% yield) as a white solid in 4:1 mixture of

cis:trans oxime: recrystallized from benzene and hexanes; X-ray (see Appendix G); mp =

168-169 °C; Rf = 0.30 (50% EtOAc in hexanes); IR (thin film): 3334, 2930, 2857, 1702,

1666, 1455, 1412, 1368, 1318, 1290, 1162, 1114 cm-1; 1H NMR (300 MHz, CDCl3): δ

7.37-7.31 (5 H, m), 7.27-7.18 (3 H, m), 7.06-7.01 (2 H, m), 6.78 (1 H, brs), 5.45 (1 H,

brs), 5.00-4.94 (2 H, m), 4.57-4.44 (2 H, m), 4.34-4.26 (2 H, m), 4.08-4.03 (1 H, m), 3.80

(3 H, s), 3.67 (3 H, s), 3.57 (1 H, brs), 3.38-3.25 (2 H, m), 2.94-2.78 (2 H, m), 2.38-2.25

(1 H, m), 2.26 (3 H, s), 2.19-1.95 (1 H, m), 1.46 (9 H, s) ppm; 13C NMR (75 MHz,

CDCl3): δ 168.0, 152.2, 150.9, 150.1, 144.3, 138.2, 128.2, 128.0, 127.9, 127.8, 127.3,

225

127.1, 125.3, 125.0, 124.2, 124.1, 80.9, 74.4, 72.7, 71.2, 60.7, 60.2, 49.9, 29.5, 28.1, 25.4,

9.2 ppm; HRMS calcd. for C38H46N3O8 (MH+) 672.3285 Da. Found 672.3298 Da.

diol 315

OMeMe

MeOOH

N

OOH

N

HO

BocH

H

To a stirred solution of silyl ether 310 (0.22 g, 0.238 mmol) in THF (7.0 mL) at 15 °C

was added AgBF4 as a solution in THF (1.7 mL from 1.0 g AgBF4 in 20 mL THF). A

precipitate immediately formed. After 15 min. at 15 °C the reaction was warmed to 25 °C

and stirred for another 4 h. The reaction was quenched with sat. aq. NaHCO3 (7 mL) and

extracted with EtOAc (3 x 15 mL). The combined extracts were washed with sat. aq.

NaCl (15 mL), dried (Na2SO4), filtered through a plug of silica gel, and concentrated

under vacuum. The resulting residue was carried to hydrogenolysis without further

purification. To a stirred solution of crude aldehyde 311 (156 mg, 0.238 mmol) in MeOH

(5 mL) at 25 °C was added Pd(OH)2 (moist, Pd content 20%, 64 mg) and the reaction

stirred under an atmosphere of H2 (balloon pressure) for 4 h. The reaction mixture was

filtered through a plug of Celite®, washed with MeOH, and concentrated under vacuum.

The residue was purified by flash column chromatography (SiO2, 50-100% EtOAc-

hexane, then 5% MeOH in EtOAc) to afford diol 315 (64 mg, 79% yield from

cyclization) as a colorless foam: Rf = 0.20 (100% EtOAc in hexanes); IR (thin film):

226

3384, 2937, 1700, 1684, 1653, 1472, 1417, 1368, 1162, 1115, 1055 cm-1; 1H NMR (300

MHz, CDCl3): δ 9.74 (1 H, s), 5.93 (1 H, brs), 5.63 (1 H, t, J = 4.3 Hz), 4.74 (1 H, s),

4.67 (1 H, brs), 3.97 (1 H, dd, J = 11.1, 3.6 Hz), 3.78 (3 H, s), 3.68 (3 H, s), 3.66-3.60 (1

H, m), 3.33 (1 H, t, J = 8.0 Hz), 3.07 (1 H, dd, J = 14.7, 2.2 Hz), 2.61 (1 H, dd, J = 14.4,

6.8 Hz), 2.57-2.47 (1 H, m), 2.41-2.37 (1 H, m), 2.24 (3 H, s), 2.21 (1 H, m), 1.43 (9 H, s)

ppm; HRMS calcd. for C24H33N2O8 (MH+) 477.2237 Da. Found 477.2219 Da.

aldehyde hydrate 316

MeOMe

MeOOH

N

OOH

NH

OHHO

H

HTFA

1

1511

16

137

5

3

18

9

Ha Hb

10

Ha

Hb17

To a stirred solution of diol 315 (40 mg, 0.084 mmol) in MeOH (1.0 mL) at 0 °C was

added 6 M HCl (1:1 H2O:MeOH, 1.0 mL). The solution was warmed to 25 °C, stirred for

3 h, and the solvents removed under vacuum. The white residue was used directly for

oxidation to quinone. Some material was purified by reverse phase HPLC (preparatory

column, rate = 8.0 mL/min, gradient = 100% to 70% H2O with 0.1% TFA in CH3CN

with 0.1% TFA over 20 min., injection volume = 1.0 mL, product elution from 17-19

min.) and lyophilized to afford 316 as white powder: IR (thin film): 3350, 2944, 1668,

1466, 1417, 1286, 1258, 1201, 1136, 1056 cm-1; 1H NMR (500 MHz, D2O): δ 5.31 (C1, 1

H, t, J = 3.0 Hz), 5.12 (C16, 1 H, d, J = 4.6 Hz), 4.37 (C11, 1 H, s), 4.28 (C13, 1 H, d, J =

6.4 Hz), 3.90 (C18, 1 H, dd, J = 11.8, 4.0 Hz), 3.79 (C3, 1 H, dt, J = 12.2, 2.4 Hz), 3.66

227

(C7-OMe, 3 H, s), 3.61 (C5-OMe, 3 H, s), 3.58 (C18, 1 H, dd, J = 11.7, 2.8 Hz), 2.98 (C4

Hb, 1 H, dd, J = 15.3, 2.6 Hz), 2.86-2.82 (C15, 1 H, m), 2.76 (C4 Ha, 1 H, dd, J = 15.1,

12.3 Hz), 2.44 (C14 Ha, 1 H, dd, J = 14.1, 9.1 Hz), 2.25 (C14 Hb , 1 H, dt, J = 14.2, 6.6

Hz), 2.13 (C6-Me, 3 H, s) ppm; 13C NMR (125 MHz, D2O, carbon signals for C6-10 are

inferred by chemical shift and analogous compounds): δ 167.4 (C17), 147.6 (C5), 144.8

(C7), 142.3 (C8), 124.9 (C9/10), 124.6 (C9/10), 118.6 (C6), 90.0 (C16), 62.1 (C18), 61.3

(C5-OMe), 60.8 (C7-OMe), 59.5 (C13), 59.1 (C11), 56.4 (C3), 52.3 (C1), 41.4 (C15),

31.6 (C14), 24.5 (C4), 9.0 (C6-Me) ppm; HRMS calcd. for C19H27N2O (MH+) 395.1818

Da. Found 395.1825 Da.

(±)-lemonomycinone amide (317)

OMe

MeOO

N

OOH

NH

OHHO

H

HTFA

1

1511

16

137

5

3

18

9

Ha Hb

10

Ha

Hb17

To a stirred solution of amine 316 (0.084 mmol) in H2O (1.0 mL) at 25 °C was added

ammonium cerium(IV) nitrate (0.138 g, 0.252 mmol) in one portion. The reaction was

monitored by reverse phase HPLC and after 4 hours no starting material remained. The

reaction solution was then purified without concentration on reverse phase HPLC (semi-

preparatory column, rate = 1.5 mL/min, gradient = 98% to 60% H2O with 0.1% TFA in

CH3CN with 0.1% TFA over 50 min., injection volume = 0.3 mL, product elution from

25-26 min.) and lyophilized to afford (±)-lemonomycinone amide (317) (14 mg, 35%

228

yield) as an orange residue: 1H NMR (500 MHz, D2O ): δ 5.13 (C16, 1 H, d, J = 4.7 Hz),

5.04 (C1, 1 H, brs), 4.38 (C11, 1 H, s), 4.31 (C13, 1 H, d, J = 6.3 Hz), 4.09 (C18, 1 H, dd,

J = 12.1, 3.4 Hz), 3.84 (C3, 1 H, dt, J = 11.5, 2.9 Hz), 3.84 (C7-OMe, 3 H, s), 3.51 (C18,

1 H, dd, J = 12.2, 2.1 Hz), 2.93 (C4-Hb, 1 H, dd, J = 16.5, 2.8 Hz), 2.81-2.77 (C15, 1 H,

m), 2.48-2.43 (C4-Ha, C14-Ha, 2 H, m), 2.26 (C14-Hb, 1 H, dt, J = 14.4, 6.5 Hz), 1.92

(C6-Me, 3 H, s) ppm; 13C NMR (125 MHz, D2O, carbon signals for C6-10 are inferred by

chemical shift and comparison with analogous compounds, no long-range carbon-

hydrogen correlation was conducted): δ 187.0 (C5), 181.5 (C8), 167.3 (C17), 155.8 (C7),

142.7 (C10), 136.5 (C9), 131.2 (C6), 89.9 (C16), 61.5 (C7-OMe), 61.0 (C18), 59.6 (C13),

58.7 (C11), 55.1 (C3), 52.2 (C1), 41.6 (C15), 31.6 (C14), 23.5 (C4), 8.7 (C6-Me) ppm;

HRMS calcd. for C18H23N2O7 (MH+) 379.1505 Da. Found 379.1535 Da.

5-Chloromethyl-2,4-dimethoxy-3-methyl benzaldehyde (322)

OMe

Me

MeO

HO

Cl

To a refluxing suspension of commercially available 2,4-dimethoxy-3-

methylbenzaldehyde (11.6 g, 64.5 mmol) and zinc chloride (9.3 g, 68.2 mmol) in 37%

aqueous formaldehyde (26 mL) was bubbled HCl gas for 90 min. The reaction solution

became homogeneous and dark orange in color. The reaction was cooled to 25 °C, poured

over ice, and extracted with Et2O (3 x 100 mL). The organic extracts were combined,

washed with aq. NaHCO3 (3 x 50 mL), sat. aq. NaCl (100 mL), dried (Na2SO4), filtered,

229

and concentrated under vacuum. The resulting brown oil was purified by flash column

chromatography (SiO2, 5-15% EtOAc-hexane) to afford benzyl chloride 322 (10.8 g,

73% yield) as a white solid: Rf = 0.41 (20% EtOAc in hexanes); mp = 67-68 °C; IR (thin

film): 2946, 2854, 1682, 1595, 1472, 1472, 1417, 1390, 1307, 1268, 1244, 1187, 1112,

1001 cm-1; 1H NMR (300 MHz, CDCl3): δ 10.29 (1 H, s), 7.79 (1 H, s), 4.63 (2 H, s),

3.92 (3 H, s), 3.90 (3 H, s), 2.28 (3 H, s) ppm; 13C NMR (75 MHz, CDCl3): δ 188.7,

163.1, 128.4, 127.7, 125.9, 125.5, 63.1, 61.3, 40.6, 9.26 ppm; HRMS calcd. for

C11H14O3Cl (MH+) 229.0631 Da. Found 229.0637 Da.

Formic acid 5-chloromethyl-2,4-dimethoxy-3-methyl-phenyl ester (323)

OMe

Me

MeOO

Cl

O

H

To a stirred solution of benzyl chloride 322 (4.0 g, 17.5 mmol) in CH2Cl2 (35 mL) at 0 °C

was added m-CPBA (77% max., 6.6 g, 29.7 mmol max.) in small portions. The reaction

was warmed to 25 °C and stirred for 3 h. The reaction was treated with sat. aq. NaHCO3,

extracted with CH2Cl2 (3 x 40 mL). The combined organic extracts were dried (Na2SO4),

filtered, and concentrated under vacuum to afford the ester 323 (4.1 g, 95% yield) as a

yellow oil: Rf = 0.44 (20% EtOAc in hexanes); IR (thin film): 2944, 2831, 1747, 1484,

1457, 1417, 1319, 1243, 1218, 1100, 1005 cm-1; 1H NMR (300 MHz, CDCl3): δ 8.27 (1

H, s), 7.04 (1 H, s), 4.60 (2 H, s), 3.82 (3 H, s), 3.75 (3 H, s), 2.25 (3 H, s) ppm; 13C NMR

230

(75 MHz, CDCl3): δ 159.0, 155.4, 139.1, 126.8, 126.6, 121.3, 61.3, 60.5, 40.4, 9.7 ppm;

HRMS calcd. for C11H14O4Cl (MH+) 245.0581 Da. Found 245.0575 Da.

5-Chloromethyl-2,4-dimethoxy-3-methyl phenol (324)

OMe

Me

MeOOH

Cl

To a stirred solution of the ester 324 (4.1 g, 16.7 mmol) in THF (94 mL) at 0 °C was

added LiBH4 (0.36 g, 16.7 mmol) in small portions. After 10 min. analysis by TLC

showed that starting material was consumed. The reaction was treated with aq. 1 N HCl

(50 mL) and extracted with EtOAc (1 x 150 mL, 2 x 50 mL). The combined extracts were

washed with sat. aq. NaCl (50 mL), dried (Na2SO4), filtered, and concentrated under

vacuum to afford the phenol as an unstable orange oil. Rf = 0.22 (20% EtOAc in

hexanes); IR (thin film): 3405, 2942, 2834, 1596, 1485, 1457, 1418, 1346, 1307, 1248,

1197, 1114, 1049, 1005 cm-1; 1H NMR (300 MHz, CDCl3): δ 6.85 (1 H, s), 5.58 (1 H,

bs), 4.60 (2 H, s), 3.80 (3 H, s), 3.79 (3 H, s), 2.26 (3 H, s) ppm; 13C NMR (75 MHz,

CDCl3): δ 150.2, 146.1, 145.2, 129.5, 127.7, 126.7, 113.9, 61.4, 60.6, 41.1, 9.6 ppm;

HRMS calcd. for C10H14O3Cl (MH+) 217.0631 Da. Found 217.0629 Da.

231

1-Chloromethyl-2,4-dimethoxy-3-methyl-5-trityloxy benzene (321)

OMe

Me

MeOOTrt

Cl

To solution of the phenol 324 (3.8 g, 16.7 mmol) and triethylamine (7.0 mL, 50.1 mmol)

in CH2Cl2 (90 mL) at 25 °C was added trityl chloride (4.7 g, 16.7 mmol). The reaction

was stirred for 2 h then quenched with sat. aq. NaHCO3 (100 mL) and extracted with

EtOAc (1 x 150 mL, 2 x 50 mL). The combined extracts were washed with sat. aq. NaCl

(50 mL), dried (Na2SO4), filtered, and concentrated under vacuum. The brown residue

was purified by flash column chromatography (SiO2, slow gradient 0-10% EtOAc-

hexane) to afford benzyl chloride 321 (3.89 g, 51% yield over 3 steps) as a white solid.

Approximately 20% decomposition was observed when stored under argon in the dark at

room temperature for two weeks. Rf = 0.46 (20% EtOAc in hexanes); recrystallized from

EtOAc/hexanes, mp = 136-140 °C; IR (thin film): 3059, 3024, 2932, 2853, 1596, 1483,

1448, 1418, 1230, 1116, 1064, 1010, 905, 701 cm-1; 1H NMR (300 MHz, CDCl3): δ 7.48

(6 H, d, J = 6.9 Hz), 7.31-7.20 (9 H, m), 6.35 (1 H, s), 4.26 (2 H, s), 3.87 (3 H, s), 3.68 (3

H, s), 2.13 (3 H, s) ppm; 13C NMR (75 MHz, CDCl3): δ 150.8, 145.3, 143.7, 128.6,

127.5, 127.0, 125.0, 124.0, 120.1, 90.9, 61.2, 60.3, 41.2, 9.4 ppm; HRMS calcd. for

C29H26O3Cl (MH+) 457.1570 Da. Found 457.1569 Da.

232

7-Benzyloxy-6-benzyloxymethyl-3-(2,4-dimethoxy-3-methyl-5-trityloxy-benzyl)-8,10-

dimethoxy-9-methyl-4-oxo-1-phenylsulfanyl-1,3,4,6,11,11a-hexahydro-pyrazino[1,2-

b]isoquinoline-2-carboxylic acid tert-butyl ester (325)

N

MeOMe

MeOOBn

OBnO

SPh

NH

OTrt

OMeMe

OMeBoc

H

To a degassed solution (freeze/pump/thaw) of thioaminal 300 (0.38 g, 0.54 mmol),

benzyl chloride 321 (0.30 g, 0.65 mmol), and 18-crown-6 (0.16 g, 0.59 mmol) in THF

(4.15 mL) at −78 °C was added KHMDS (0.5 M in toluene, 1.3 mL, 0.65 mmol)

dropwise within 2 min. The reaction became orange and remained clear. After 20 min. no

starting material was observed by TLC analysis. The reaction was quenched with sat. aq.

NaHCO3 (25 mL) and extracted with EtOAc (3 x 25 mL). The combined extracts were

washed with sat. aq. NaCl (25 mL), dried (Na2SO4), filtered, and concentrated under

vacuum. The yellow oil was purified by flash column chromatography (SiO2, 15-25%

EtOAc-hexane) to afford alkylated product 325 (0.595 g, 98% yield) as a colorless foam:

Rf = 0.35 (30% EtOAc in hexanes); IR (thin film): 2936, 1701, 1668, 1479, 1449, 1368,

1306, 1162, 1064, 1011, 907, 733 cm-1; 1H NMR (300 MHz, CDCl3), pronounced

carbamate resonance was observed giving sharp signals in ratio of 1:2, (high temperature

NMR [100 °C] led to decomposition): δ 7.66-7.53 (4 H, m), 7.44-7.06 (26 H, m), 6.20

(0.3 H, s), 6.11 (0.7 H, s), 6.02 (0.3 H, t, J = 6.2 Hz), 5.97 (0.7 H, t, J = 6.2 Hz) 5.26 (0.3

H, s), 5.17 (0.3 H, s), 5.13 (1.4 H, s), 5.06 (1 H, t, J = 5.2 Hz), 4.53-4.46 (2.3 H, m), 4.37

(0.7 H, m), 3.86-3.65 (2.0 H, m), 3.80 (2.3 H, s), 3.76 (0.7 H, s), 3.59 (2.3 H, s), 3.54 (0.7

H, s), 3.48 (0.7 H, s), 3.42 (2.3 H, s), 3.36 (3 H, s), 3.06-2.76 (3 H, m), 2.55-2.25 (2 H,

233

m), 2.22 (0.7 H, s), 2.20 (2.3 H, s), 1.53 (0.7 H, s), 1.49 (2.3 H, s), 1.31 (6.3 H, s), 1.17

(2.7 H, s) ppm; 13C NMR (75 MHz, CDCl3): δ 167.9, 153.2, 152.5, 152.2, 150.9, 150.6,

150.0, 145.1, 144.1, 138.6, 137.7, 134.5, 131.4, 129.5, 129.0, 128.8, 128.6, 128.3, 128.1,

128.0, 127.7, 127.4, 127.3, 127.1, 126.8, 126.7, 123.9, 123.2, 122.7, 122.3, 91.3, 81.0,

80.7, 74.3, 72.3, 72.2, 72.0, 69.0, 68.7, 60.5, 60.4, 60.3, 60.2, 60.1, 60.0, 59.0, 58.6, 54.8,

54.7, 48.3, 33.3, 32.3, 28.1, 27.6, 24.9, 24.2, 9.1, 8.9 ppm; HRMS calcd. for

C70H73N2O10S (MH+) 1133.4986 Da. Found 1133.4980 Da.

7-Benzyloxy-6-benzyloxymethyl-3-(2,4-dimethoxy-3-methyl-5-trityloxy-benzyl)-8,10-

dimethoxy-9-methyl-4-oxo-1-phenylsulfanyl-1,3,4,6,11,11a-hexahydro-pyrazino[1,2-

b]isoquinoline-2-carboxylic acid tert-butyl ester (326)

N

MeOMe

MeOOBn

OBnO

SPh

NH

OTrt

OMeMe

OMeBoc

H

To a degassed solution (freeze/pump/thaw) of 325 (0.132 g, 0.117 mmol) in THF (1.2

mL) at −78 °C was added t-BuLi (1.3 M in pentane, 0.224 mL, 0.291 mmole). Upon

complete addition the solution became deep orange/brown in color. Within 10 seconds of

complete t-BuLi addition a degassed solution (freeze/pump/thaw) of butylated

hydroxytoluene (0.103 g, 0.466 mmol) in THF (0.25 mL) was added in one portion. The

brown color faded to pale yellow. The solution was diluted with sat. aq. NaHCO3 (10

mL) and extracted with EtOAc (3 x 15 mL). The combined extracts were washed with

sat. aq. NaCl (15 mL), dried (Na2SO4), filtered, and concentrated under vacuum. 1H

234

NMR of the crude product revealed a 6:1 ratio of 326 to 325. The mixture was purified

by flash column chromatography (SiO2, very slow gradient of 10-20% EtOAc-hexane) to

yield 326 (0.064 g, 48% yield) as a colorless foam and a 3:2 mixture of 326:325 (0.033 g,

25% yield). 326: Rf = 0.39 (30% EtOAc in hexanes); IR (thin film): 3061, 3032, 2936,

2863, 1695, 1653, 1479, 1456, 1417, 1386, 1330, 1289, 1250, 1160, 1115, 1064, 1011,

910, 699 cm-1; 1H NMR (300 MHz, CDCl3) carbamate resonance was observed giving

broad signals rather than sharp splitting: δ 7.57-7.20 (30 H, m), 6.50 (1 H, s), 6.18 (1 H,

s), 5.85 (1 H, brs), 5.15 (2 H, brs), 4.56 (2 H, s), 4.45 (1 H, brs), 3.87 (3 H, s), 3.84 (3 H,

s), 3.86-3.77 (2 H, m), 3.70-3.60 (1 H, m), 3.63 (3 H, s), 3.54 (3 H, s), 3.50-3.43 (1 H,

m), 3.06 (1 H, m), 2.80 (1 H, m), 2.63 (1 H, m), 2.28 (3 H, brs), 2.18 (3 H, brs), 1.32 (2

H, brs), 1.18 (7 H, brs) ppm; 13C NMR (75 MHz, CDCl3): δ 167.4, 153.0, 151.4, 151.1,

150.2, 148.7, 145.0, 144.7, 144.2, 138.6, 137.2, 134.5, 133.3, 131.2, 129.1, 128.9, 128.2,

127.9, 127.8, 127.6, 127.5, 127.4, 127.2, 127.0, 126.8, 126.7, 126.4, 126.3, 124.4, 124.0,

120.7, 90.5, 80.9, 74.5, 72.7, 72.3, 63.0, 60.7, 60.2, 60.1, 60.0, 58.8, 57.3, 49.7, 35.8,

27.8, 27.5, 26.9, 9.6, 9.3 ppm; HRMS calcd. for C70H73N2O10S (MH+) 1133.4986 Da.

Found 1133.5018 Da.

235

7-Benzyloxy-6-benzyloxymethyl-3-(5-hydroxy-2,4-dimethoxy-3-methyl-benzyl)-8,10-

dimethoxy-9-methyl-4-oxo-1-phenylsulfanyl-1,3,4,6,11,11a-hexahydro-pyrazino[1,2-

b]isoquinoline-2-carboxylic acid tert-butyl ester (327)

N

MeOMe

MeOOBn

OBnO

SPh

NH

OH

OMeMe

OMeBoc

H

To a degassed solution (freeze/pump/thaw) of 325 (90 mg, 0.080 mmol) in THF (1.2 mL)

at −78 °C was added t-BuLi (1.1 M in pentane, 0.18 mL, 0.20 mmol). Upon complete

addition the solution became deep orange/brown in color. After 10 seconds a degassed

solution (freeze/pump/thaw) of butylated hydroxyltoluene (0.103 g, 0.466 mmol) in THF

(0.25 mL) was added in one portion. The brown color faded to pale yellow. To the

reaction mixture was added anhydrous HCl (1.0 M in Et2O, 1.0 mL). The solution was

warmed to 0 °C and added HCl in small portions over 1 h until the trityl ether could not

be detected by TLC. The solution was diluted with sat. aq. NaHCO3 (10 mL) and

extracted with EtOAc (2 x 15 mL). The combined extracts were washed with sat. aq.

NaCl (15 mL), dried (Na2SO4), filtered, and concentrated under vacuum. The resulting

yellow residue was purified by flash column chromatography (SiO2, slow gradient, 10-

25% EtOAc-hexane) to afford phenol 327 (0.052 g, 73% yield) as a colorless oil: Rf =

0.26 (30% EtOAc in hexanes); IR (thin film): 3395, 2936, 2863, 1686, 1653, 1456, 1417,

1385, 1334, 1255, 1162, 1115, 1009, 735 cm-1; 1H NMR (300 MHz, CDCl3): δ 7.56 (2 H,

d, J = 7.6 Hz), 7.40-7.17 (13 H, m), 6.64 (0.2 H, s), 6.34 (0.8 H, s), 6.04 (0.8, s), 5.86 (0.2

H, s), 5.86-5.81 (1 H, m), 5.03 (2 H, s), 5.01-4.93 (1 H, m), 4.80 (1 H, dd, J = 8.8, 3.4

Hz), 4.54 (2 H, s), 3.90-3.45 (4 H, m), 3.84 (3 H, s), 3.74 (6 H, s), 3.63 (3 H, s), 3.21-3.00

236

(3 H, m), 2.26 (3 H, s), 2.22 (3 H, s), 1.28 (2.3 H, s), 1.12 (6.7 H, s) ppm; 13C NMR (75

MHz, CDCl3): δ 167.4, 153.5, 151.0, 150.5, 150.3, 144.5, 144.4, 143.9, 138.5, 137.0,

134.0, 131.9, 129.6, 129.1, 128.9, 128.6, 128.4, 128.3, 128.1, 127.8, 127.7, 127.5, 127.2,

127.0, 126.6, 124.4, 123.8, 123.7, 123.5, 114.7, 114.2, 81.1, 74.4, 72.3, 65.2, 61.3, 60.8,

60.3, 60.2, 58.0, 57.5, 56.3, 49.9, 49.5, 35.3, 34.7, 27.7, 27.4, 26.8, 9.6, 9.2 ppm; HRMS

calcd. for C51H59N2O10S (MH+) 891.3890 Da. Found 891.3887 Da

phenol 332

N

MeOMe

MeOOBn

OBnO

NHH

H

HOOMe

Me

OMeH

H

To a solution of 327 (0.043 g, 0.048 mmol) in dry chlorobenzene (1.6 mL) at 25 °C was

added AgBF4 (0.20 mL of a solution of 0.18 g AgBF4 in 0.50 PhCl). A precipitate formed

and the solution remained pale yellow in color. The reaction was complete upon first

TLC analysis (within 5 minutes). After 10 minutes the reaction mixture was diluted with

20% aq. Rochelle’s salt (15 mL) and extracted with EtOAc (3 x 15 mL). The combined

extracts were washed with sat. aq. NaCl (15 mL), dried (Na2SO4), filtered, and

concentrated under vacuum. The resulting yellow residue was purified by flash column

chromatography (SiO2 treated with triethylamine, 100% EtOAc to 5% MeOH in EtOAc)

to afford 332 (0.019 g, 56% yield) as a pale yellow cloudy oil: Rf = 0.17 (5% MeOH in

EtOAc); IR (thin film): 3304, 3030, 2937, 2862, 1635, 1457, 1412, 1361, 1302, 1254,

237

1193, 1112, 1060, 1007, 911, 733 cm-1; 1H NMR (300 MHz, CDCl3): δ 7.42-7.24 (5 H,

m), 7.12-7.10 (3 H, m), 6.81 (2 H, d, J = 3.5 Hz), 5.74 (1 H, t, J = 3.8 Hz), 5.04 (1 H, d, J

= 11.0 Hz), 4.96 (1 H, d, J = 11.0 Hz), 4.60 (1 H, d, J = 3.3 Hz), 4.06-3.95 (3 H, m), 3.80

(3 H, s), 3.78-3.67 (2 H, m), 3.65 (3 H, s), 3.62 (3 H, s), 3.59 (3 H, s), 3.54-3.37 (2 H, m),

3.23-3.03 (2 H, m), 2.33 (1 H, dd, J = 15.0, 12.9 Hz), 2.26-2.22 (NH, ArOH, 2 H, m),

2.23 (3 H, s), 2.16 (3 H, s) ppm; 13C NMR (75 MHz, CDCl3): δ 169.5, 151.0, 149.9,

149.6, 144.6, 143.3, 142.0, 138.5, 137.2, 128.2, 128.1, 127.9, 127.9, 127.8, 127.7, 127.6,

127.5, 126.6, 126.5, 126.2, 123.9, 123.3, 122.9, 117.8, 74.5, 72.5, 72.0, 61.1, 60.5, 60.1,

59.8, 53.2, 49.6, 48.8, 29.5, 26.2, 9.4, 9.2 ppm; HRMS calcd. for C40H45N2O8 (MH+)

681.3176 Da. Found 681.3173 Da.

N-methylamine 333

N

MeOMe

MeOOBn

OBnO

NH

H

HOOMe

Me

OMeH

H

Me

To a stirred solution of phenol 327 (0.20 g, 0.224 mmol) in dry chlorobenzene (7.5 mL)

at 25 °C was added AgBF4 (0.93 mL of a solution of 1.0 g AgBF4 in 2.8 PhCl). A

precipitate formed and the solution remained pale yellow in color. The reaction was

complete upon first TLC analysis (within 5 minutes). After 10 min. the reaction mixture

was diluted with 20% aq. Rochelle’s salt (25 mL) and extracted with EtOAc (3 x 20 mL).

The combined extracts were washed with sat. aq. NaCl (20 mL), dried (Na2SO4), filtered,

238

and concentrated under vacuum. The yellow residue was dissolved in MeOH (16 mL).

37% Aq. formaldehyde (1.2 mL), sodium cyanoborohydride (0.282 g, 4.48 mmol), and

acetic acid (4.0 mL) were added. The yellow solution became colorless. After 40 min. the

solution was concentrated under vacuum, the resultant residue treated with sat. aq.

NaHCO3 (25 mL) and extracted with EtOAc (3 x 25 mL). The combined extracts were

washed with sat. aq. NaCl (20 mL), dried (Na2SO4), filtered, and concentrated under

vacuum. The pale yellow residue was purified by flash column chromatography (SiO2

treated with triethylamine, 50-100% EtOAc in hexanes) to afford methylamine 333

(0.101 g, 65% yield over two steps) as a pale yellow oil: Rf = 0.27 (100% EtOAc in

hexanes); IR (thin film): 3319, 2986, 2937, 2869, 1635, 1455, 1412, 1361, 1302, 1257,

1109, 1058, 1008, 753 cm-1; 1H NMR (300 MHz, CDCl3): δ 7.44-7.27 (5 H, m), 7.11-

7.08 (3 H, m), 6.80-6.77 (2 H, m), 5.92 (1 H, ArOH, s), 5.75 (1 H, t, J = 3.1 Hz), 5.06 (1

H, d, J = 11.0 Hz), 4.97 (1 H, d, J = 11.0 Hz), 4.31 (1 H, d, J = 3.0 Hz), 4.04 (1 H, d, J =

12.1 Hz), 3.98 (1 H, d, J = 12.2 Hz), 3.91-3.83 (1 H, m), 3.80 (3 H, s), 3.72-3.67 (2 H,

m), 3.64 (3 H, s), 3.59 (3 H, s), 3.58 (3 H, s), 3.43-3.38 (2 H, m), 3.14 (1 H, dd, J = 18.1,

6.7 Hz), 2.98 (1 H, d, J = 18.0 Hz), 2.46 (3 H, s), 2.33 (1 H, dd, J = 15.0, 12.9 Hz), 2.23

(3 H, s), 2.15 (3 H, s) ppm; 13C NMR (75 MHz, CDCl3): δ 170.4, 151.0, 149.9, 149.3,

144.6, 143.4, 142.9, 138.4, 137.2, 128.2, 128.0, 127.8, 127.7, 127.6, 126.6, 126.5, 126.2,

126.1, 123.8, 122.7, 116.4, 74.4, 72.5, 71.9, 60.5, 60.1, 59.8, 59.2, 57.0, 55.0, 49.6, 40.1,

25.8, 24.6, 9.4, 9.2 ppm; HRMS calcd. for C41H47N2O8 (MH+) 695.3332 Da. Found

695.3337 Da.

239

diol 334

N

MeOMe

MeOOH

OBnO

NH

H

HOOMe

Me

OMeH

H

Me

To a solution of methylamine 333 (0.31 g, 0.446 mmol) in MeOH (9.0 mL) at 25 °C was

added Pd(OH)2 (moist, Pd content 20%, 62 mg) and the reaction was stirred under an

atmosphere of H2 (balloon pressure) for 1 h. The reaction mixture was filtered through a

plug of Celite®, washed with MeOH, and concentrated under vacuum. The pale yellow

residue was purified by flash column chromatography (SiO2 treated with triethylamine,

75-100% EtOAc in hexanes then 2% MeOH in EtOAc) to afford diol 334 (0.229 g, 85%

yield) as a fine white solid: recrystallized from MeOH with slow diffusion of EtOAc;

X-ray (see Appendix H); mp = 209-211°C (dec.); Rf = 0.45 (6% MeOH in EtOAc); IR

(thin film): 3326, 2938, 2870, 1635, 1464, 1414, 1362, 1302, 1273, 1193, 1109, 1059,

1006, 910 cm-1; 1H NMR (300 MHz, CDCl3): δ 7.19-7.12 (3 H, m), 6.91-6.88 (2 H, m),

6.04 (1 H, s), 5.77 (2 H, brs), 4.33 (1 H, s), 4.20 (1 H, d, J = 12.1 Hz), 4.11 (1 H, d, J =

12.1 Hz), 4.01-3.96 (1 H, m), 3.76 (3 H, s), 3.74-3.68 (1 H, m), 3.64-3.58 (1 H, m), 3.63

(3 H, s), 3.62 (3 H, s), 3.59 (3 H, s), 3.49-3.40 (1 H, m), 3.14 (1 H, dd, J = 18.1, 6.8 Hz),

2.99 (1 H, d, J = 18.0 Hz), 2.48 (3 H, s), 2.27-2.20 (1 H, m), 2.22 (3 H, s), 2.17 (3 H, s)

ppm; 13C NMR (75 MHz, CDCl3): δ 149.3, 148.1, 144.4, 143.4, 142.9, 142.1, 138.0,

127.8, 126.9, 126.6, 125.3, 122.6, 119.6, 77.0, 72.8, 71.7, 60.6, 60.5, 59.8, 59.3, 57.1,

55.0, 49.4, 40.0, 25.6, 24.5, 9.4 ppm; HRMS calcd. for C34H41N2O8 (MH+) 605.2863 Da.

Found 605.2864 Da.

240

triol 335

N

MeOMe

MeOOH

OHO

NH

H

HOOMe

Me

OMeH

H

Me

To a solution of methylamine 333 (0.10 g, 0.144 mmol) in MeOH (2.9 mL) at 25 °C was

added Pd(OH)2 (moist, Pd content 20%, 45 mg), and the reaction was stirred under an

atmosphere of H2 (balloon pressure) for 6 h. The reaction mixture was filtered through a

plug of Celite®, washed with MeOH, and concentrated under vacuum. The pale yellow

residue was purified by flash column chromatography (SiO2 treated with triethylamine,

75-100% EtOAc in hexanes then 5% MeOH in EtOAc) to afford triol 335 (0.073 g, 94%

yield) as a fine white solid: Rf = 0.36 (6% MeOH in EtOAc); IR (thin film): 3333, 2937,

1634, 1463, 1413, 1362, 1302, 1272, 1192, 1110, 1060, 1005 cm-1; 1H NMR (300 MHz,

DMSO-d6): δ 8.74 (1 H, s), 8.48 (1 H, s), 5.45 (1 H, t, J = 4.9 Hz), 4.26 (1 H, brs), 4.19 (1

H, d, J = 3.2 Hz), 3.71 (1 H, dt, J = 12.7, 2.7 Hz), 3.58 (6 H, s), 3.54 (3 H, s), 3.53 (3 H,

s), 3.53-3.51 (1 H, m), 3.28 (1 H, d, J = 15.3 Hz), 3.17 (1 H, m), 2.96 (2 H, m), 2.63 (1 H,

d, J = 17.6 Hz), 2.28 (3 H, s), 2.09 (3 H, s), 2.08 (3 H, s), 2.01 (1 H, dd, J = 15.0, 13.0

Hz) ppm; 13C NMR (75 MHz, DMSO-d6): δ 170.7, 148.3, 147.4, 144.7, 144.1, 144.0,

143.2, 125.7, 122.5, 122.3, 122.1, 120.4, 117.6, 63.7, 60.8, 60.2, 59.7, 59.0, 57.8, 54.8,

50.5, 39.0, 25.6, 24.2, 9.6 ppm; HRMS calcd. for C27H35N2O8 (MH+) 515.2393 Da.

Found 515.2394 Da.

241

bis-isoquinolinequinone 337

N

MeO

MeOO

OHO

NH

OOMe

Me

OMe

1 137

5

3

22

9

10

21

11H16

1518

17

19Ha Hb

HaHb

To a solution of the triol 335 (73 mg, 0.135 mmol) in dry acetonitrile (5.0 mL) at −15 °C

was added ammonium cerium(IV) nitrate (0.369 g, 0.674 mmol) as a solution in water

(0.2 mL), pre-cooled to 0 °C. The orange/brown solution was stirred for 15 minutes as

the temperature rose to −5 °C. The solution was diluted with CHCl3 (15 mL) and 5% aq.

NaHCO3 (5.0 mL), aqueous layer extracted with CHCl3 (2 x 10 mL). The combined

organic extracts were washed with sat. aq. NaCl (5.0 mL), dried (Na2SO4), filtered, and

concentrated under vacuum. The brown residue was purified by flash column

chromatography (SiO2 treated with triethylamine, 50-100% EtOAc in hexanes) to afford

bis-isquinolinequinone 337 (0.036 g, 55% yield) as a burnt orange residue; Rf = 0.35 (5%

MeOH in EtOAc); IR (thin film): 3365, 2925, 2853, 1653, 1617, 1429, 1307, 1232, 1150

cm-1; The numbering scheme used in the isolation paper8 is not consistent with other

renieramycins. Using the numbering scheme consistent with other renieramycins: 1H NMR (500 MHz, CD2Cl2): δ 5.23 (C1, 1 H, dd, J = 4.0, 1.4 Hz), 4.14 (C11, 1 H, brs),

3.98 (C7/17-OMe, 3 H, s), 3.97 (C7/17-OMe, 3 H, s), 3.89 (C3, 1 H, brd, J = 11.4 Hz),

3.73 (C22, 1 H, dd, J = 11.0, 3.5 Hz), 3.68 (C13, 1 H, d, J = 6.7 Hz), 3.43 (C22, 1 H, dd,

J = 11.2, 4.1 Hz), 3.05 (C4-Hb, 1 H, dd, J = 16.4, 2.6 Hz), 2.88 (C14-Hb, 1 H, dd, J =

20.7, 6.8 Hz), 2.66 (C14-Ha, 1 H, brd, J = 20.6 Hz), 2.65 (OH, 1 H, brs), 2.00 (NMe, 3 H,

s), 1.95 (C6/16-Me, 3 H, s), 1.94 (C6/16-Me, 3 H, s), 1.68 (C4-Ha, 1 H, ddd, J = 16.3,

242

12.3, 1.3 Hz) ppm; 13C NMR (125 MHz, CD2Cl2): δ 186.8 (C15), 185.7 (C5), 182.7

(C18), 181.2 (C8), 171.2 (C21), 156.0 (C17), 155.9 (C7), 142.3 (C20/10), 142.2

(C20/10), 137.1 (C9), 135.2 (C19), 129.7 (C6/C16), 129.2 (C6/C16), 65.6 (C22), 61.3

(C7 and C17-OMe), 59.2 (C13), 57.0 (C3), 53.4 (C11), 52.4 (C1), 39.9 (NMe), 25.8 (C4),

24.3 (C14), 8.9 (C6 and C16-Me) ppm; HRMS calcd. for C25H27N2O8 (MH+) 483.1767

Da. Found 483.1786 Da.

(±)-renieramycin G (2g)

N

MeO

MeOO

OO

NH

OOMe

Me

OMe

OMe

Me

H

1 137

5

3

22

9

10

21

11H16

24

15

25

18

26

17

19Ha Hb

HaHb

To a solution of alcohol 337 (6 mg, 0.012 mmol) in CH2Cl2 (0.30 mL) at 25 °C was

added angeloyl chloride9 (approx. 15 μL, 0.120 mmol) and the solution was allowed to

stand for 24 h at 25 °C in the dark. The solution was concentrated under vacuum then

purified by reverse phase HPLC (preparatory column, rate = 8.0 mL/min, gradient = 65%

to 30% H2O without TFA in CH3CN without TFA over 20 min., then to 0% H2O over

next 10 min., injection volume = 1.0 mL, product elution from 23-24 min.) and

lyophilized to afford (±)-renieramycin G (2g) (5 mg, 74% yield) as an orange residue: Rf

243

= 0.25 (100% EtOAc); IR (thin film): 2929, 2854, 1717, 1655, 1616, 1420, 1373, 1351,

1307, 1263, 1229, 1149, 1120 cm-1;

Renieramcyin G was originally characterized in CD2Cl2, however trace amounts of

CH2Cl2 and water made NMR interpretation difficult. C6D6 proved to be a better solvent

for analysis of all signals without overlap or distortion.

The numbering scheme used in the isolation paper8 is not consistent with other

renieramycins. Using the numbering scheme consistent with other renieramycins: 1H NMR (500 MHz, C6D6): δ 5.56 (C1, 1 H, d, J = 2.2 Hz), 5.34 (C26, 1 H, q of q, J =

7.3, 1.4 Hz), 4.92 (C22, 1 H, dd, J = 11.6, 2.4 Hz), 4.52 (C22, 1 H, dd, J = 11.7, 2.3 Hz),

3.82 (C11, 1 H, d, J = 3.5 Hz), 3.79 (C7-OMe, 3 H, s), 3.62 (C17-OMe, 3 H, s), 3.47

(C13, 1 H, d, J = 7.2 Hz), 3.36 (C3, ddd, J = 12.3, 3.2, 3.0 Hz), 3.10 (C4-Hb, 1 H, dd, J =

16.5, 2.7 Hz), 2.87 (C14-Ha, 1 H, d, J = 20.9 Hz), 2.59 (C14-Hb, 1 H, dd, J = 20.5, 7.0

Hz), 1.90 (C6-Me, 3 H, s), 1.87 (C16-Me, 3 H, s), 1.80 (NMe, 3 H, s), 1.55 (C26-Me, 3

H, dq, J = 7.3, 1.5 Hz), 1.51 (C4-Ha, 1 H, ddd, J = 16.7, 12.2, 2.0 Hz), 1.37 (C25-Me, 3

H, dq, J = 1.6, 1.4 Hz) ppm; 13C NMR (125 MHz, C6D6): δ 185.9 (C15), 185.0 (C5),

182.7 (C18), 180.4 (C8), 169.9 (21), 166.8 (C24), 156.4 (C7), 155.5 (C17), 142.2 (C20),

141.1 (C10), 139.1 (C26), 136.3 (C9), 135.1 (C19), 128.8 (C16), 127.4 (C6), 126.9

(C25), 62.8 (C22), 60.6 (C7-OMe), 60.5 (C17-OMe), 59.3 (C13), 56.0 (C3), 53.3 (C11),

50.8 (C1), 39.3 (NMe), 25.9 (C4), 23.6 (C14), 20.3 (C25-Me), 15.3 (C26-Me), 8.6 (C16-

Me and C6-Me) ppm; HRMS calcd. for C30H33N2O9 (MH+) 565.2186 Da. Found

565.2203 Da.

244

245

246

247

248

249

250

251

5.2. REFERENCES

1) Perrin, D. D.; Armarego, W. L. F. Purification of Laboratory Chemicals; Third ed.; Pergamon Press: New York, 1993. 2) Larock, R. C.; Doty, M. J.; Cacchi, S. J. J. Org. Chem. 1993, 58, 4579. 3) Ireland, R. E.; Anderson, R. C.; Badoud, R.; Fitzsimmons, B. J.; McGarvey, G. J.; Thaisrivongs, S.; Wilcox, C. S. J. Am. Chem. Soc. 1983, 105, 1988. 4) Turos, E.; Ren, X. F.; Lake, C. H.; Churchill, M. R. J. Org. Chem. 1995, 60, 6468. 5) D. Saito, T.; Morimoto, M.; Akiyama, C.; Matsumoto, T.; Suzuki, K. J. Am. Chem. Soc. 1995, 117, 10757. 6) Kaufman, T. S. Synlett 1997, 1377. 7) Clayden, J.; Knowles, F. E.; Baldwin, I. R. J. Am. Chem. Soc. 2005, 127, 2412. 8) Davidson, B. S. Tetrahedron Lett. 1992, 33, 3721. 9) Beeby, P.J. Tetrahedron Lett. 1977, 38, 3379.

252

Appendix A: X-Ray Data for the Chloride 198

NCO2Me

SPh

ClH

H

198

253

Table 1. Crystal data and structure refinement for chloride 198

Empirical formula C19 H20 Cl N O2 S

Formula weight 361.87

Temperature 153(2) K

Wavelength 0.71073 Å

Crystal system Monoclinic

Space group P21/c

Unit cell dimensions a = 12.1743(2) Å α = 90°

b = 13.8873(2) Å β = 105.929(1)°

c = 10.3218(1) Å γ = 90°

Volume 1678.08(4) Å3

Z 4

Density (calculated) 1.432 Mg/m3

Absorption coefficient 0.364 mm-1

F(000) 760

Crystal size 0.40 x 0.22 x 0.06 mm

Theta range for data collection 2.93 to 27.48°

Index ranges -15<=h<=15, -13<=k<=18, -13<=l<=13

Reflections collected 6556

Independent reflections 3841 [R(int) = 0.0182]

Completeness to theta = 27.48° 99.9 %

Absorption correction None

Refinement method Full-matrix least-squares on F2

Data / restraints / parameters 3841 / 0 / 298

254

Goodness-of-fit on F2 1.055

Final R indices [I>2sigma(I)] R1 = 0.0341, wR2 = 0.0796

R indices (all data) R1 = 0.0442, wR2 = 0.0841

Largest diff. peak and hole 0.329 and -0.246 e.Å-3

255

Table 2. Atomic coordinates ( x 104) and equivalent isotropic displacement parameters (Å2 x 103) for chloride 198. U(eq) is defined as one third of the trace of the orthogonalized Uij tensor.

________________________________________________________________________

x y z U(eq) ________________________________________________________________________

N1 2227(1) 4245(1) 1789(1) 17(1) C2 1713(1) 3812(1) 452(1) 17(1) C3 2483(1) 3972(1) -453(1) 17(1) C4 2038(1) 4207(1) -1806(2) 21(1) C5 2768(1) 4333(1) -2623(2) 25(1) C6 3936(1) 4224(1) -2087(2) 26(1) C7 4383(1) 4007(1) -732(2) 24(1) C8 3660(1) 3883(1) 97(1) 19(1) C9 4091(1) 3631(1) 1564(2) 21(1) C10 3460(1) 4153(1) 2468(1) 18(1) C11 1514(1) 4777(1) 2321(1) 18(1) O12 485(1) 4834(1) 1843(1) 23(1) O13 2097(1) 5236(1) 3449(1) 22(1) C14 1413(2) 5837(1) 4064(2) 29(1) C15 1456(1) 2747(1) 659(1) 19(1) S16 763(1) 2072(1) -836(1) 23(1) C17 -701(1) 2385(1) -1181(1) 20(1) C18 -1129(1) 3120(1) -538(2) 22(1) C19 -2296(1) 3304(1) -904(2) 26(1) C20 -3037(2) 2768(1) -1900(2) 31(1) C21 -2604(2) 2036(1) -2532(2) 32(1) C22 -1447(1) 1837(1) -2180(2) 25(1) C23 3627(1) 3601(1) 3794(2) 25(1) Cl24 5093(1) 3600(1) 4749(1) 34(1)

________________________________________________________________________

256

Appendix B: X-Ray Data for the Bicyclo[3.3.1] 213

NO

CO2Me

SPh213

257

Table 1. Crystal data and structure refinement for Bicyclo[3.3.1] 213

Empirical formula C19 H19 N O3 S

Formula weight 341.41

Temperature 153(2) K

Wavelength 0.71073 Å

Crystal system Monoclinic

Space group P21/c

Unit cell dimensions a = 11.0472(2) Å α = 90°

b = 7.6308(1) Å β = 92.423(1)°

c = 19.6869(3) Å γ = 90°

Volume 1658.10(4) Å3

Z 4

Density (calculated) 1.368 Mg/m3

Absorption coefficient 0.212 mm-1

F(000) 720

Crystal size 0.29 x 0.25 x 0.04 mm

Theta range for data collection 3.25 to 27.50°

Index ranges -14<=h<=14, -9<=k<=9, -25<=l<=25

Reflections collected 6798

Independent reflections 3797 [R(int) = 0.0218]

Completeness to theta = 27.50° 99.7 %

Absorption correction None

Refinement method Full-matrix least-squares on F2

Data / restraints / parameters 3797 / 0 / 294

258

Goodness-of-fit on F2 1.025

Final R indices [I>2sigma(I)] R1 = 0.0391, wR2 = 0.0753

R indices (all data) R1 = 0.0566, wR2 = 0.0811

Extinction coefficient 5.2(10)x10-6

Largest diff. peak and hole 0.27 and -0.26 e.Å-3

259

Table 2. Atomic coordinates ( x 104) and equivalent isotropic displacement parameters (Å2 x 103) for Bicyclo[3.3.1] 213. U(eq) is defined as one third of the trace of the orthogonalized Uij tensor.

________________________________________________________________________

x y z U(eq) ________________________________________________________________________

O1 2818(1) 5353(1) 2863(1) 23(1) C2 2587(2) 7130(2) 2654(1) 28(1) C3 3362(1) 8469(2) 3045(1) 25(1) C4 4713(2) 8445(2) 2906(1) 28(1) C5 5424(1) 7073(2) 3310(1) 21(1) C6 6657(1) 6836(2) 3206(1) 25(1) C7 7325(2) 5578(2) 3564(1) 27(1) C8 6766(1) 4511(2) 4029(1) 26(1) C9 5546(1) 4744(2) 4144(1) 22(1) C10 4879(1) 6052(2) 3801(1) 19(1) C11 3565(1) 6370(2) 3967(1) 19(1) C12 2723(1) 5110(2) 3572(1) 20(1) N13 3204(1) 8163(2) 3775(1) 22(1) S14 1176(1) 5428(1) 3869(1) 28(1) C15 591(1) 3277(2) 3768(1) 25(1) C16 734(1) 2315(2) 3178(1) 29(1) C17 251(2) 647(2) 3116(1) 41(1) C18 -394(2) -61(3) 3635(1) 51(1) C19 -548(2) 901(3) 4219(1) 54(1) C20 -55(2) 2573(3) 4291(1) 40(1) C21 3331(1) 9559(2) 4203(1) 26(1) O22 3395(1) 11071(1) 4022(1) 37(1) O23 3347(1) 9053(1) 4861(1) 30(1) C24 3490(2) 10477(3) 5345(1) 42(1) ________________________________________________________________________

260

Appendix C: X-Ray Data for the Lactam 229

NNH

CO2Me

O229

261

Table 1. Crystal data and structure refinement for lactam 229

Empirical formula C13 H14 N2 O3

Formula weight 246.26

Temperature 153(2) K

Wavelength 0.71073 Å

Crystal system Orthorhombic

Space group P212121

Unit cell dimensions a = 7.5720(3) Å α = 90°

b = 10.0310(5) Å β= 90°

c = 15.7540(8) Å γ = 90°

Volume 1196.59(10) Å3

Z 4

Density (calculated) 1.367 Mg/m3

Absorption coefficient 0.099 mm-1

F(000) 520

Crystal size 0.36 x 0.35 x 0.06 mm

Theta range for data collection 2.98 to 27.49°

Index ranges -9<=h<=9, -12<=k<=13, -20<=l<=20

Reflections collected 2615

Independent reflections 2615 [R(int) = 0.0000]

Completeness to theta = 27.49° 99.5 %

Absorption correction None

Refinement method Full-matrix least-squares on F2

Data / restraints / parameters 2615 / 0 / 220

262

Goodness-of-fit on F2 1.018

Final R indices [I>2sigma(I)] R1 = 0.0410, wR2 = 0.0784

R indices (all data) R1 = 0.0696, wR2 = 0.0878

Absolute structure parameter -1.4(12)

Extinction coefficient 3.2(3)x10-5

Largest diff. peak and hole 0.159 and -0.166 e.Å-3

263

Table 2. Atomic coordinates ( x 104) and equivalent isotropic displacement parameters (Å2 x 103) for lactam 229. U(eq) is defined as one third of the trace of the orthogonalized Uij tensor.

________________________________________________________________________

x y z U(eq) ________________________________________________________________________

N1 882(2) 3998(2) 5360(1) 30(1) C2 2512(2) 3639(2) 5140(1) 26(1) C3 3977(2) 4687(2) 5212(1) 25(1) C4 4293(2) 5291(2) 4348(1) 25(1) C5 5648(3) 4855(2) 3819(1) 30(1) C6 5902(3) 5440(2) 3032(1) 35(1) C7 4802(3) 6463(2) 2776(1) 36(1) C8 3468(3) 6912(2) 3299(1) 31(1) C9 3199(2) 6336(2) 4096(1) 27(1) C10 1778(3) 6874(2) 4676(1) 33(1) C11 1754(3) 6331(2) 5590(1) 27(1) C12 362(3) 5278(2) 5739(1) 32(1) N13 3471(2) 5740(2) 5803(1) 28(1) O14 2844(2) 2522(1) 4847(1) 32(1) C15 4251(3) 5800(2) 6577(1) 30(1) O16 5564(2) 5177(1) 6773(1) 41(1) O17 3424(2) 6672(1) 7090(1) 40(1) C18 4122(4) 6759(3) 7947(1) 48(1) ________________________________________________________________________

264

Appendix D: X-Ray Data for the Amino Alcohol 283

Me

MeOOBn

OMe

NHOH

OBn

H

H

283

265

Table 1. Crystal data and structure refinement for amino alcohol 283

Empirical formula C28 H33 N O5

Formula weight 463.55

Temperature 153(2) K

Wavelength 0.71073 Å

Crystal system Triclinic

Space group P-1

Unit cell dimensions a = 9.3397(2) Å α = 79.242(2)°

b = 11.4953(3) Å β = 83.470(2)°

c = 12.0017(3) Å γ = 80.402(2)°

Volume 1243.62(5) Å3

Z 2

Density (calculated) 1.238 Mg/m3

Absorption coefficient 0.084 mm-1

F(000) 496

Crystal size 0.30 x 0.30 x 0.10 mm

Theta range for data collection 2.93 to 27.46°

Index ranges -12<=h<=12, -14<=k<=14, -15<=l<=15

Reflections collected 9792

Independent reflections 5646 [R(int) = 0.0454]

Completeness to theta = 27.46° 99.0 %

Max. and min. transmission 0.9916 and 0.9751

Refinement method Full-matrix least-squares on F2

Data / restraints / parameters 5646 / 0 / 316

266

Goodness-of-fit on F2 0.995

Final R indices [I>2sigma(I)] R1 = 0.0512, wR2 = 0.1074

R indices (all data) R1 = 0.1546, wR2 = 0.1376

Extinction coefficient 1.9(2)x10-5

Largest diff. peak and hole 0.261 and -0.253 e.Å-3

267

Table 2. Atomic coordinates ( x 104) and equivalent isotropic displacement parameters (Å2 x 103) for amino alcohol 283. U(eq) is defined as one third of the trace of the orthogonalized Uij tensor.

________________________________________________________________________

x y z U(eq) ________________________________________________________________________ N1 4257(2) 1779(2) 4475(1) 31(1) C2 4021(2) 2924(2) 3678(2) 30(1) C3 2419(2) 3466(2) 3740(2) 28(1) C4 1954(2) 4521(2) 3008(2) 31(1) C5 485(2) 5003(2) 3003(2) 33(1) C6 -564(2) 4448(2) 3724(2) 32(1) C7 -72(2) 3435(2) 4492(2) 32(1) C8 1393(2) 2953(2) 4535(2) 30(1) C9 1865(2) 1935(2) 5468(2) 37(1) C10 3479(2) 1814(2) 5604(2) 32(1) C11 4584(2) 2656(2) 2501(2) 32(1) O12 6110(1) 2243(1) 2525(1) 35(1) C13 6766(2) 1930(2) 1469(2) 43(1) C14 8343(2) 1470(2) 1604(2) 35(1) C15 8934(3) 308(2) 1505(2) 57(1) C16 10380(3) -122(3) 1718(2) 69(1) C17 11219(3) 613(3) 2031(2) 58(1) C18 10645(3) 1775(3) 2103(2) 58(1) C19 9227(2) 2192(2) 1895(2) 49(1) O20 2971(1) 5106(1) 2271(1) 34(1) C21 3355(2) 6103(2) 2671(2) 44(1) C22 4429(2) 6681(2) 1817(2) 34(1) C23 5426(2) 6035(2) 1133(2) 46(1) C24 6431(3) 6591(3) 380(2) 58(1) C25 6466(3) 7789(3) 309(2) 65(1) C26 5478(3) 8435(3) 976(2) 62(1) C27 4460(3) 7888(2) 1730(2) 47(1) O28 46(2) 6084(1) 2303(1) 44(1) C29 241(3) 6069(2) 1113(2) 66(1) C30 -2167(2) 4939(2) 3690(2) 43(1) O31 -1086(2) 2924(1) 5287(1) 40(1) C32 -1589(2) 1926(2) 4983(2) 53(1) C33 3979(2) 671(2) 6404(2) 36(1) O34 5532(2) 397(1) 6374(1) 38(1) ________________________________________________________________________

268

Appendix E: X-Ray Data for Thioaminal 300

Me

MeOOBn

OMe

N

OBn

H

HNBoc

O

SPh

300

269

Table 1. Crystal data and structure refinement for thioaminal 300

Empirical formula C41 H46 N2 O7 S

Formula weight 710.86

Temperature 153(2) K

Wavelength 0.71070 Å

Crystal system Monoclinic

Space group P21/c

Unit cell dimensions a = 17.3357(3) Å α = 90°

b = 18.3236(4) Å β = 92.4980(13)°

c = 11.5161(5) Å γ = 90°

Volume 3654.64(19) Å3

Z 4

Density (calculated) 1.292 Mg/m3

Absorption coefficient 0.142 mm-1

F(000) 1512

Crystal size 0.30 x 0.28 x 0.10 mm

Theta range for data collection 3.05 to 27.43°

Index ranges -18<=h<=21, -21<=k<=23, -13<=l<=14

Reflections collected 18879

Independent reflections 7594 [R(int) = 0.0804]

Completeness to theta = 27.43° 91.0 %

Absorption correction None

Refinement method Full-matrix least-squares on F2

Data / restraints / parameters 7594 / 0 / 460

270

Goodness-of-fit on F2 1.143

Final R indices [I>2sigma(I)] R1 = 0.0819, wR2 = 0.1831

R indices (all data) R1 = 0.1809, wR2 = 0.2095

Largest diff. peak and hole 0.660 and -0.299 e.Å-3

271

Table 2. Atomic coordinates ( x 104) and equivalent isotropic displacement parameters (Å2 x 103) for thioaminal 300. U(eq) is defined as one third of the trace of the orthogonalized Uij tensor.

________________________________________________________________________

x y z U(eq) ________________________________________________________________________ N1 2863(2) 6022(2) 2742(3) 24(1) C2 2909(3) 5958(2) 1577(4) 28(1) C3 3562(3) 6321(3) 974(4) 31(1) N4 4164(2) 6641(2) 1738(3) 23(1) C5 4214(2) 6405(2) 2930(4) 23(1) C6 3430(2) 6448(2) 3448(4) 24(1) C7 3405(2) 6236(2) 4724(4) 25(1) C8 2600(3) 6374(2) 5097(4) 26(1) C9 2440(3) 6779(2) 6078(4) 27(1) C10 1694(3) 7013(2) 6298(4) 30(1) C11 1109(3) 6832(2) 5493(4) 29(1) C12 1243(3) 6397(2) 4536(4) 27(1) C13 1989(3) 6166(2) 4329(4) 25(1) C14 2178(3) 5723(2) 3280(4) 26(1) O15 2422(2) 5621(2) 968(3) 42(1) C16 4637(3) 7149(2) 1279(4) 26(1) O17 4625(2) 7288(2) 237(3) 32(1) O18 5084(2) 7467(2) 2107(3) 36(1) C19 5723(3) 7970(2) 1850(4) 31(1) C20 5428(3) 8599(3) 1119(6) 58(2) C21 6339(3) 7551(3) 1282(6) 61(2) C22 5979(4) 8226(4) 3047(5) 69(2) S23 4552(1) 5442(1) 3045(1) 36(1) C24 5488(3) 5519(2) 2490(4) 32(1) C25 6133(3) 5571(3) 3239(5) 43(1) C26 6866(3) 5594(3) 2792(6) 53(2) C27 6946(4) 5573(3) 1613(6) 56(2) C28 6305(4) 5525(3) 874(6) 57(2) C29 5574(3) 5495(3) 1293(5) 43(1) O30 3054(2) 7014(2) 6821(3) 30(1) C31 3316(3) 6473(3) 7645(4) 40(1) C32 1532(3) 7461(3) 7351(4) 42(1) O33 367(2) 7100(2) 5667(3) 35(1) C34 179(3) 7714(3) 4934(5) 45(1) O35 669(2) 6242(2) 3694(3) 31(1) C36 -42(3) 5932(3) 4053(4) 44(1)

272

C37 -415(3) 5567(2) 3020(4) 30(1) C38 -254(3) 5752(3) 1891(4) 35(1) C39 -606(3 ) 5407(3) 955(5) 45(1) C40 -1128(3) 4854(3) 1113(6) 52(2) C41 -1299(3) 4666(3) 2232(6) 56(2) C42 -962(3 ) 5022(3) 3182(5) 46(1) C43 2304(3) 4905(2) 3549(4) 31(1) O44 1615(2) 4551(2) 3845(3) 41(1) C45 1151(3) 4325(3) 2843(4) 39(1) C46 1448(3) 3651(2) 2269(4) 30(1) C47 1348(3) 2963(3) 2754(4) 40(1) C48 1597(3) 2347(3) 2208(5) 46(1) C49 1937(3) 2400(3) 1151(5) 48(2) C50 2052(3) 3070(3) 664(5) 45(1) C51 1801(3) 3695(3) 1218(4) 39(1) ________________________________________________________________________

273

Appendix F: X-Ray Data for the Allylated Product 301

Me

MeOOBn

OMe

N

OBn

H

HNBoc

O

SPh

301

274

Table 1. Crystal data and structure refinement for allylated product 301

Empirical formula C47 H53 N2 O7 S

Formula weight 789.97

Temperature 153(2) K

Wavelength 0.71073 Å

Crystal system Triclinic

Space group P-1

Unit cell dimensions a = 11.1857(2) Å α = 87.112(1)°

b = 12.2691(3) Å β = 74.288(1)°

c = 16.4143(4) Å γ = 76.143(1)°

Volume 2105.14(8) Å3

Z 2

Density (calculated) 1.246 Mg/m3

Absorption coefficient 0.130 mm-1

F(000) 842

Crystal size 0.30 x 0.30 x 0.10 mm

Theta range for data collection 2.92 to 27.47°

Index ranges -14<=h<=14, -15<=k<=15, -21<=l<=18

Reflections collected 15723

Independent reflections 9454 [R(int) = 0.0895]

Completeness to theta = 27.47° 98.1 %

Absorption correction None

Refinement method Full-matrix least-squares on F2

Data / restraints / parameters 9454 / 0 / 515

275

Goodness-of-fit on F2 0.972

Final R indices [I>2sigma(I)] R1 = 0.0577, wR2 = 0.1094

R indices (all data) R1 = 0.2078, wR2 = 0.1489

Extinction coefficient 1.49(11)x10-5

Largest diff. peak and hole 0.308 and -0.348 e.Å-3

276

Table 2. Atomic coordinates ( x 104) and equivalent isotropic displacement parameters (Å2 x 103) for allylated product 301. U(eq) is defined as one third of the trace of the orthogonalized Uij tensor.

________________________________________________________________________

x y z U(eq) ________________________________________________________________________ N1 4072(2) 2290(2) 3859(2) 24(1) C2 5273(3) 2124(3) 3182(2) 28(1) C3 5120(3) 1980(3) 2305(2) 28(1) N4 3980(2) 2418(2) 2149(1) 24(1) C5 3864(3) 2376(3) 1275(2) 28(1) C6 2864(3) 3364(3) 1126(2) 25(1) C7 3037(3) 3982(3) 387(2) 27(1) C8 2079(3) 4902(3) 287(2) 29(1) C9 917(3) 5201(3) 900(2) 28(1) C10 764(3) 4564(3) 1632(2) 26(1) C11 1725(3) 3675(3) 1759(2) 24(1) C12 1637(3) 3061(3) 2578(2) 25(1) C13 2870(3) 2944(3) 2838(2) 24(1) C14 2882(3) 2308(2) 3653(2) 24(1) C15 4198(3) 2103(3) 4670(2) 28(1) O16 5227(2) 1907(2) 4827(1) 38(1) O17 3065(2) 2187(2) 5239(1) 29(1) C18 3024(3) 2029(3) 6148(2) 29(1) C19 3504(3) 2939(3) 6458(2) 51(1) C20 1609(3) 2152(3) 6559(2) 41(1) C21 3766(3) 863(3) 6278(2) 45(1) C22 5969(3) 3069(3) 3154(2) 35(1) C23 5242(3) 4200(3) 2974(2) 38(1) C24 4761(3) 5035(3) 3518(2) 56(1) O25 6071(2) 1514(2) 1746(1) 37(1) C26 3603(3) 1239(3) 1135(2) 33(1) O27 3577(2) 1168(2) 277(1) 37(1) C28 3202(3) 174(3) 118(2) 40(1) C29 1846(3) 174(3) 575(2) 30(1) C30 1539(3) -756(3) 997(2) 41(1) C31 276(4) -770(4) 1386(2) 55(1) C32 -689(4) 156(5) 1367(3) 63(1) C33 -378(4) 1088(4) 960(3) 67(1) C34 868(4) 1108(3) 564(2) 53(1) O35 4200(2) 3720(2) -225(1) 31(1) C36 4150(3) 3237(3) -1002(2) 37(1)

277

C37 5453(3) 3021(3) -1598(2) 26(1) C38 6284(3) 3701(3) -1611(2) 32(1) C39 7465(3) 3504(3) -2185(2) 37(1) C40 7841(3) 2631(3) -2772(2) 36(1) C41 7019(3) 1953(3) -2773(2) 39(1) C42 5846(3) 2142(3) -2185(2) 33(1) O43 2263(2) 5514(2) -451(1) 36(1) C44 3038(3) 6302(3) -475(2) 43(1) C45 -107(3 ) 6192(3) 776(2) 36(1) O46 -375(2) 4885(2) 2276(1) 30(1) C47 -1253(3) 4199(3) 2298(2) 39(1) S48 2718(1) 852(1) 3595(1) 33(1) C49 1028(3) 1035(3) 3885(2) 27(1) C50 287(3) 1513(3) 4659(2) 41(1) C51 -1031(3) 1647(3) 4881(2) 48(1) C52 -1603(3) 1283(3) 4341(2) 44(1) C53 -871(3) 801(3) 3571(2) 36(1) C54 442(3) 678(3) 3338(2) 31(1) C1A -759(5) 4897(4) 4501(3) 74(1) C2A 369(5) 5220(3) 4161(3) 67(1) C3A 1128(4) 5324(3) 4664(3) 68(1) ________________________________________________________________________

278

Appendix G: X-Ray Data for Oxime 314

NN

H

O

MeOMe

OBn

H

H

OBn

Boc

MeO

NHOH

H

314

279

Table 1. Crystal data and structure refinement for oxime 314

Empirical formula C38 H45 N3 O8

Formula weight 671.77

Temperature 153(2) K

Wavelength 0.71073 Å

Crystal system Triclinic

Space group P-1

Unit cell dimensions a = 8.8203(5) Å α = 102.634(1)°

b = 12.4682(3) Å β = 90.790(1)°

c = 16.9791(2) Å γ = 98.378(1)°

Volume 1800.57(11) Å3

Z 2

Density (calculated) 1.239 Mg/m3

Absorption coefficient 0.087 mm-1

F(000) 716

Crystal size 0.30 x 0.13 x 0.12 mm

Theta range for data collection 3.05 to 27.49°

Index ranges -11<=h<=11, -15<=k<=16, -21<=l<=21

Reflections collected 13638

Independent reflections 13638

Completeness to theta = 27.49° 98.6 %

Absorption correction None

Refinement method Full-matrix least-squares on F2

Data / restraints / parameters 13638 / 14 / 459

280

Goodness-of-fit on F2 1.443

Final R indices [I>2sigma(I)] R1 = 0.0905, wR2 = 0.1693

R indices (all data) R1 = 0.2415, wR2 = 0.1939

Extinction coefficient 1.15(12)x10-5

Largest diff. peak and hole 0.331 and -0.309 e.Å-3

281

Table 2. Atomic coordinates ( x 104) and equivalent isotropic displacement parameters (Å2 x 103) for oxime 314. U(eq) is defined as one third of the trace of the orthogonalized Uij tensor.

________________________________________________________________________

x y z U(eq) ________________________________________________________________________ N1 2139(3) 5707(2) 2757(2) 29(1) C2 992(5) 5067(3) 3052(3) 37(1) C3 1126(4) 3882(3) 3018(2) 34(1) C4 2281(4) 3829(3) 3689(2) 38(1) C5 3848(4) 4178(3) 3350(2) 31(1) C6 3485(4) 4076(3) 2433(2) 27(1) C7 3307(4) 5177(3) 2249(2) 25(1) C8 4784(4) 6009(3) 2308(2) 28(1) C9 4394(4) 7007(3) 2035(2) 24(1) C10 5278(4) 7502(3) 1494(2) 29(1) C11 4933(4) 8455(3) 1263(2) 27(1) C12 3690(4) 8875(3) 1572(2) 31(1) C13 2750(4) 8381(3) 2102(2) 29(1) C14 3122(4) 7440(3) 2316(2) 25(1) C15 2106(4) 6907(3) 2880(2) 30(1) O16 -82(3) 5495(2) 3402(2) 43(1) N17 1935(3) 3400(2) 2305(2) 30(1) C18 1277(5) 3084(3) 1530(3) 36(1) O19 2021(3) 2888(2) 935(2) 41(1) O20 -260(3) 3005(2) 1559(2) 43(1) C21 -1235(5) 2635(4) 794(3) 59(1) C22 -1037(6) 1465(4) 396(3) 80(2) C23 -869(5) 3448(4) 257(3) 78(2) C24 -2811(5) 2687(5) 1121(3) 94(2) C25 5049(5) 3488(4) 3477(3) 45(1) N26 6495(5) 3641(4) 3449(3) 44(1) O27 6859(4) 4710(3) 3297(2) 45(1) N26' 6281(12) 4208(12) 3701(12) 57(6) O27' 7691(13) 3875(9) 3854(7) 44(3) O28 6604(3) 7077(2) 1218(2) 33(1) C29 6290(4) 6202(3) 498(2) 47(1) C30 5960(4) 9007(3) 715(2) 42(1) O31 3272(3) 9818(2) 1343(2) 41(1) C32 3645(5) 10833(3) 1937(3) 52(1) O33 1525(3) 8866(2) 2452(1) 32(1) C34 178(4) 8757(3) 1930(2) 46(1)

282

C35 -925(4) 9407(3) 2387(2) 31(1) C36 -858(4) 10517(3) 2400(2) 36(1) C37 -1898(4) 11156(3) 2825(3) 41(1) C38 -2996(4) 10636(4) 3248(2) 39(1) C39 -3076(4) 9531(3) 3261(2) 38(1) C40 -2037(5) 8930(3) 2834(3) 43(1) C41 2468(4) 7492(3) 3769(2) 34(1) O42 4065(3) 7565(2) 3953(2) 32(1) C43 4431(4) 8002(3) 4802(2) 37(1) C44 6111(4) 8167(3) 4955(2) 27(1) C45 6829(4) 9079(3) 5522(2) 36(1) C46 8409(5) 9235(3) 5676(2) 41(1) C47 9289(5) 8497(4) 5253(2) 40(1) C48 8577(5) 7578(3) 4702(2) 40(1) C49 7007(4) 7428(3) 4562(2) 34(1) ________________________________________________________________________

283

Appendix H: X-Ray Data for the Diol 334

OMeMe

MeOOH

NN

HOOMe

Me

OMeMe

H

H

H

H

OBnO

334

284

Table 1. Crystal data and structure refinement for diol 334

Empirical formula C34.50 H42 N2 O8.50

Formula weight 620.70

Temperature 153(2) K

Wavelength 0.71073 Å

Crystal system Triclinic

Space group P-1

Unit cell dimensions a = 9.0359(4) Å α = 87.720(2)°

b = 12.6771(6) Å β = 78.039(2)°

c = 14.4654(7) Å γ = 79.337(2)°

Volume 1593.03(13) Å3

Z 2

Density (calculated) 1.294 Mg/m3

Absorption coefficient 0.093 mm-1

F(000) 662

Crystal size 0.50 x 0.06 x 0.05 mm

Theta range for data collection 2.99 to 27.41°

Index ranges -11<=h<=8, -16<=k<=13, -18<=l<=18

Reflections collected 9982

Independent reflections 6911 [R(int) = 0.0950]

Completeness to theta = 27.41° 95.3 %

Absorption correction None

Refinement method Full-matrix least-squares on F2

Data / restraints / parameters 6911 / 0 / 424

285

Goodness-of-fit on F2 0.956

Final R indices [I>2sigma(I)] R1 = 0.0781, wR2 = 0.1116

R indices (all data) R1 = 0.3121, wR2 = 0.1601

Extinction coefficient 5.9(10)x10-6

Largest diff. peak and hole 0.260 and -0.236 e.Å-3

286

Table 2. Atomic coordinates ( x 104) and equivalent isotropic displacement parameters (Å2 x 103) for diol 334. U(eq) is defined as one third of the trace of the orthogonalized Uij tensor.

________________________________________________________________________

x y z U(eq) ________________________________________________________________________ N1 5746(4) 3576(3) 6039(2) 27(1) C2 4944(6) 2807(3) 5948(3) 27(1) C3 5811(5) 1723(3) 5589(3) 29(1) C4 6107(5) 1001(3) 6429(3) 32(1) C5 7517(5) 1162(3) 6776(3) 28(1) C6 7862(6) 659(3) 7599(3) 35(1) C7 9238(6) 673(4) 7896(3) 35(1) C8 10268(6) 1225(3) 7325(3) 34(1) C9 9953(6) 1733(3) 6506(3) 30(1) C10 8569(6) 1733(3) 6237(3) 27(1) C11 8185(5) 2346(3) 5375(3) 27(1) C12 7381(5) 3515(3) 5569(3) 27(1) C13 8185(5) 4131(3) 6135(3) 29(1) C14 7377(5) 5269(3) 6281(3) 26(1) C15 8162(6) 6120(3) 6297(3) 30(1) C16 7397(6) 7175(3) 6406(3) 29(1) C17 5818(5) 7376(3) 6461(3) 27(1) C18 5022(6) 6553(3) 6424(3) 29(1) C19 5796(5) 5486(3) 6357(3) 24(1) C20 4853(5) 4606(3) 6465(3) 30(1) N21 7238(4) 1821(3) 4888(2) 30(1) O22 3533(4) 2936(2) 6234(2) 36(1) C23 8048(5) 769(3) 4473(3) 36(1) O24 6787(4) 93(2) 8141(2) 44(1) C25 5721(6) 754(4) 8852(3) 63(2) C26 9590(6) 103(4) 8774(3) 55(2) O27 11689(4) 1243(2) 7556(2) 41(1) C28 11604(6) 2102(4) 8210(3) 59(2) O29 11004(4) 2260(2) 5933(2) 35(1) O30 9765(4) 5912(2) 6170(2) 36(1) C31 10350(6) 5475(4) 6975(3) 54(2) C32 8237(5) 8087(3) 6481(3) 34(1) O33 5059(3) 8446(2) 6503(2) 36(1) C34 4155(5) 8752(3) 7417(3) 47(1) O35 3446(4) 6775(2) 6501(2) 32(1) C36 4214(6) 4447(4) 7513(3) 42(1)

287

O37 5445(4) 4127(2) 7981(2) 50(1) C38 4971(7) 3914(4) 8950(3) 66(2) C39 6364(9) 3775(4) 9411(4) 64(2) C40 6378(11) 3215(5) 10233(5) 126(3) C41 7588(13) 3145(6) 10704(6) 124(4) C42 8800(11) 3615(6) 10320(6) 108(3) C43 8823(8) 4190(6) 9492(5) 95(2) C44 7586(8) 4272(5) 9035(4) 68(2) O1A 6645(10) -1764(6) 9328(5) 89(3) C2A 7563(16) -2716(9) 8954(9) 121(6) ________________________________________________________________________

288

Abbreviations

Ac acetyl AIBN 2,2’-azobisisobutyronitrile Ar aryl atm atmosphere BHT butylated hydroxytoluene Bn benzyl Boc tert-butyloxycarbonyl BOM benzyloxymethyl BOPCl bis(2-oxo-3-oxazolidinyl)phosphinic chloride br broad Bu butyl CAN ammonium cerium(IV) nitrate cat. catalytic Cbz benzyloxycarbonyl CI chemical ionization m-CPBA m-chloroperoxybenzoic acid CSA camphorsulfonic acid d doublet DABCO 1,3-diazabicyclo[2.2.2]octane DBU 1,8-diazabicyclo[5.4.0]undec-7-ene DCC 1,3-dicyclohexylcarbodiimide DCE 1,2-dichloroethane DCM dichloromethane dd double doublet DDQ 2,3-dichloro-5,6-dicyano-1,4-benzoquinone DEAD diethyl azodicarboxylate DIAD diisopropyl azodicarboxylate DIBAL diisobutylaluminium hydride DMAP 4-N,N-dimethylaminopyridine DMDO dimethyldioxirane DMF N,N-dimethylformamide DMP Dess-Martin periodinane DMSO dimethylsulfoxide DMTS dimethylthexylsilyl equiv. equivalents Et ethyl Fmoc 9-fluorenylmethoxycarbonyl HRMS high resolution mass spectrometry imid. imidazole IR infrared KHMDS potassium bis(trimethylsilyl)amide LAH lithium aluminum hydride

289

m multiplet m meta M molar Me methyl mol mole MOM methoxymethyl mp melting point Ms methanesulfonyl MS mass spectrometry NCS N-chlorosuccinimide NIS N-iodosuccinimide NMR nuclear magnetic tesonance o ortho p para PCC pyridinium chlorochromate Ph phenyl Pht phthalimidyl PIFA [bis(trifluoroacetoxy)iodo]benzene PMB para-methoxybenzyl ppm parts per million Pr propyl pyr. pyridine PyBOP® benzotriazolyloxy-tris[pyrrolidino]-phosphonium hexafluorophosphate ORTEP Oak Ridge thermal ellipsoid program oxid. oxidation R alkyl q quartet s singlet t triplet TBAF tetrabutylammonium fluoride TBDPS tert-butyldiphenylsilyl TBDMS or TBS tert-butyldimethylsilyl TEA triethylamine TFA trifluoroacetic acid TFAA trifluoroacetic anhydride THF tetrahydrofuran TIPS triisopropylsilyl TLC thin layer chromatography TMEDA N,N,N',N'-tetramethylethylenediamine TMS trimethylsilyl TMSOTf trimethylsilyl trifluoromethanesulfonate Trt trityl Ts toluenesulfonyl TsOH or pTSA toluenesulfonic acid

290

Vita

Kenneth Stanley Matthews was born in Stockton, CA on December 16th, 1976,

the son of Mr. Stanley and Gay Matthews. He attended Lincoln High School in Stockton,

CA and graduated in May of 1995. From September 1995 to June 1999 he attended The

University of California at San Diego and graduated with a Bachelor of Science degree in

chemistry. After graduation he began work at Corvas International, Inc. as a Research

Associate until leaving for graduate school in August 2000. He entered the graduate

program of The University of Texas at Austin to obtain a Ph.D. in organic chemistry

under the supervision of Prof. Philip D. Magnus.

Permanent address: 3717 Moultrie Dr. Stockton CA 95129

This dissertation was typed by the author.