a de novo approach towards glycosylation using iterative ...rx915s49r/... · behind my success and...

187
A De Novo Approach Towards Glycosylation using Iterative Pd-/B-Dual Catalysis: Applications in Natural Products Synthesis, Digitoxin analogues, Oligomannose motifs and their SAR Studies by Sumit O. Bajaj B.Sc. in Chemistry, Physics, Maths, Amravati University M.Sc. in Chemistry, Sant Gadge Baba Amravati University A dissertation submitted to The Faculty of the College of Science of Northeastern University in partial fulfillment of the requirements for the Doctor of Philosophy in the July 10, 2015 Dissertation directed by George A. O’Doherty Professor of Chemistry and Chemical Biology

Upload: phungtram

Post on 05-Feb-2018

214 views

Category:

Documents


0 download

TRANSCRIPT

  • A De Novo Approach Towards Glycosylation using Iterative Pd-/B-Dual Catalysis: Applications in Natural Products Synthesis, Digitoxin analogues, Oligomannose

    motifs and their SAR Studies

    by Sumit O. Bajaj

    B.Sc. in Chemistry, Physics, Maths, Amravati University M.Sc. in Chemistry, Sant Gadge Baba Amravati University

    A dissertation submitted to

    The Faculty of the College of Science of

    Northeastern University in partial fulfillment of the requirements

    for the Doctor of Philosophy in the

    July 10, 2015

    Dissertation directed by

    George A. ODoherty Professor of Chemistry and Chemical Biology

  • ii

    Dedication

    To the loving memory of my beloved Grandfather Late Seth Shri Mansaramji Hariramji Bajaj, Lohi, Maharashtra, India, the person behind my success.

    ToMy Grandmother Shrimati Saraswatibai Mansaramji Bajaj, the inspiration behind my success and my positive attitude.

  • iii

    Acknowledgements

    First, I would like to express my sincere gratitude towards my advisor Prof. George A.

    ODoherty for his excellent mentorship, guidance, encouraging conversation during the course of

    my studies at Northeastern University. I am grateful that he was always there to support me

    when circumstances became difficult in my graduate studies. I am very thankful to him for

    providing an excellent opportunity to work on research projects and learn new techniques.

    Whenever I encountered problem, he was always been very helpful to resolve these through

    critical thinking, discussion and positive attitude. Whenever I told him that the reaction is not

    working, his favorite quote Try it again. I want to express my gratitude to Prof. James Aggen,

    Prof. Roman Manetsch, Prof. Ke Zhang, Prof. Graham Jones and Prof. Michael Pollastri, to be

    on/part of my dissertation committee, discussing chemistry and their valuable suggestions on my

    thesis. Everyone was friendly and supportive.

    I feel that I am fortunate to work in the ODoherty group where all the group members

    both past and present, were most helpful during my tenure. Specially, I would like to thank the

    ODoherty group members- Dr. Miaosheng Li, Dr. Mingde Shan, Dr. Xiaomei Yu, Dr. Sang-

    woo Kang, Dr. Qian Chen, Dr. Rajender Vemula, Dr. Yalan Xing, Dr. Bulan Wu, Dr. Mingzong

    Li, Dr. Hongyan Li, Dr. Leo Wang, Dr. Ehesan Sharif, Dr. Qi Zhang, Dr. Yanping Wang, Dr.

    Michael Cuccarese, Melvin Rajaratnam, Dr. Yashan Zhong, Dr. Pei Shi, Debarpita Ray, Yu Li,

    Jiamin Zheng, Yuzi Ma, Chao Liang, Xiaofan Liu, Alhanouf Aljahdali and all the other graduate

    students, postdocs and undergraduates for their friendship and assistance in the department of

    chemistry and chemical biology at Northeastern University, Boston MA.

    I would like to thank my grandfather late Shri. Mansaramji Hariramji Bajaj, to whom this

    dissertation is dedicated for the enormous love, care and strong blessings in the form of

  • iv

    Ashirwad. His positive attitude, guidance to work hard and honestly made me a completely

    different personality than what I was before. Most importantly I would like to thank my

    Grandmother Shrimati Saraswatibai Mansaramji Bajaj for her enormous love, care and advice

    she gave me for my successful career. I would like to thank my maternal grandparents Shri.

    Narayandasji Lahoti and Sau. Shantabai N. Lahoti. I would like to thank my grandfather Shri.

    Laxminarayanji H. Bajaj.

    I would like to bow my head towards my parents Shri. Omprakash M. Bajaj, Sau. Pushpa

    O. Bajaj for their enormous love, education, guidance they showered on me. I would like to

    thank my elder brother Amit O. Bajaj and younger brother Satish O. Bajaj for their guidance and

    affection they showered on me. I would like to especially thank my sister-in-law Sau. Jyoti A.

    Bajaj and Sau. Kirti S. Bajaj for their enormous care, very delicious food and affection they

    showed towards me during my PhD career. The most important personality which reminds me of

    my childhood days, my loving niece, my sweetheart Palak Amit Bajaj, I would like to thank her

    about this, its totally a different feeling when I hear Sumit Chachu from her.

    Then comes the turn of all my uncles Shri Manaklalji M. Bajaj, Kamalkishorji M. Bajaj,

    Madhusudanji L. Bajaj, Chandraprakashji M. Bajaj and Sanjay N. Lahoti, all aunts for their love,

    inspiration and guidance.

    I am so happy that I met my beloved wife, Ms. Deepa R. Atal during my visit to India in

    2015. I would like to thank her for enormous care, love and being a very strong positive

    motivator, supporter during my final Ph.D dissertation writing, even words are shorter to express

    my feelings towards her. I would also like to thank my Father-in-law, Advocate Ratanlalji B.

    Atal and mother-in-law Prabhadevi R. Atal for their enormous support and love. Special thanks

  • v

    to Dr. Mohanji Biyani, Sandeepji Rathi, Advocate Riteshji G. Boob, Mr. Manishji Daga, Dr.

    Jyoti M. Biyani, Mrs. Kanchan M. Daga, Ms. Vrunda R. Atal (CA), Girdhar M. Biyani, Krishna

    Daga, Soham Daga, Nandini Biyani, Naman Biyani for their love, guidance and support in my

    decisions.

    I would like to thank my brothers Deepak M. Bajaj, Harshal K. Bajaj, Swapnil Bajaj,

    Niket C. Bajaj, to all my sister-in-laws Sau. Aarti D. Bajaj, Neha H. Bajaj, to all my sisters

    Hemal M. Bajaj, Suhati K. Bajaj, Shital K. Bajaj, and Mangla M. Bajaj and Monali C. Bajaj, also

    to all other nieces Urvi D. Bajaj, Rucha D. Bajaj, Devanshi R. Rathi, Harshita R. Harkanth,

    nephew Manan N. Gandhi, Rishab Rathi and Vidhan N. Gandhi. I would also like to thanks my

    brother in laws for their encouragement throughout my studies.

    I would like to thank all my friends/roommates Dr. Ehesan Sharif, Dr. Ashish Bambal, Javeeria

    Ghani, Dr. Somu Chatterjee, Dr. Subodh Kumar, Dr. Manoj Gupta, Mrs Nikita Manoj Gupta, Dr.

    Yamini Sharma, Ritika Tulshan, Manjari Tulshan, Amol bhai, Sandeep Lahiri, Chander Prasad

    Padamalai, Sanjiv Kaushik, Valmik Doshi, Nimit Shah, Harsha Shekhar, Tathagat Pathak, Pratik

    Patel, Kunal Sharma, Ankith Gupta, Arun, Sanjog Rathi, Rakesh Dand, Nimish Somaiya, Alok

    Vishwakarma, Nilesh Dhurve, Swati Gupta, Subhash for their help, friendship and guidance in

    daily chores. I would like to thank Dr. Novruz G. Akhmedov and Dr. Roger Kautz to help me to

    crack down some oligosaccharide structures through NMR confirmation and their friendship. I

    would like to thank the Northeastern University for the financial support. I would also like to

    thank Cara Shockley, Andy Bean for their help and constant support in the administrative work.

    I would also like to thanks Dr. Stewart Campbell, Dr. Jeff Arnold, Mr. Richard Laura, Dr. Greg

    Mercer and Dr. Tesmol George for their mentorship, useful suggestions/discussions during my

  • vi

    internship at Corden Pharma, MA. Finally, I would like to thanks every single individual whom I

    interacted. I would like to thanks people from my village for the care, love and blessings they

    showered on me. I would like to Pranam and thanks Ashutoshji Swami and Avdhoot Swamiji for

    their spiritual guidance.

    Finally, I bow my head to the ALMIGHTY GOD who gave me enough patience, strength,

    courage, positive attitude and good people in the form of parents, brothers, sisters, wife, in-laws,

    niece, nephews, friends and mentors to handle and solve the problems during my PhD career.

    Without the above mentioned people and gods grace, I would not have been here defending my

    thesis.

  • vii

    Abstract of Dissertation

    Carbohydrates containing natural products regularly play an impressive role in the

    biological activities, like target binding, tissue targeting, cell-cell interaction, cell-cell

    recognition and membrane transportation. Despite of the important biological properties, these

    highly complex oligosaccharide natural products are still challenging synthetic targets to make.

    The ODoherty group has been using a de novo asymmetric approach to build on the desired

    functionality and chirality within the molecule starting from achiral moiety. The importance of

    this approach comes from the fact that all the stereocenter(s)/chirality in the molecule are built

    using an achiral starting material (2-acetyl furan), in contrast to the traditional approaches which

    starts from the known carbohydrate starting materials. This approach relies on a highly dia-

    stereoselective Palladium (0)-catalyzed glycosylation reaction to control the anomeric

    stereochemistry, followed by post-glycosylation transformations (Luche reduction, Upjohn

    dihydroxylation) to install the desired functionality and stereochemistry in the sugar moiety. In

    this regard, we have developed Pd-/B-dual catalysis for the regio- and stereo-selective synthesis

    of complex oligosaccharide class of natural products which in turn help us to expand the SAR

    network towards the anti-cancer studies.

    Oligo-mannose related motifs got most attention as they are ubiquitously present on the

    surface of the mammalian cells and has been looked as an emerging field towards the studies

    related to cancer/HIV vaccine development. Building oligosaccharide motifs in a regioselective

    manner was always seen as a challenge, to address this issue we have developed and utilized the

    Pd-/B-dual catalysis for the regioselective glycosylation of mannose related oligosaccharides

    based on protecting group free approach. Working towards this goal, we successfully bis-

    glycosylated -D-mannose to form trisaccharides and pentasaccharide oligomannose motifs

  • viii

    using the Pd-/B-catalysis.

    Mezzettiasides, a class of partially acetylated highly complex oligosaccharide natural

    products possess anti-cancer activities in the M range and were isolated from the fruit and stem

    bark of Mezzettia leptopoda. They are divided into 3 subclass i.e., disaccharides (Mezzettiaside-

    9-11), trisaccharides (Mezzettiaside 2-3 and 8), tetrasaccharides (Mezzettiaside 5-7) attached to

    each other in 1,3-linked fashion. Inspired from the synthesis and biological importance of a

    related class of natural products, the cleistriosides and cleistetrosides, we synthesized and

    evaluated the SAR studies of mezzettiasides by exploiting a de novo asymmetric approach (i.e.,

    building chirality in the molecules from achiral starting material). This highly divergent first total

    synthesis features the iterative use of Pd-/B-dual catalyst using nucleophilic boron/electrophilic

    palladium for the regio-/stereo-selective glycosylation. It is worth noting that syntheses of all this

    10 divergent members were completed in a range of 13-22 linear steps, yet only 42 total steps

    were required. Only 1 protecting group (AcCl) was used throughout the synthesis of 9 final

    products whereas, for the synthesis of 10th member, the same protecting group (AcCl) was used

    two times. Along the line, four new disaccharide unnatural analogues were synthesized for

    studying the different acetylation pattern (Ac/n-Pr/i-Pr/t-Bu) and concluded that smaller the

    group, better the activity and less sterically hindered acetylating agent are more potent than the

    sterically hindered ones. Chapter 2 describes this in details.

    Based on the developed Pd-/B-dual catalyzed glycosylation methodology, we planned to

    utilize this concept towards the synthesis of digitoxin analogues using protecting group free

    strategy, planning towards the synthesis of digitoxin 1,3-linked analogues. Digitoxin is a

    naturally occurring cardiac glycoside which has been used in the treatment of congestive heart

    failure but due to toxicity, its applications in the clinics are limited. Recent study focuses

  • ix

    digitoxins potential application in cancer and viral (HCMV) infections. In this regards, we and

    several other groups have recently reported that digitoxin can be studied towards its potential as

    an anti-cancer agent. Our final goal is to synthesize the novel molecule(s) which will selectively

    target the tumor cells while maintaining the efficacy and high potency with low toxicity. In this

    regard, we have made a library of digitoxin oligosaccharide (mono- to tetra-saccharides) with -

    L/D and performed SAR studies. From the SAR studies it was concluded that sugar

    stereochemistry plays an important role in the anticancer studies. Chapter 3 describes this in

    detail.

    With the great success of 1,3-linked digitoxin oligosaccharide analogues, we planned to

    shift our focus towards the amino version of the digitoxin. As amines are the most interesting

    pharmaceuticals and are present as a backbone in the most active molecules, may be because

    amine containing compound are much more soluble, can form H-bonding, interact with the

    proteins and can pass through the membrane faster and has a tremendous effect on the potency.

    Working towards this aim of synthesizing digitoxin amino analogues, we invested our Pd-/B-

    methodology for the synthesis of mono and di-saccharide amino analogues and finally compared

    the importance of NH2 vs OH group towards the potency by varying the installation of amine

    group on different carbohydrate chain. Along this path, we have also successfully glycosylated

    the natural digitoxin molecule on the C-4 position of the last digitoxose sugar so that we can

    make the tetrasaccharide amino compounds with -L/D stereochemistry and check its

    applications towards the cancer cell lines.

  • x

    Table of Contents

    Dedication....ii

    Acknowledgements........iii

    Abstract of dissertation..vii

    Table of contents..x

    List of figures....xiii

    List of schemes.xiv

    List of tables..xvi

    List of abbreviations....xvii

    Chapter 1: Introduction to carbohydrates and development of Pd-/B-mediated dual catalyzed

    regio/stereo-selective glycosylation methodology...1

    1.1 Background of carbohydrates..1

    1.2 Classical methods for glycosylation1

    1.3 De Novo approach to carbohydrates and synthesis of Boc-pyranones3

    1.4 De Novo approach using Pd-catalysis towards glycosylation..5

    1.5 The aims...7

    1.6 Development of Pd-/B-mediated dual catalysis towards regio/stereo-selective

    glycosylations8

    1.7 Effect of C-4 substitution on the regioselectivity..13

    1.8 Iterative use of Pd-/B-dual catalysis towards Mannose oligosaccharide structures..15

    1.9 Conclusion.20

    1.10 References21

  • xi

    Chapter 2: First Total Synthesis of Mezzettiaside natural products via the iterative use of a Pd-

    /B-mediated dual catalysis and SAR study22

    2.1 Background of Mezzettiasides...22

    2.2 De Novo synthesis of Boc-pyranone (2.1.1 or 1.2.3).24

    2.3 Retrosynthetic analysis of Mezzettiaside family members24

    2.4 Synthesis of the key intermediate 2.4.2 using a dual Pd-/B-catalysis for the regioselective

    glycosylation26

    2.5 Synthetic approach to mezzettiaside 2-11..28

    2.6 Synthesis of four new disaccharide analogues...35

    2.7 Bioactivity of all 10 mezzettiasides and four new disaccharide analogues towards anti-

    cancer H460 cancer cell line/anti-bacterial strain....37

    2.8 Conclusion.38

    2.9 References..39

    Chapter 3: Synthesis of cardiac glycosides 1,3-linked oligosaccharides via enzymatic like

    regio/stereo-selective glycosylation and SAR study.....41

    3.1 Introduction to Na+/K+-ATPase pump and Cardiac Glycosides (CGs)42

    3.2 Synthesis of monosaccharide digitoxin analogues46

    3.3 Synthesis of 1,3-linked digitoxin analogues (di-/tri-/tetra-saccharide)..48

    3.4 Biological evaluations of 1,3-linked oligosaccharides..52

    3.5 Studies directed towards the synthesis of monosaccharide C4 amino-digitoxin

    analogues....53

    3.6 Carbohydrate as mimic to hydrated ions...55

    3.7 Synthesis of Boc-azide as a glycosyl donor...56

  • xii

    3.8 Regioselective synthesis of disaccharide amino-digitoxin analogues...57

    3.9 Conclusion.59

    3.10 References60

    Chapter 4: Experimental Section..62

    4.1 Chapter 1 Experimental Procedure62

    4.2 Chapter 2 Experimental Procedure73

    4.3 Chapter 3 Experimental Procedure..128

    Appendix: 1H and 13C NMR Spectrum

  • xiii

    List of Figures

    Figure 2.1: Mezzettiaside family of natural products.23

    Figure 3.1: General representation of cardiac Glycosides ..45

    Figure 3.2: Crystal structure of Ouabain, a cardiac glycoside.....46

    Figure 3.3: General representation of digitoxin...........46

    Figure 3.4: Carbohydrate as a mimic to hydrated ions....55

  • xiv

    List of Schemes

    Scheme 1.1: Classical method for O-glycosylation...2

    Scheme 1.2: De Novo synthesis of Boc-pyranones ( / -L/D-Boc-pyranones).4

    Scheme 1.3: Diastereoselective Palladium catalyzed glycosylation through Pd- -allyl

    Complex6

    Scheme 1.4: Post-glycosylation transformations...7

    Scheme 1.5: Tin-oxide mediated Benzylation...8

    Scheme 1.6: General representation of Cleistrio-/Cleistetro-side natural products..9

    Scheme 1.7: Regioselective glycosylation using Taylors catalyst.10

    Scheme 1.8: Regio-/Stereo-selective glycosylation11

    Scheme 1.9: Regio-/Stereo-selective synthesis of disaccharide using Pd-/B-dual catalysis...13

    Scheme 1.10: General representation of effect of C-4 substitution on regioselectivity......14

    Scheme 1.11: Plausible mechanistic representation of bis-glycosylation on Mannose...17

    Scheme 1.12: Bis-glycosylation on methyl- -D-mannose..18

    Scheme 1.13: Iterative Bis-glycosylation on trisaccharide..19

    Scheme 1.14: Utilization of Pd-/B-methodology towards bis-glycosylation..20

    Scheme 2.1: Synthesis of -L-Boc-pyranone..24

    Scheme 2.2: Retrosynthetic analysis of mezzettiaside 2-11....25

    Scheme 2.3: Synthesis of key intermediate 2.3.426

    Scheme 2.4: Synthesis of key intermediate 2.4.227

    Scheme 2.5: Synthesis of mezzettiaside-9 (9).28

    Scheme 2.6: Synthesis of mezzettiaside-10 (10) 29

    Scheme 2.7: Synthesis of mezzettiaside-11 (11).29

  • xv

    Scheme 2.8: Synthetic approach to mezzettiaside-8 (8)..30

    Scheme 2.9: Approach to mezzettiaside-4 (4) ....31

    Scheme 2.10: Approach to mezzettiaside-2 (2)...31

    Scheme 2.11: Synthesis of mezzettiaside-3 (3)...32

    Scheme 2.12: Approach towards mezzettiaside-5 (5) .33

    Scheme 2.13: Synthesis of mezzettiaside-6 (6) and mezzettiaside-7 (7).34

    Scheme 2.14: Approach towards unnatural disaccharide analogues (2.14.1-2.14.2)..36

    Scheme 2.15: Approach towards unnatural disaccharide analogues (2.15.1-2.15.2)..36

    Scheme 3.1: De Novo synthesis of glycosyl donors and -L-/D-Digitoxin

    monosaccharides.47

    Scheme 3.2: Regioselective glycosylation using Pd-/B-catalysis.......49

    Scheme 3.3: Synthesis of trisaccharide-digitoxin analogues...50

    Scheme 3.4: Synthesis of tetrasaccharide-digitoxin analogues...51

    Scheme 3.5: Synthesis of C-4-amino-digitoxin monosaccharide analogues..54

    Scheme 3.6: Synthesis of -L-Boc-azide56

    Scheme 3.7: Synthesis of disaccharide amino-digitoxin analogue..57

    Scheme 3.8: Synthesis of digitoxin disaccharide amino analogue..58

    Scheme 3.9: Synthesis of diamino-digitoxin disaccharide analogue...59

  • xvi

    List of Tables

    Table 1.1: Optimization of regio-/stereo-selective glycosylation....12

    Table 1.2: Effect of C-4 substitution on regioselectivity ....14

    Table 2.1: Optimization of the regio-selectivity of glycosylation...27

    Table 2.2: Anticancer activity ( M) for the isolated mezzettiasides...35

    Table 2.3: Anticancer/antibacterial data for the 10 natural and 4 synthetic disaccharide

    analogues37

    Table 3.1: Anticancer data of digitoxin analogues using H460 cell line.52

    Table 3.2: Anticancer data of monosaccharide C-4 amino-digitoxin analogues.55

  • xvii

    List of Abbreviations

    Ac Acetyl

    Anal. Analysis

    Bn Benzyl

    Boc t-Butoxycarbonyl

    bp Boiling point

    Bu Butyl

    BuLi n-Butyllithium

    Calcd Calculated

    CI Chemical Ionization

    ClAc Chloroacetyl

    CTAB Cetyltrimethylammonium bromide

    d Doublet

    DBA trans, trans-dibenzylideneacetone

    DBU 1,8-Diazabicyclo[5.4.0]undec-7-ene

    de Diastereomeric excess

    DEAD Diethyl azodicarboxylate

    Chemical shift (ppm)

    DMAP 4-Dimethylaminopyridine

    DMSO Dimethyl sulfoxide

    ee Enantiomeric excess

    EI Electron ionization

    ent Enantiomer

  • xviii

    equiv Equivalent(s)

    ESI Electron spray ionization

    Et Ethyl

    EtOAc Ethyl acetate

    g Gram(s)

    h Hour(s)

    HRMS High resolution mass spectrum

    Hz Hertz (cycles per second)

    IR Infrared

    J Spin-spin coupling constant

    mol Mole(s)

    m Multiplet

    Me Methyl

    mg Milligram(s)

    MHz Megahertz

    MIC Minimum inhibitory concentration

    min Minute(s)

    mmol Millimole(s)

    mp Melting point

    MS Mass spectrum

    MTT 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide

    NBS N-Bromosuccinimide

    NBSH o-Nitrobenzenesulfonylhydrazide

  • xix

    NMO N-Methylmorpholine N-oxide

    NMR Nuclear magnetic resonance

    Ph Phenyl

    PMB p-Methoxybenzyl

    Ppm Parts per million

    Py Pyridine

    q Quartet

    Rf Ratio to front

    rt Room temperature

    t Triplet

    THF Tetrahydrofuran

    TLC Thin layer chromatography

    p-TsOH p-toluene sulfonic acid

    TBAI Tetrabutylammonium iodide

  • 1

    Chapter 1

    Introduction to carbohydrates and development of Pd-/B-mediated dual

    catalyzed regio-/stereo-selective glycosylation methodology

    1.1 Background of Carbohydrates

    Carbohydrates containing natural products play an impressive role in the biological processes

    (like cell death, cell-cell recognition, target binding, cell-cell interaction, tissue targeting and

    membrane transportation).1,2 These attracted the synthetic community to study the role of

    carbohydrates in biologically active natural/unnatural products. Although, nature provides

    biologically active carbohydrate motifs but chemists are hampered by the limited number of

    sugar isomers when they try to perform carbohydrate based structure activity relationship (SAR)

    studies. Often times aglycon core of natural products are devoid of activities when the

    carbohydrate motif is removed (e.g., Jadomycin A vs Jadomycin B3 and Digitoxigenin vs

    Digitoxin). Hence the need for alternative carbohydrate synthetic methodologies which

    systematically installs rare natural and unnatural sugars are required to enable drug discovery

    efforts. These highly complex carbohydrate natural products are challenging synthetic targets. A

    practical total synthetic approach toward these natural/unnatural products is one that enables the

    supply of abundant material for SAR studies.

    1.2 Classical methods for glycosylation

    Glycosylation is one of the most meticulously studied reactions in chemistry. The reaction

    is a process in which a nucleophile acts as a glycosyl acceptor and attacks the electrophile to

    form a glycosidic bond in presence of an activator or Lewis acid catalyst.3 The new glycosidic

    bond is formed by the displacement of the leaving group (e.g., -OTf, -Cl, -Br, -I, -OC=(NH)-R, t-

  • 2

    Boc) at the anomeric position of the sugar by a nucleophile, often in the presence of an activator.

    In this process, the nucleophile is referred as glycosyl acceptor, whereas, the incoming group

    acts as a glycosyl donor (electrophile).

    The most typically used glycosylation methods are the Koenigs-Knorr glycosylation and

    Schmidt glycosylation. The Koenigs-Knorr glycosylation4,5 involves the nucleophilic

    displacement of halide (chloride/bromide as leaving group) at the anomeric position in presence

    of promoter. Whereas, the Schmidt glycosylation6 displaces a trichloroacetamidate. The

    generally accepted mechanism involves the ionization of the leaving group at the anomeric

    center followed by a nucleophilic addition.

    Scheme 1.1: Classical method for O-glycosylation

    Using traditional glycosylation methodologies, the nucleophile can attack from two

    possible faces: top ( ) or bottom ( ) face (Scheme 1.1). If the nucleophile attacks from the top

    face, a -isomer is formed, whereas, if the nucleophile attacks from the bottom face, the -

    isomer is obtained. The traditional glycosylation is a powerful methodology but does suffer from

    the following drawbacks:

    a) It often produces a mixture of anomers ( / ) in varying ratios thus affecting the yield (Scheme

    1.1).

    b) It often requires stoichiometric amount of promoter/catalyst (e.g., TMSOTf, TfOH, AgOTf).

  • 3

    c) It requires protection/deprotection manipulation which increases total number of steps.

    d) It is sensitive to moisture, temperature, solvent dependent and highly relies upon the structure

    of donor and acceptor.

    In order to overcome these problems, there is a continuing need to develop new

    glycosylation methodology which will address the concerns and rely on catalytic amount of

    activator. In this regards, the ODoherty lab has developed a Pd-catalyzed glycosylation

    methodology under mild reaction conditions thus maintaining the good anomeric selectivity

    (Section 1.4).

    1.3 De novo Achmatowicz approach to carbohydrates and synthesis of Boc-pyranones

    In contrast to the classical glycosylation methods where carbohydrates containing starting

    materials being used for the synthesis of carbohydrate structures, the ODoherty group used a de

    novo asymmetric approach to build carbohydrate motifs. This de novo approach to sugars installs

    the desired functionality and chirality from achiral moieties (e.g., 2-acetylfuran 1). The

    uniqueness of this approach comes from the fact that all the stereocenters in the molecule are

    built onto the achiral framework of 2-acetylfuran 1. We call this concept a de novo asymmetric

    approach towards sugars. This approach relies on a highly diastereoselective palladium(0)-

    catalyzed glycosylation in combination with asymmetric catalysis to control the anomeric

    stereochemistry.7

    The de novo asymmetric approach begins with the synthesis of the key glycosyl donors

    (Boc-pyranones) using Noyori reduction and Achmatowicz oxidative rearrangement. The

    synthesis of Boc-pyranone (1.2.3, 1.2.4, (ent)-1.2.3 and (ent)-1.2.4) starts with achiral 2-

    acetylfuran 1, which underwent an enantioselective Noyori asymmetric hydrogenation using the

  • 4

    Noyori catalyst (Noyori (S,S) or Noyori (R,R)) and sodium formate (hydrogen source). This

    highly scalable procedure provided the corresponding chiral furanols (1.2.1 and ent-1.2.1) with

    excellent enantiocontrol. The furan alcohols (1.2.1/ent-1.2.1) underwent Achmatowicz oxidative

    rearrangement8 upon exposure to aqueous NBS conditions (NBS/NaOAc/H2O) to provide an

    equilibrating mixture of hemiacetals 1.2.2/ent-1.2.2. Hemiacetal 1.2.2 with the C-5 L-

    configuration was protected with Boc2O/DMAP ( 78 C) to provide a separable mixture of -L-

    Boc-pyranone 1.2.3 and -L-Boc-pyranone 1.2.4 in ~3:1 ratio. Alternatively, when hemiacetal

    ent-1.2.2 was heated to 80 C with Boc2O/DMAP in benzene, -D-/ -D- oc-pyranone (ent-

    1.2.3/ent-1.2.4) were obtained in 1:1 ratio (Scheme 1.2).7

    Scheme 1.2: De Novo synthesis of Boc-pyranones ( / -L/D-Boc-pyranones)

    Thus in just a three step sequence, achiral starting material 1 was converted into any possible

    stereoisomers of the Boc-pyranones. This practical procedure provides the four stereoisomers on

    multigram scale. Once the / are separated, the stereochemistry can be transformed with the

    complete control using our Pd-chemistry. In turn, this stereochemistry is used to install the

  • 5

    remaining stereocenters with the post glycosylation reaction (Luche reduction (NaBH4/CeCl3)

    and Upjohn dihydroxylation (OsO4/NMO)) to provide an acceptor (i.e., a glycosyl donor was

    converted to glycosyl acceptor using post-glycosylation transformation).

    1.4 De Novo approach using Pd-catalysis towards glycosylation

    The ODohertys Pd-catalyzed steroselective glycosylation has been used for the synthesis

    of natural/unnatural carbohydrates motif(s) with interesting biological activity. This Pd-catalyzed

    reaction is highly stereospecific (i.e., -pyranones results in -anomeric products) and occurs

    under mild reaction conditions (i.e., 0 C) in the presence of catalytic amount (1-5 mol%) of

    Pd2(dba)3CHCl3/4PPh3 as a catalyst. The advantage of this Pd-catalyzed methodology is: a) its

    compatibility with wide variety of solvents (e.g., CH2Cl2, THF, DMF, CH3CN); b) its reliance on

    a catalytic amount of Pd-catalyst and c) its predictable stereochemistry.

    The reaction proceeds through a Pd- -allyl intermediate complex which serves as the

    glycosyl donor and reacts with an alcohol nucleophile (glycosyl acceptor) with net retention of

    stereochemistry. Thus the incoming nucleophile retains the anomeric stereochemistry of the

    starting material, (i.e., double inversion of stereochemistry, Scheme 1.3). This is seen as

    advantageous over traditional glycosylation reactions which often provide mixture of products at

    the anomeric position.7

  • 6

    Scheme 1.3: Diastereoselective Palladium catalyzed glycosylation through Pd- -allyl complex This Pd-catalyzed glycosylation can be applied towards a wide range of alcohol nucleophiles

    under mild reaction condition [Pd2(dba)3CHCl3/4PPh3 (2.5 mol%)] with decent yields. The

    enone portion of the Boc-pyranone not only serves as an anomeric directing group during Pd(0)

    catalyzed glycosylation, it also serves as a triol protecting group (i.e., masked enone as an atom-

    less protecting group). That is to suggest that the post-glycosylation transformations function as

    triol deprotection steps, for example, reduction of the enone 1.4.1 under Luche conditions

    (NaBH4/CeCl3) gave C-4 allylic alcohol 1.4.2. Then the resulting allylic alcohol 1.4.2 was

    subsequently dihydroxylated using the Upjohn condition (OsO4/NMO) to form a manno-triol

    1.4.3a (X = -OH). Similarly, when a diimide (NBSH/Et3N) reduction was performed on the

    double bond, amicetose sugar 1.4.3b (X = -H) was obtained. The allylic alcohol 1.4.2 can act as

    an acceptor and can be expanded to prepare series of 1,4-linked motifs using Pd-catalyzed

    glycosylation with Boc-pyranone 1.2.3, thus providing disaccharide 1.4.5 after post

    glycosylation transformations (Scheme 1.4).

  • 7

    Scheme 1.4: Post-glycosylation transformations

    1.5 The aims

    The initial aim of my PhD studies was to develop a general methodology for regio-/stereo-

    selective Pd-catalyzed glycosylation of manno-1,2 diols at the equatorial C-3 position. In this

    regards, we have developed a Pd-/B-dual catalyzed glycosylation methodology for the regio-

    /stereo-selective glycosylations of 1,2 and 1,3 diols. An important feature of this methodology is

    how it can enables the minimal use of protecting groups and in some applications protecting

    group free synthesis of natural/unnatural oligosaccharide structures. The applications of this Pd-

    /B-dual catalyzed methodology have been successfully applied towards the following synthetic

    applications:

    One pot regio/stereo-selective bis-glycosylation of mannose related oligosaccharides.

    First total synthesis of mezzettiaside 2-11, anti-cancer natural products using divergent

    approach (see Chapter 2).

  • 8

    Protecting group free synthesis of digitoxin oligosaccharide and amino analogues

    towards cancer studies (see Chapter 3).

    1.6 Development of Pd-/B-mediated dual catalysis towards regio-/stereo-selective

    glycosylations

    The regio-/stereo-selective functionalization of a single hydroxyl group in presence of other

    unmasked hydroxyl group(s) in the carbohydrate motif is seen as a challenge as it is the key for

    building oligosaccharide with the minimal use of protecting groups. Even when enzymatic

    approaches are available, there is no assurance that the enzymatic glycosylation will be general

    (e.g., able to transfer a / -D-/L-sugar to its corresponding / -D-/L products). Previous

    regioselective studies on carbohydrate mainly focused on organotin mediated regioselective

    glycosylation using stoichiometric dibutyltin oxide (Bu2SnO), tributyltin oxide (Bu3Sn)2O and

    dibutyltin dimethoxide (Bu2Sn(OMe)2) (Scheme 1.5).

    Scheme 1.5: Tin-oxide mediated Benzylation

    The problems associated with the tin chemistry were its toxicity, requirement for stoichiometric

    amount, absolute need for anhydrous conditions (Dean-Stark) and long reaction time. Most

  • 9

    problematically, in our hands these reactions suffered from poor reproducibility in terms of

    regio-selectivity and yields. Other problems associated with tin chemistry are the requirement for

    a two steps procedure, in which the sugar (glycosyl acceptor 1.6.1) is first reacted with tin oxide

    in methanol or benzene/toluene to form an intermediate tin-acetal 1.6.2. Then intermediate 1.6.2

    reacts with the corresponding electrophile Boc-pyranone 1.2.3 to give the glycosylated product

    in presence of palladium catalyst. For instance, our Pd-catalyzed regioselective glycosylation

    between 1.6.2 and 1.2.3 yielded a mixture of C-3:C-2 (7:1) products. This particular reaction was

    the representative of the issue of poor reproducibility (Scheme 1.6).9

    Scheme 1.6: General representation of Cleitrio/Cleistetro-side natural product

    While progress was made towards the regioselective acylation/tosylation/alkylation (Prof.

    Taylors work) of the carbohydrate motifs10, the regioselective glycosylation of the unprotected

    carbohydrates to form a specific linkage by using tin-chemistry has been still an unsolved

    problem. In this regards, the scope for the development of new methodology for stereo- and

  • 10

    regio-selective glycosylation that does not rely on tin chemistry was seen as a potential major

    improvement.

    To address the issues related to regio-/stereo-selectivity, reproducibility and yield in the

    glycosylation reaction, there was a strong desire from the synthetic carbohydrate community to

    develop alternative methodology which could use a catalytic method for the regio-/stereo-

    selective glycosylation of polyol sugars. The most advantageous part of this endeavor would be

    the avoidance of unnecessary protection/deprotection steps.

    In this regard, Prof. Taylor recently developed a borinate ester (Ph2BOCH2CH2NH2)

    catalyst that promoted the regioselective acylation/tosylation/alkylation/glycosylation. The

    Taylors catalyst (Ph2BOCH2CH2NH2) via intermediate of a cyclic boronate can direct the

    incoming group to the equatorial C-3 of an unprotected rhamnose, mannose and pyranose sugars,

    using 1.1 equiv. of Ag2O for glycosylation (Scheme 1.7).10

    Scheme 1.7: Regioselective glycosylation using Taylors catalyst

  • 11

    Inspired by this approach, we were interested in developing a general methodology which not

    only provides the desired regioselectivity, but also imparts / -stereoselectivity. This in turn

    allows libraries of biologically active carbohydrates structures to be synthesized in fewer steps

    with complete control of regio/stereo-selectivity. Working towards this aim, we have developed

    a new methodology which utilizes a combination of nucleophilic boron and electrophilic

    palladium(0) catalysis to install the incoming glycosyl donor selectively to get an equatorial C-3

    and/or C-6 hydroxy of a pyranose with complete control of -stereochemistry. Discussed in this

    Chapter is the development of Pd-/B-mediated dual catalyzed glycosylation methodology, in

    which the nucleophilic boron imparts the regioselectivity whereas, the electrophilic Pd controls

    the -selectivity.

    With the above strategy in mind, we started exploring our initial attempts for the

    regioselective glycosylation on the C-4 substituted rhamnose motif 1.8.1 (glycosyl acceptor)

    with Boc-pyranone (glycosyl donor 1.2.3). Initially, rhamnose 1.8.1 underwent regio/stereo-

    selective Pd-catalyzed glycosylation in presence of tin oxide (Bu2SnO, 1.1 eqvi.) to give an

    inseparable mixture of C-3:C-2 (5:1) glycosylated product (1.8.2 and 1.8.3).

    Scheme 1.8: Regio/Stereo-selective glycosylation

  • 12

    Inspiring by this problem, we investigated the use of a Pd-/B-dual catalyzed glycosylation. The

    Taylor catalyst ultimately led to the regio-/stereo-selective C-3 glycosylation in good yields of

    C-4 substituted sugars. We began our studies with the C-4 substituted rhamnose 1.8.1. When diol

    1.8.1 was reacted with 15 mol% borinate catalyst and glycosyl donor 1.2.3 (see Chapter 2, for

    application to the mezzettiasides) using CH2Cl2 as a reaction solvent, a regiomeric ratio for

    1.8.2:1.8.3 of ~2.5:1 was observed (Table 1.1).

    Table 1.1: Optimization of regio/stereo-selective glycosylation

    Reducing the amount of borinate catalyst to 10 mol% in CH3CN/CH2Cl2 had no significant effect

    on the regiomeric ratio. Whereas, when 15 mol% catalyst was used in CH3CN/THF (1:0.1), the

    regiomeric ratio improved two fold (i.e., 6:1). Finally, the glycosylation condition was optimized

    to 30 mol% borinate catalyst in CH3CN/THF (1:0.1) which provided the best regioselectivity

    ratio (7.5:1) in decent yield (77%) (Table 1.1).11

    The plausible mechanism for the Pd-/B-mediated dual catalyzed glycosylation is shown in

    Scheme 1.9. Initially, the diol forms an anionic tetra-coordinated borinate intermediate 1.8.1a

  • 13

    whereas, the Boc-pyranone forms a cationic palladium -allyl complex. From the observations,

    we can conclude that the reaction solvent plays a critical role in regioselectivity, as shown in

    Table 1.1.

    Scheme 1.9: Regio/Stereo-selective synthesis of disaccharide using Pd-/B-dual catalysis

    1.7 Effect of C-4 substitution on the regio-selectivity

    With the successful development of the iterative Pd-/B-mediated dual catalysis

    methodology, we explored a range of C-4 substituted sugars to gauge its effect on the

    regioselectivity. When rhamnose 1.10.4 was used as starting material (i.e., C-4 = -OH), the

    pyranone donor was regio/stereo-selectively delivered exclusively at the C-3 position of

    rhamnose (Entry 1, Table 1.2) to provide 1.10.5. Whereas, when C-4 was changed to an acetoxy

    group 1.10.6, the ratio of the incoming group being transferred to the C-3 vs C-2 was 15:1 (Entry

    2). In addition, when the steric at C-4 was further increased to a hexanoate (i.e., C-4 = -

    OC(O)(CH2)4CH3, 1.8.1), the ratio dropped to 7.5:1 (Entry 3). Finally, when C-4 was changed to

    an azide (i.e, C-4 = -N3), the C-3/C-2 ratio was found to be 16:1 (Entry 4). From these

    observations we concluded that both steric and electron withdrawing group at C-4 affects the

  • 14

    regioselectivity, electron withdrawing groups at C-4 reduces the selectivity. Smaller the

    substituent at C-4 being better for the regioselectivity (Table 1.2)

    Scheme 1.10: General representation of effect of C-4 substitution on regioselectivity

    Table 1.2: Effect of C-4 substitution on regioselectivity

    With the successful development of an iterative Pd-/B-mediated dual catalysis methodology

    towards regio- and stereo-selective glycosylation, we wanted to expand its application towards

    the synthesis of biologically active natural/unnatural compounds having range of sugar units

  • 15

    (mono- to oligo-saccharides).

    We were also fascinated at the possibility of using this methodology to exploit an SAR

    study of a biologically active natural products. In this regard, we have successfully used this

    methodology for the first total synthesis of the mezzettiasides.11,12 The mezzettiasides are a

    family of anti-cancer natural products (details will be discussed in Chapter 2). Finally Chapter 3

    talks about the use of Pd-/B-dual catalysis towards the protecting group free synthesis of

    digitoxin oligosaccharide analogues and its amino sugar analogues.

    Presented in this chapter, is the use of this Pd-/B-dual catalyzed regio/stereo-selective

    glycosylation in the iterative bis-glycosylation of mannose to form oligosaccharide structures. In

    this regards, we initially started to explore the glycosylation reactions between methyl- -D-

    mannose 1.11.1 and Boc-pyranone (ent)-1.2.3. The glycosyl acceptor (methyl- -D-mannose

    1.11.1) underwent bis-glycosylation using the Pd-/B-dual catalysis to form trisaccharide enone

    1.11.3 (Scheme 1.12). The most interesting feature of this methodology is that, in just 6 steps

    starting material methyl- -D-mannose was efficiently converted to a fully functionalized

    pentasaccharide motif.

    1.8 Iterative use of Pd-/B-dual catalysis towards Mannose oligosaccharide structures

    Carbohydrates not only act as energy storage but also play key roles in number of

    biological processes (e.g., viral/bacterial infections, angiogenesis, tumor cell metastasis,

    inflammation, immune response).13 Due to this vast scope of carbohydrates in biology, it has

    become an emerging subject in drug discovery.

    Working towards this goal, we planned to synthesize mannose related oligosaccharide

    motifs to uncover the potential applications of our above mentioned Pd-/B-catalyzed

  • 16

    glycosylation methodology towards the synthesis of oligosaccharide motifs. Of the

    oligosaccharides, the oligomannose containing motifs has gotten most attention as oligomannose

    related structures are ubiquitously present on the surface of the mammalian cells and are

    involved in processes such as protein quality control, cell-cell communication, cell-cell

    differentiation, viral infection and tumor migration.13

    Herein, we report regioselective and stereoselective Pd-/B-dual catalysis strategy for the

    protecting group free synthesis of 3,6-linked mannose trisaccharides. The approach started with

    the bis-glycosylation of methyl- -D-mannopyranoside 1.11.1 and -pyranose (glycosyl donor,

    ent-1.2.3). Initially it forms a six membered cyclic intermediate 1.11.1a (Scheme 1.11) which

    reacts with the palladium -allyl complex at C-6 position to give a C-6 glycosylated product

    1.11.2. Subsequently it then forms a five membered ring tetra-coordinated boron intermediate

    1.11.2a which then react regioselectively to install the -D-Boc-pyranone (glycosyl donor)

    equatorially at the C-3 position 1.11.3. Thus in a net one pot reaction 1.11.1 is converted into

    trisaccharide 1.11.3 cleanly in a 68% yield (Scheme 1.11). The most important feature of this

    iterative bis-glycosylation is that no protecting groups were required for the synthesis of

    oligosaccharides. A schematic representation of bis-glycosylated oligosaccharide is shown in

    Scheme 1.11.

  • 17

    Scheme 1.11: Plausible mechanistic representation of bis-glycosylation on Mannose

    The synthesis of oligosaccharide motifs starts from commercially available methyl- -D-

    mannopyranoside 1.11.1 which upon treatment with borinate catalyst (35 mol%) and

    Pd2(dba)3CHCl3/4PPh3 catalyst (5 mol%) underwent iterative regio/stereo-selective bis-

    glycosylation using -D-Boc-pyranone ent-1.2.3 to provide trisaccharide dienone 1.11.3 (68%).

    Dienone 1.11.3 was reduced (NaBH4/CeCl3) to corresponding bis-allylic alcohol 1.12.1 in 90%

    yield. Alcohol 1.12.1 was dihydroxylated using OsO4/NMO to provide fully functionalized

    trisaccharide compound 1.12.2 (79%) (Scheme 1.12).

  • 18

    Scheme 1.12: Bis-glycosylation of methyl- -D-mannose

    Building upon this success we were intrigued to see if we could explore Pd-/B-dual catalysis on

    the trisaccharide moiety 1.12.2 at the C-3/C-3 hydroxyl positions respectively to form a

    pentasaccharide motif without using any protecting group. This was accomplished by bis-

    glycosylating trisaccharide motif 1.12.2 with -D-Boc-pyranone ent-1.2.3 regoiselectively at C-

    3/C-3 positions respectively with full control of -selectivity to give pentasaccharide dienone

    1.13.1. Dienone 1.13.1 underwent a Luche reduction (NaBH4/CeCl3) followed by Upjohn

    dihyroxylation (OsO4/NMO)) to form pentasaccharide 1.13.2 (Scheme 1.13).

  • 19

    Scheme 1.13: Iterative Bis-glycosylation on trisaccharide

    Finally, we have shown that we can successfully regio/stereo-selectively glycosylate in

    one pot using the Pd-/B-dual catalysis. The importance of this bis-glycosylation comes from the

    fact that pentasaccharide motif 1.13.2 can be prepared in just 6 steps, without the use of any

    protecting group(s). In order to demonstrate the stereochemical variability, we glycosylated

    methyl- -D-mannose 1.11.1 with -L-Boc-pyranone 1.2.3 to obtain trisaccharide enone 1.14.1,

    which further underwent post-glycosylation transformations (NaBH4/CeCl3 and OsO4/NMO)

    providing fully functionalized trisaccharide moiety 1.14.2 (Scheme 1.14).

    Scheme 1.14: Utilization of Pd-/B-methodology towards bis-glycosylation

  • 20

    1.9 Conclusion

    In summary, we have successfully developed a de novo methodology using nucleophilic

    boron and electrophilic palladium which can be used towards regioselective installation of sugars

    with complete retention of anomeric stereochemistry. Utilization of this method was shown in

    the iterative bis-glycosylation of manno-pyranose motif to form mannose related oligosaccharide

    structures. The most important feature of the Pd-/B-dual catalyzed reaction is the lack of

    protecting groups. For instance, the synthesis of the pentasaccharide motif only required 6 steps.

    Thus the synthesis is highly efficient and affordable toward carbohydrate motifs for medicinal

    chemistry studies.

    Future studies focusing on utilizing galactose (as an acceptor) and cyclitols/carbasugar

    (as donors) are under investigation in the ODoherty group. Applications of this developed

    methodology towards different carbohydrate structures can be seen in the total synthesis of

    mezzettiaside natural products using minimal protecting group and in the protecting group free

    synthesis of digitoxin oligosaccharide analogues as discussed in Chapter 2 and 3 respectively.

  • 21

    1.10 References

    1. For useful reviews see: (a) P. R. Crocker, T. Feizi, Curr. Opin. Struct. Biol., 1996, 6, 679; (b) E. Herbert, Biosci. Rep., 2000, 20, 213.

    2. For useful reviews see: (a) H. Lis, N. Sharon, Chem. Rev., 1998, 98, 637; (b) S. Houlton,

    Chem. Br., 2002, 38, 46. 3. (a) L. Wang, R. L. White, L. C. Vining, Microbiology, 2002, 148, 1091; (b) M. Shimizu, H.

    Togo, M. Yokoyama, Synthesis, 1998, 799. 4. W. Koenig, E. Knorr, Ber., 1901, 34, 957.

    5. H. Paulsen, Angew. Chem. Int. Ed. Engl., 1982, 21, 155; (b) G-J. Boons, Carbohydrate Chemistry; Blackie Publishers; London, 1998; (c) S. H. Khan, R. A. ONeill, Modern Methods in Carbohydrates Synthesis; Harwood Academic: Amsterdam, 1996; (d) K. C. Nicolaou, H. J. Mitchell, Angew. Chem. Int. Ed., 2001, 40, 1576; (e) D. P. Galoni, D. Y. Gin, Nature, 2007, 446, 1000.

    6. R. R. Schmidt, J. Michel, Angew. Chem. Int. Ed. Engl., 1980, 19, 731; (b) R. R. Schmidt,

    Angew. Chem. Int. Ed. Engl., 1986, 25, 212.

    7. (a) R. S. Babu, G. A. ODoherty, J. Am. Chem. Soc. 2003, 125, 12406; (b) Concurrent with these studies was the similar discovery by Feringa, Lee; A. C. Comely, R. Eelkema, A. J. Minnaard, B. L. Feringa, J. Am. Chem. Soc., 2003, 125, 8714; (c) H. Kim, H. Men, C. Lee, J. Am. Chem. Soc., 2004, 126, 1336; (d) The poor reactivity in Pd-catalyzed allylation reaction of alcohols as well as a nice solution to this problem was reported, see: H. Kim, C. Lee, Org. Lett., 2002, 4, 4369; (e) For a related Rh system, see: P. A. Evans, L. J. Kennedy, Org. Lett., 2000, 2, 2213; (f) S. O. Bajaj, J. R. Farnsworth, G. A. ODoherty, Org. Synth., 2014, 91, 338.

    8. O. Achmatowicz, R. Bielski, Carbohydr. Res., 1977, 55, 165.

    9. B. Wu; M. Li, G. A. ODoherty, Org. Lett., 2010, 12(23), 5466.

    10. (a) D. Lee, M. S Taylor, J. Am. Chem. Soc., 2011, 133, 3724; (b) C. Gouliaras, D. Lee, L. Chan; M. S. Taylor, J. Am. Chem. Soc., 2011, 133, 13926; (c) C. A. McClary, M. S. Taylor, Carbohydr. Res., 2013, 381, 112; (d) E. Dimitrijevic, M. S. Taylor, Chem. Sci., 2013, 4, 3298; (e) T. M. Beale, M. S. Taylor, Org. Lett., 2013, 15, 1358.

    11. S. O. Bajaj, E. U. Sharif, N. G. Akhmedov, G. A. ODoherty, Chem. Sci., 2014, 5, 2230. 12. S. O. Bajaj, P. Shi, P. J. Beuning and G. A. ODoherty, Med. Chem. Comm., 2014, 5, 1138.

  • 22

    Chapter 2

    First total synthesis of Mezzettiaside natural products via the iterative use of a

    Pd-/B-mediated dual Catalysis and SAR study

    2.1 Background of Mezzettiasides

    The zeal for natural products with interesting biological properties, led to the discovery of

    Mezzettiasides, a class of partially acetylated oligosaccharide natural products which are known

    to be active against cancer cell lines, particularly towards Lu1, Col2 and KB (5 to > 20 M).1,2

    This family of oligorhamnoside was isolated from the fruit and stem bark of Mezzettia leptopoda

    in Malaysia in 1986.1 The important feature of this class of anti-cancer natural products is that,

    all the carbohydrate units have unique acylation pattern and are 1,3-linked oligorhamnose motifs.

    This unique acylation pattern makes them a challenging target for synthesis. Mezzettiaside

    family members are divided into three subclasses: disaccharide (Mezzettiaside 9-11),

    trisaccharide (Mezzettiaside 2-4 & 8) and tetrasaccharide (Mezzettiaside 5-7) (Fig. 2.1).

    As part of a program aimed at the synthesis and biological investigation of

    anticancer/antibiotic oligosaccharide natural products,3 we became intrigued in the synthesis of

    mezzettiasides. The interest came out from our synthesis and study of a related class of natural

    products, the cleistriosides and cleistetrosides3, where we found that the pattern of acylation had

    a significant effect on the biological activity. As a continuation of these studies, we desired

    access to all the ten members of the mezzettiaside natural products.3,4 While re-isolation of this

    material from its various sources was considered, we rather planned to synthesize all the

    mezzettiasides using our de novo asymmetric approach.

    Thus, we envisioned the synthesis of all the mezzettiasides using asymmetric divergent

    approach utilizing Pd-catalyzed glycosylation in combination with the nucleophilic boron (Pd-

  • 23

    /B-) dual catalysis methodology as discussed in Chapter 1.5 The divergent approach allowed us

    the use of minimal protecting group (i.e., only one protecting group (AcCl) was used in the

    synthesis of 9 natural products, whereas for the 10th member uses AcCl twice). Herein, we

    disclose our successful de novo asymmetric approach to all the ten members of the mezzettiaside

    where all the stereocenters were derived from achiral starting material 2-acetyl furan 1. Figure

    2.1 shows all the 10 mezzettiasides natural products.

    Figure 2.1: Mezzettiasides family of natural products

    (Contributions: Sumit O. Bajaj performed all the synthetic work presented here under the

    guidance of Prof. ODoherty and Dr. Sharif. Dr. Akhmedov helped with the structure analysis of

    some final molecules using detailed NMR studies. The biological evaluation was performed by

    Dr. Shi under the guidance of Prof. ODoherty and Prof. Beuning).

  • 24

    Along with the first total synthesis of this family of natural products, we have also

    planned to synthesize four new disaccharides analogues (2.14.1-2.14.2 and 2.15.1-2.15.2) with

    different (R1 = n-Pr, i-Pr, t-Bu, i-Bu) acylation pattern. These products enabled structure activity

    relationship (SAR) studies6 where the importance of the sugar chain length and acylation pattern

    at various positions of the sugar moiety on the biological activity was investigated.

    2.2 De Novo synthesis of Boc-pyranone (2.1.1 or 1.2.3)

    The synthesis of Boc-pyranone ( -L) 2.1.1 from inexpensive achiral starting material 2-

    acetyl furan 1 is outlined in Scheme 2.1. Pyranone 2.1.1 is a key building block for all the

    mezzettiaside family of natural products. The uniqueness of this de novo route is that we can

    install the stereochemistry in the sugar products from achiral material 2-acetyl furan 1, using

    asymmetric catalysis. This concept is referred to as a de novo approach to carbohydrates.7

    Boc-pyranone 2.1.1 could be easily prepared in three steps (Noyori (S,S) enantioselective

    asymmetric hydrogenation9, Achmatowicz oxidative rearrangement (NBS/NaOAc/H2O) and

    Boc-protection (Boc2)O) from achiral 2-acetyl furan 1 (Chapter 1). In the Pd-catalyzed

    glycosylation reaction, the Boc-pyranone 2.1.1 or 1.2.3 acts as a glycosyl donor and the reacting

    alcohol is the glycosyl acceptor.7,8

    Scheme 2.1: Synthesis of -L-Boc-pyranone

    2.3 Retrosynthetic analysis of Mezzettiaside family members

    The retrosynthetic route we devised for the mezzettiaside natural products is outlined in

  • 25

    Scheme 2.2. We envisioned that the three tetrasaccharides (mezzettiasdie 5-7) and the four

    trisaccharides (mezzettiaside 2-4 & 8) could be obtained from a common pyranone trisaccharide

    intermediate 2.8.1. In turn, intermediate 2.8.1 as well as the final three disaccharides

    (mezzettiaside-9-11) could be prepared from a disaccharide allylic alcohol. Finally, allylic

    alcohol 2.4.2 could be obtained from ester diol 2.3.4, whereas, diol 2.3.4 can be made from Boc-

    protected pyranone 2.1.1 in 4 steps (Scheme 2.2).

    Scheme 2.2: Retrosynthetic analysis of mezzettiaside 2-11

    We planned to synthesize all the mezzettiaside using divergent approach. Key to the

    success of this approach is the strategic use of enone atomless/minimal protecting groups (enone

    of a pyranone as a masked triol and the minimal use of chloroacetate as the only other protecting

    group) in combination with the iterative use of a highly regio- and stereo-selective B-/Pd-

    catalyzed glycosylation makes this route more interesting.

  • 26

    2.4 Synthesis of the key intermediate 2.4.2 using a dual Pd-/B-catalysis for the regio-

    selective glycosylation

    The synthesis of key intermediate 2.4.2 began with 2-acetyl furan 1. In a 3 steps sequence

    acetyl furan 1 (Noyori reduction, Achmatowicz reaction9 and Boc-protection) was converted to

    Boc-pyranone 2.1.1. Pyranone 2.1.1 in a palladium catalyzed glycosylation with octan-1-ol

    provided enone 2.3.1. Enone 2.3.1 was reduced under Luche reduction condition (NaBH4/CeCl3)

    followed by esterification (hexanoic acid, DCC/DMAP) provided C-4 hexanoate motif 2.3.3.

    Finally Upjohn syn-dihydroxylation (OsO4/NMO)10 provided diol 2.3.4 (Scheme 2.3).

    Scheme 2.3: Synthesis of key intermediate 2.3.4

    The C-4 hexanoate diol 2.3.4 was studied in the regioselective glycosylation reaction at O-

    3 hydroxy group with stoichiometric amount of tin catalyst (Bu2SnO).3 The main drawback of

    the tin chemistry was the requirement for stoichiometric amount. Also, the reproducibility in

    regio-isomeric ratio and yield was the hardest part to optimize. As discussed in Chapter 1, we

    have developed a new methodology based on catalytic borinate ester to control the

    regiochemistry in the glycosylation process. Our optimized condition for the glycosylation

    (Entry 2, Table 2.1) uses 30 mol% borinate catalyst (Ph2BOCH2CH2NH2)11,12 and 2.5%

    palladium catalyst to provide a mixture of C3:C2 (2.3.5/2.3.6) in 7.5:1 ratio.

  • 27

    .

    Table 2.1: Optimization of the regio-selectivity of glycosylation

    The glycosylated mixture 2.3.5/2.3.6 was chloroacetylated ((ClAc)2O/Py) to provide 2.4.1 and its

    C-2 regiomer followed by Luche reduction (NaBH4/CeCl3) to provide desired pure allylic

    alcohol 2.4.2 in excellent yield (77%, 2 steps) (Scheme 2.4).

    Scheme 2.4: Synthesis of key interemediate 2.4.2

  • 28

    2.5 Synthetic approach to mezzettiaside 2-11

    The key intermediate 2.4.2 was transformed into disaccharides mezzettiaside 9-11 using a

    divergent route. In order to obtain mezzettiaside-9, disaccharide allylic alcohol 2.4.2 was

    chloroacetylated ((ClAc)2O/Py) at the C-4 position, followed by dihydroxylation to give diol

    2.5.1 (77%, 2 steps). Peracetylation of 2.5.1 (Ac2O/DMAP) then removal of both chloroacetates

    (thiourea/NaHCO3/n-Bu4NI)13 provided mezzettiaside-9 (9) (83%, 2 steps) (Scheme 2.5).

    Scheme 2.5: Synthesis of mezzettiaside-9 (9)

    In a similar fashion, disaccharide allylic alcohol 2.4.2 was acetylated (Ac2O/DMAP), followed

    by dihydroxylation (OsO4/NMO) to give C-4 acetate diol 2.6.1 (80%, 2 steps). Diol 2.6.1

    underwent treatment with (CH3C(OEt)3/p-TsOH/AcOH) to regioselectively install an axial C-2

    acetate in alcohol 2.6.2 (85%). Alcohol 2.6.2 upon chloroacetate deprotection

    (thiourea/NaHCO3/n-Bu4NI) furnished mezzettiaside-10 (10) (86%) (Scheme 2.6).

  • 29

    Scheme 2.6: Synthesis of mezzettiaside-10 (10)

    Allylic alcohol 2.4.2 was dihydroxylated under Upjohn condition (OsO4/NMO), to provide

    a triol which upon peracetylation (Ac2O/DMAP) gave triacetate 2.7.1 (81%, 2 steps). Finally

    deprotection of the chloroacetate group (thiourea/NaHCO3/n-Bu4NI) gave the desired

    mezzettiaside-11 (11) (82%) (Scheme 2.7).

    Scheme 2.7: Synthesis of mezzettiaside 11(11)

    As mentioned earlier, this route is divergent so the disaccharide intermediate 2.6.2 was

    utilized towards the synthesis of trisaccharides mezzettiaside 2-4 and 8 (Scheme 2.8-2.11).

    Alcohol 2.6.2 upon treatment with Boc-pyranone 2.1.1 underwent a Pd-catalyzed glycosylation

    to give trisaccharide enone 2.8.1 (68%) in presence of Pd2(dba)3CHCl3/4PPh3. Enone 2.8.1 upon

  • 30

    post-glycosylation transformations provided trisaccharide triol 2.8.2 (79%, 2 steps). Triol 2.8.2

    was then regioselectively C-2 acetylated ((CH3C(OEt)3/p-TsOH/AcOH) to give trisaccharide

    diol 2.8.3 (79%). Diol 2.8.3 upon subsequent chloroacetate deprotection provided mezzettiaside-

    8 (8) (85%) (Scheme 2.8).

    Scheme 2.8: Synthetic approach to mezzettiaside-8 (8)

    In order to obtain mezzettiaside-4, trisaccharide enone 2.8.1 was reduced (NaBH4/CeCl3)

    and acetylated (Ac2O/Py) to give allylic acetate 2.9.1 (71%, 2 steps). Allylic acetate 2.9.1 was

    dihydroxylated (OsO4/NMO) and deprotected (CH3C(OEt)3/p-TsOH/AcOH) to give

    mezzettiaside-4 (4) (70%, 2 steps) (Scheme 2.9).

  • 31

    Scheme 2.9: Approach to mezzettiaside-4 (4)

    Allylic acetate 2.9.1 upon syn-dihydroxylation (OsO4/NMO) gave diol 2.10.1 (85%).

    Exposure of diol 2.10.1 to the orthoester mediated C-2 acylation ((CH3C(OEt)3/p-TsOH/AcOH))

    gave alcohol 2.10.2 (84%). Removal of chloroacetate group (thiourea/NaHCO3/TBAI)13 from

    alcohol 2.10.2 gave mezzettiaside-2 (2) (77%) (Scheme 2.10).

    Scheme 2.10: Approach to mezzettiaside-2 (2)

    To regioselectively install a C-3 acetate on diol 2.10.1, the Taylor type boron mediated

    acylation (Ph2BOCH2CH2NH2/AcCl) was utilized providing intermediate C-2 alcohol 2.11.1

    (71%).11 Chloroacetate deprotection (thiourea/NaHCO3/TBAI) of alcohol 2.11.1 provided

    mezzettiaside-3 (3) (82%) (Scheme 2.11).

  • 32

    Scheme 2.11: Synthesis of mezzettiaside-3 (3)

    Similarly, tetrasaccharides mezzettiaside 5-7 were prepared from trisaccharide intermediate

    2.10.1. Intermediate diol 2.10.1 underwent a Pd-/B-dual catalyzed C-3 regioselective

    glycosylation (Ph2BOCH2CH2NH2/Pd(PPh3)2) with Boc-pyranone 2.1.1 to provide

    tetrasaccharide enone 2.12.1 (76%) with exclusively -stereochemistry. Enone 2.12.1 upon

    subsequent post-glycosylation transformations (NaBH4/CeCl3 then OsO4/NMO) gave triol 2.12.2

    (73%, 2 steps). Triol 2.12.2 upon C-3 regioselective acetylation (Ph2BOCH2CH2NH2,

    AcCl/DIPEA)11 and chloroacetate deprotection (thiourea/NaHCO3/TBAI) provided

    mezzettiaside-5 (5) in good yield (58%, 2 steps) (Scheme 2.12).

  • 33

    Scheme 2.12: Approach towards mezzettiaside-5 (5)

    Finally, the synthesis of the last two tetrasaccharides mezzettiaside 6-7 began with intermediate

    2.12.2, which upon chloroacetate deprotection gave mezzettiaside-7 (7) (83%). Whereas,

    exposure of intermediate 2.12.2 to the regioselective C-2 acylation (CH3C(OEt)3/p-TsOH) gave

    mezzettiaside-6 (6) after deprotection (thiourea/NaHCO3/TBAI) of the chloroacetate group

    (61%, 2 steps) (Scheme 2.13).

  • 34

    Scheme 2.13: Synthesis of mezzettiaside-6 (6) and mezzettiaside-7 (7)

    With all the mezzettiasides 2-11 in hand, we next wanted to explore the effect of the various

    acylation patterns on anticancer and antibacterial activities. In addition we also wanted to explore

    analogues of the simplest disaccharide mezzettiasides 9-11, as these offered the greatest

    opportunity for structural modification. The reported activities for Mezzettiaside 9-11 showed a

    great dependence on the degree of acylation. To address this question we decided to make C-3

    ester analogues of mezzettiaside-10. The anticancer data reported for the isolated material

    [disaccharide (mezzettiaside 9-11) and trisaccharide (mezzettiaside 2-4 & 8)] were found to have

    the broadest range of activity against the lung/colon (Lu1, Col2, Table 2.2) cancer cell lines. We

    decided to use the NCI H460 non-small cell lung cancer cell line for our evaluation. For

    cleistetrosides/cleistriosides, we found the greatest antibacterial activity was seen with the

    B.subtilis strain, so we decided to use this strain for MIC screening of antibacterial activity.

    Cell Line M-2 M-3 M-4 M-8 M-9 M-10 M-11

    Lu1 8.6 11.8 19.4 19.7 5.4 >20 6.1

    Col2 4.3 4.9 6.2 8.2 >20 >20 9.0

    KB 6.2 14.3 15.4 >20 11.3 >20 12.7

    aResults are expressed as ED50 values ( g/mL) bLu1 = human lung cancer; Co12 = human colon cancer; KB = human oral epidermoid carcinoma

  • 35

    Table 2.2: Anticancer activity ( M) for the isolated mezzettiasides

    We performed an MTT anti-cancer assay on NCI-H460 cancer cell line, which confirmed that

    among the disaccharides/trisaccharides/tetrasaccharides, trisaccharides mezzettiaside 2-4 & 8

    were more active than disaccharides 9-11 and tetrasaccharides 5-7. Initially, we were concerned

    about the stability of the acetate groups towards ester hydrolysis or 1,2 migration and no

    evidence of this has been observed.

    Among the three types of mezzettiasides (di-, tri- and tetra-saccharides) a strong

    dependency upon the degree and location of acylation on activity can be seen. To further explore

    this effect, we decided to install the bulkier group (n-butyrate, i-butyrate, i-valerate and pivalate)

    on the disaccharide motif at C-3 position of the intermediate 2.6.2. These more hindered esters

    which might be stable towards hydrolysis and then compared the effect of acylation vs

    glycosylation on the disaccharide 2.6.2 at C-3 position. Working towards this, we planned to

    synthesize four new disaccharide unnatural analogues and investigated their SAR studies.

    2.6 Synthesis of four new disaccharide analogues

    The synthesis of new analogues started with disaccharide alcohol intermediate 2.6.2.

    Disaccharide 2.6.2 upon esterification with n-butanoic acid/DCC/DMAP provided C-3 butanoate

    which upon chloroacetate group deprotection (thiourea/NaHCO3/Bu4NI) gave disaccharide

    2.14.1 (70%, 2 steps). In a similar fashion, intermediate 2.6.2 upon treatment with pivaloyl

    chloride/Py. underwent acylation followed by deprotection (thiourea/NaHCO3/Bu4NI) of the

    chloroacetate to provide disaccharide analogue 2.14.2 (58%, 2 steps) (Scheme 2.14).

  • 36

    Scheme 2.14: Approach towards unnatural disaccharide analogues 2.14.1-2.14.2

    Along these lines, intermediate 2.6.2 was esterified with iso-butyryl chloride/pyridine and the

    chloroacetate was deprotected (thiourea/NaHCO3/Bu4NI) yielding disaccharide 2.15.1 (68%, 2

    steps). Similarly when 2.6.2 was treated with iso-valeric acid in presence of DCC/DMAP, it

    underwent esterification to provide disaccharide analogue 2.15.2 (58%, 2 steps) (Scheme 2.15).6

    Scheme 2.15: Approach towards unnatural disaccharide analogues 2.15.1-2.15.2

    2.7 Bioactivity of all 10 mezzettiasides and four new disaccharide analogues towards anti-

    cancer H460 cancer cell line/anti-bacterial strain

    All 10 synthetically prepared natural compounds and 4 new disaccharide analogues were

    tested for cytotoxicity against cancer cell lines and bacterial strains. These results from these

    studies are outlined in Table 2.3. Disaccharide mezzettiaside-11 and trisaccharides mezzettiaside

  • 37

    2-4 and 8 were found to be the most cytotoxic towards H460 cell. Whereas, mezzettiaside-4, 8

    and 9 were most cytotoxic against the B.Subtilis strain.6 The synthetic unnatural disaccharide

    analogues 2.14.1-2.14.2 and 2.15.1-2.15.2 were synthesized to compare the effect of

    glycosylation (mezzettiaside 2-4 and 8) to acylation. These acetylated products had reduced

    activities which led us to conclude that installing sugar at C-3 of the disaccharide was more

    effective than acylation (2.14.1-2.14.2 and 2.15.1-2.15.2).

    a All the MIC ( M) are an average of at least three independent experiments bAll the IC50 ( M) are an average of at least three independent experiments

    Table 2.3: Anti-cancer/anti-bacterial data for the 10 natural and 4 synthetic disaccharide (2.14.1-2.14.2 and 2.15.1-2.15.2) analogues

    R R1 R2 MICa ( M)

    IC50b( M)

    B.subtilis H460 Mezze-2 Ac H Ac >128 17.0 Mezze-3 Ac Ac H >128 10.0 Mezze-4 Ac H H 8 9.0 Mezze-8 H H Ac 16 24.4 Mezze-5 H Ac H >128 40.0 Mezze-6 H H Ac 64 259.6 Mezze-7 H H H 32 17.7

    R R1 MICa ( M)

    IC50b ( M)

    B.subtilis H460 Mezze-9 H Ac 16 151.4

    Mezze-10 Ac H >128 >500 Mezze-11 Ac Ac >128 15.2

    2.14.1 Ac n-PrCO >128 46.6 2.14.2 Ac t-BuCO >128 141.1 2.15.1 Ac i-PrCO >128 32.0 2.15.2 Ac i-BuCO >128 79.3

  • 38

    2.8 Conclusion

    In conclusion, we have shown the utility of our highly divergent de novo Pd-/B-dual

    catalyzed approach towards the first total synthesis of mezzettiaside family of natural products.

    In addition, this synthetic approach allowed the synthesis of the entire family of mezzettiaside

    (10 members). The synthesis concluded the regio/stereo-selectively installation of the sugars at

    the C-3 position to form 1,3-linked oligo-rhamnoside natural products. The key feature of this

    synthesis is the use of a divergent approach to synthesize all 10 natural products. The synthesis

    occured in the range of 13-22 longest linear steps and yet only 42 total steps were required for

    the synthesis of all members. In combination with the natural products mezzettiaside 2-11, four

    new disaccharide analogues 2.14.1-2.14.2 and 2.15.1-2.15.2 were successfully synthesized with

    different pattern of acylation (Ac/n-Pr/i-Pr/t-Bu) to compare the effect of glycosylation vs

    acylation.

    All fourteen molecules (Mezze-2-11, 2.14.1-2.14.2 and 2.15.1-2.15.2) were tested against

    NCI H460 cancer cell lines and B. Subtilis bacterial strain to confirm that sugar chain length,

    substitution at various positions varies the biological activity. Finally, this SAR study uncovered

    the anti-cancer data for the tetrasaccharide mezzettiaside 5-7. The synthesis makes use of

    inexpensive, commercially available achiral starting material (2-acetyl furan 1) and builds on

    chirality using a de novo approach. Overall 10-20 chiral centers were built and masked enone

    was used as an atom-less protecting group. In order to build all the 10 members of the natural

    product library, only one protecting group (AcCl) was used. The synthesis was amenable for the

    preparation of enough quantities for the biological testing against cancer-cell line and bacterial

    strains.

  • 39

    2.9. References

    1. (a) J. T Etse, A. I. Gray, C. Lavaud, G. Massiot, J-M. Nuzillard and P. G. Waterman, J. Chem. Soc., Perkin Trans. 1, 1991, 861; (b) D. A. Powell, W. S. York, H. V. Halbeek, J. T. Etse, A. I. Gray and P. G. Waterman, Can. J. Chem., 1990, 68, 1044. 2. B. Cui, H. Chai, T. Santisuk, V. Reutrakul, N. R. Farnsworth, G. A. Cordell, J. M. Pezzuto, A. D. Kinghorn, J. Nat. Prod., 1998, 61, 1535. 3. B. Wu, M. Li, G. A. ODoherty, Org. Lett., 2010, 12 (23), 5466; (b) V. Seidel, F. Bailleul, P. G. Waterman, J. Nat. Prod., 2000, 63, 6. 4. P. Shi, M. C. Silva, H. L. Wang, N. G. Akhmedov, M. Li, P. J. Beuning, G. A. ODoherty, ACS Med. Chem. Lett., 2012, 3, 1086. 5. (a) S. O. Bajaj, E. U. Sharif, N. G. Akhmedov, G. A. ODoherty, Chem. Sci., 2014, 5, 2230; (b) S. Y. Ko, A. W. Lee, S. Masamune, L. A. Reed, K. B. Sharpless, Science, 1983, 20, 949; (c) A. B. Northrup, D. W. C. MacMillan, Science, 2004, 305, 1752; (d) Md. M. Ahmed, B. P. Berry, T. J. Hunter, D. J. Tomcik, G. A. O'Doherty, Org. Lett., 2005, 7, 745; (e) J. M. Harris, M. D. Keranen, H. Nguyen, V. G. Young, G. A. O'Doherty, Carbohydr. Res., 2000, 328, 17; (f) J. M. Harris, M. D. Keranen, G. A. O'Doherty, J. Org. Chem., 1999, 64, 2982. 6. S. O. Bajaj, P. Shi, P. J. Beuning, G. A. ODoherty, Med. Chem. Comm., 2014, 5, 1138.

    7. (a) M. Li, J. G. Scott, G. A. O'Doherty, Tetrahedron Lett., 2004, 45, 1005; (b) H. Guo, G. A. O'Doherty, Org. Lett., 2005, 7, 3921. 8. S. O. Bajaj, J. R. Farnsworth, G. A. ODoherty, Org. Synth., 2014, 91, 338.

    9. (a) O. Achmatowicz, P. Bukowski, B. Szechner, Z. Zwierzchowska, A. Zamojski, Tetrahedron, 1971, 27, 1973; (b) A. R. Noyori, T. Ohkuma, M. Kitamura, J. Am. Chem. Soc., 1987, 109, 5856.

    10. In general, we have found the NaBH4 (with and without CeCl3) reduction of -pyranones and the OsO4 catalyzed dihydroxylation of the resulting allylic alcohol to proceed with virtually complete diastereocontrol, see ref. 4, 6, 7 and 10. 11. (a) D. Lee, M. S Taylor, J. Am. Chem. Soc., 2011, 133, 3724; (b) C. Gouliaras, D. Lee, L. Chan, M. S. Taylor, J. Am. Chem. Soc., 2011, 133, 13926; (c) C. A. McClary, M. S. Taylor, Carbohydr. Res., 2013, 381, 112; (d) E. Dimitrijevic, M. S. Taylor, Chem. Sci., 2013, 4, 3298; (e) T. M. Beale, M. S. Taylor, Org. Lett., 2013, 15, 1358. 12. While the Taylor catalyst has been used in regioselective glycosylations (glycosylbromide with excess Ag2O (>1.0 equiv.), ref. 12), this is the first example of its use to catalytically induce regiocontrol in a catalyzed glycosylation.

  • 40

    13. (a) M. H. Clausen, R. Madsen, Chem.-Eur. J., 2003, 9, 3821; (b) M. Naruto, K. Ohno, N. Takeuchi, Tetrahedron Lett., 1979, 251. S. O. Bajaj, E. U. Sharif, N. G. Akhmedov, G. A. ODoherty, Chem. Sci., 2014, 5, 2230. Reproduced by permission of The Royal Society of Chemistry (www.rsc.org/follow). S. O. Bajaj, P. Shi, P. J. Beuning, G. A. ODoherty, Med. Chem. Comm., 2014, 5, 1138. Reproduced by permission of The Royal Society of Chemistry (www.rsc.org/follow).

  • 41

    Chapter 3

    Synthesis of cardiac glycosides 1,3-linked oligosaccharides via enzymatic like

    regio/-stereo-selective glycosylation and SAR study

    Synthetic chemistry has always been challenged by natures ability to assemble natural

    products. This challenge comes from natures seemingly limitless ability to make complex

    structures with specific biological properties. Nature accomplishes this synthetic feat by

    designing enzymes that selectively zip these structures together without the formation of

    byproducts or the use of protecting groups. Nowhere is this synthetic efficiency more pronounces

    than in oligosaccharide biosynthesis, where glycosyltranferase enzymes transfer the desired

    sugar regio- and stereo-selectively onto a polyol containing carbohydrate without the need for

    protecting groups. To do this the glycosyltransferase utilizes two distinct binding sites, one to

    selectively recognize and activate the nucleophilic component (glycosyl acceptor) and the other

    to selectively recognize and activate the electrophilic component (glycosyl donor). Both these

    enzyme binding sites require great structural complexity in order to ensure substrate selectivity,

    which is compensated by the catalysts large turnover number. It is worth noting that this

    substrate selectivity perforces an inherant substrate inflexibility when trying to use a given

    glycosyltransferase for the transfer of unnatural substrates.

    In an effort to match natures prowess at using enzymes to assemble oligosaccharide motifs

    without the use of protecting groups, we have been exploring the use of asymmetric catalysis for

    the protecting group free synthesis of oligosaccharide structures. This alternative approach offers

    several advantages over biosynthetic approaches, in terms of the range of substrate

  • 42

    stereochemistry that can be added and the divergent synthetic direction the glycosylated product

    can be taken. In this regard, we have shown that our de novo approach to carbohydrates could be

    used to construct stereochemically diverse libraries of oligosaccharides containing D- and L-

    sugar.1,2 For example, in only seven steps all D-sugar, all L-sugar or mixed D-/L-sugar

    trisaccharide with rhamno- or amiceto-substitution/stereochemistry can be assembled without the

    use of protecting groups.1,2 This approach, which solely focuses on the use of Pd- -allyl catalysis

    to activate the glycosyl donor, relys on the glycosyl acceptor to contain a single or singlely

    reactive alcohol. Thus to more truly mimic glycosyltranferase like catalysis, an additional

    component, which regioselectively activates the glycosyl acceptor, needs to be added to this

    process. To do this we turned to borinate chemistry, which has been shown to regioselectively

    activate 1,2- and 1,3-diols (Chapter 1). More specifically, the selective functionalization of the 3-

    and 6-positions of manno- and galacto-pyranose sugars. Herein, we describe the use of this

    approach for the synthesis and SAR-study of the oligosaccharide portion of the cardiac

    glycosides as anticancer agents.

    3.1 Introduction to Na+/K+-ATPase pump and Cardiac Glycosides (CGs)

    Na+/K+-ATPase are known to regulate several vital cellular functions, like homeostasis,

    cell volume (ions inside and outside the cells), signal transducer and membrane potential.3,4,5 The

    pump is important for the movement of ions across cell membranes (e.g., muscle contraction)

    and also for creating charge imbalances across the cell membranes (e.g., electrical impulses).3,4,5

    The pumping process begins with the binding of two extracellular K+, three intracellular Na+ ions

    and a molecule of ATP. Once all the ions are bound ATP is converted into ADP and inorganic

    phosphate. The ion exchange occurs across the cell membrane, with the energy for the process

  • 43

    coming from ATP hydrolysis.6,7 The Na+/K+-ATPase is divided into two subunits and .8 The

    and subunits are known to exist in various isoforms (i.e., 1, 2, 3, 4 and 1, 2, 3),

    the expression of which is associated with various tissue types.9 For instance, the 1 isoform is

    present ubiquitously,whereas, the 2 is expressed mostly in the heart muscle and plays a key role

    in the maintenance of blood pressure and cardiac function.10

    Cardiac glycosides are known for inhibiting Na+/K+-ATPase and increase intracellular

    sodium ions. The Na+/K+-ATPase is a P-type pump that actively transports two K+ inside and

    three Na+ outside, because of this the intracellular sodium levels are low, which helps to sustain

    an adequate electrochemical gradient across the plasma membrane. Electrochemical gradients

    are necessary for proper cellular function.3,4,5 In addition to the Na+/K+-pump, the Na+/Ca2+

    exchange pump extrudes Na+ ions out of the cell and introduces Ca2+ ions into the cell.11 In heart

    muscle cells, the inhibition of the Na+/K+-ATPase leads to modulation of heart contractions,

    whereas, in cancer cells this leads to apoptotic induced cell death.12,13,14

    Recent reports suggest that some cancers have highly expressed 115,16,17,18,19 and/or

    320,21,22 isoforms and the selective binding to these isoforms may lead to a better cancer

    therapy.23,24,25 By targeting the 1 and 3 isoform selectively, one can inhibit cancer cell growth

    and reduce the off target toxicity. More recently, we and others have shown that cardiac

    glycosides with improved anticancer activity can be found by modifying the carbohydrate

    portion of the molecule.12 Specifically, we prepared 1,4-linked mono-, di- and tri-saccharide

    cardiac glycosides with varying substituion and stereochemistry and screened for anticancer

    cytotoxicity (Scheme 3.1).26 In general, these studies have found that improved anti-cancer

    activity can be found by changing the substitution and stereochemistry of the sugars (e.g., -L-

  • 44

    rhamno- and -L-amiceto-). In addition, we found that reducing the length of the trisaccharide to

    a monosaccharide significantly increases the anticancer activity. In fact, a similar dependance of

    chain length can be found when one looks at the cardiotonic activity of the cardiac glycosides.

    Taken as a whole, one can conclude from the evidence that natures choice of a trisaccharide is

    not to increase activity but rather increase the selectivity for the heart muscle tissue.

    On the basis of crystal structure of the Na+/K+-ATPase bound to ouabain (Fig. 3.2), a

    related cardiac glycoside, it could be seen that cardiac glycosides binds to the -subunit of

    Na+/K+-ATPase. The crystal structure of ouabain also shows that there is little or no opportunity

    for SAR-substituion of the aglycon region whereas, the solvent exposed cone shaped region

    where the carbohydrate resides is an ideal space for SAR-type exploration. Following natures

    lead, we chose to modify the carbohydrate subunits of digitoxin (a component of digitalis) to

    explore this space and to find new -1 and/or -3-selective Na+/K+-ATPase inhibitor as

    anticancer agents. We chose to use a synthesis led SAR-study to map out this extracellular

    carbohydrate binding site, which is circumscribed by the structurally ill-defined-extracellular

    loops27 of the Na+/K+-ATPase. This large solvent exposed pocket that allows for the exchange of

    Na+ and K+ ions is flexible enough to fit a wide range of polar structures, like sugars. As this

    space is still chiral, it should response differently to the addition of a D- vs L-sugar to a given

    ligand untill the growing oligosaccharide chain is free from the protein environment.

    Cardiac glycosides structures (digitoxin, digoxin and ouabain) are divided into two

    portions: the aglycon (steroid core) having an unsaturated lactone at C17 position and the

    carbohydrate motif (sugar) attached to the core at the C3 position (Fig. 3.1).28 Of particular

    interest to us was digitoxin, a cardiac glycoside used in the treatment of congestive heart failure,

    arrthymia by enhancing the cardiac contraction.29 Digitoxin was isolated from the flower of

  • 45

    digitalis purpurea, (aka, foxglove) which has long been known for its inotropic effect (increases

    strength of muscular contraction). Despite the narrow therapeutic window, digitoxin is still in

    clinical use around the world.30

    Figure 3.1. General representation of Cardiac Glycosides

    Our previous efforts to explore this space with oligosaccharides led to the synthesis and

    evaluation of several 1,4-linked di- and tri-saccharide structures, which all had significantly

    reduced anti-cancer activities. Having found no success with 1,4-linked oligosaccharide, we

    decided to explore 1,3-linked oligosaccharide. Herein, we describe the use of our dual Pd-/B-

    catalyzed glycosylation for the exploration of the carbohydrate binding region of the Na+/K+-

    ATPase. The ultimate aim of this project is to identify novel carbohydrate motifs that can

    selectively target Na+/K+-ATPase, which in turn can be used as anticancer agents. The ideal

    anticancer drug will not only be effective but also selective against tumor cells.

  • 46

    Figure 3.2: Crystal Structure of Ouabain, a cardiac glycoside

    The general representation of digitoxin, a cardiac glycoside is shown in Fig. 3.3, where the

    steroid core (digitoxigenin) is a pharmacophore and the carbohydrate portion controls the

    isoform selectivity.

    Figure 3.3. General representation of digitoxin

    3.2 Synthesis of monosaccharide digitoxin analogues

    From the initial studies by Dr. Hua-Yu Leo Wang on the cardiac glycosides, he

    concluded that L-sugars with rhamno-3.1.3a and amiceto-3.1.3b substitution gave the most

  • 47

    active analogues.13 The synthesis of these monosaccharide began with the preparation of the D-

    /L-Boc-pyranones (glycosyl-donors) from achiral furan. Specifically, 2-acetyl furan 1 underwent

    a three step procedure (Noyori reduction, Achmatowicz rearrangement and Boc-protection) to

    form the D- and L-Boc-pyranones 3.1.4 and 3.1.1 respectively.

    The Boc-pyranones 3.1.1/3.1.4 can be further treated with digitoxigenin under Pd-

    catalyzed glycosylation to provide enones, which were further reduced to allylic alcohols

    3.1.2/3.1.5 using Luche reduction condition (NaBH4/CeCl3). Allylic alcohols 3.1.2/3.1.5 were

    dihydroxylated (OsO4/NMO) to provide 3.1.3a/3.1.6a or reduced using o-nitrobenzenesulfonyl

    hydrazine (NBSH) reduction to give 3.1.3b/3.1.6b (Scheme 3.1). The four monosaccharides

    were screened for their cyctotoxicity against H460 cancer cell lines (Table 1). This data indicated

    that the -L-monosaccharides were better than its corresponding -D analogues.26

    IC50 (nM)X =

    Mono--OH H

    -L-Sugar 35 39-D-Sugar 706 387

    Scheme 3.1: De Novo synthesis of glycosyl donors and -L-/D-Digitoxin monosaccharides

  • 48

    We next decided to explore the effects of further carbohydrate substitution at the C-3

    position of the -L-rhamnose sugar on the most active monosaccharide 3.1.3a. To do this, we

    envisioned the addition of either -L- or D-sugar with either rhamno- or amiceto-substitution

    (3.2.3a/3.2.6a or 3.2.3b/3.2.6b). This stereodivergent substituion served as stereochemical

    control element to ensure the space being occupied is still under the influence of the extracellular

    loops of the protein.

    3.3 Synthesis of 1,3-linked digitoxin analogues (di-/tri-/tetra-saccharide)

    To install the D- and L- sugars we turned our attention towards the use of the dual Pd-/B-

    catalyzed glycosylation (Scheme 3.2). The synthesis of disaccharides starts with the

    monosaccharide rhamno-digitoxin 3.1.3a. Monosaccharide 3.1.3a upon exposure to our

    regioselective Pd-/B-dual catalysis glycosylation methodology reacted with the incoming donor

    (Boc-pyranones 3.1.1/3.1.4) selectively at C-3 equatorial alcohol position with -selectivity to

    obtain corresponding enones 3.2.1/3.2.4. The enones 3.2.1/3.2.4 were further reduced under

    NaBH4/CeCl3 condition to give disaccharide allylic alcohols 3.2.2/3.2.5 respectively. An Upjohn

    dihydroxylation (OsO4/NMO) provided 3.2.3a/3.2.6a. Alternatively, a diimide reduction using

    NBSH was used to give 3.2.3b/3.2.6b (Scheme 3.2).

  • 49

    Scheme 3.2: Regioselective glycosylation using Pd-/B-catalysis

    The four di-saccharides were screened for their cyctotoxicity against the human non-

    small cell lung cancer cell line, H460 (Table 3.1). These results showed that only the bis-L-

    rhamno-disaccharide 3.2.3a retained its potency (3.2.3a: IC50 = 39 nM). The chiral nature of the

    space was indicated by the ne