Disclaimer - Inha저작자표시 2.0 대한민국 이용자는 아래의 조건을 따르는...

165
저작자표시 2.0 대한민국 이용자는 아래의 조건을 따르는 경우에 한하여 자유롭게 l 이 저작물을 복제, 배포, 전송, 전시, 공연 및 방송할 수 있습니다. l 이차적 저작물을 작성할 수 있습니다. l 이 저작물을 영리 목적으로 이용할 수 있습니다. 다음과 같은 조건을 따라야 합니다: l 귀하는, 이 저작물의 재이용이나 배포의 경우, 이 저작물에 적용된 이용허락조건 을 명확하게 나타내어야 합니다. l 저작권자로부터 별도의 허가를 받으면 이러한 조건들은 적용되지 않습니다. 저작권법에 따른 이용자의 권리는 위의 내용에 의하여 영향을 받지 않습니다. 이것은 이용허락규약 ( Legal Code) 을 이해하기 쉽게 요약한 것입니다. Disclaimer 저작자표시. 귀하는 원저작자를 표시하여야 합니다.

Transcript of Disclaimer - Inha저작자표시 2.0 대한민국 이용자는 아래의 조건을 따르는...

  • 저작자표시 2.0 대한민국

    이용자는 아래의 조건을 따르는 경우에 한하여 자유롭게

    l 이 저작물을 복제, 배포, 전송, 전시, 공연 및 방송할 수 있습니다. l 이차적 저작물을 작성할 수 있습니다. l 이 저작물을 영리 목적으로 이용할 수 있습니다.

    다음과 같은 조건을 따라야 합니다:

    l 귀하는, 이 저작물의 재이용이나 배포의 경우, 이 저작물에 적용된 이용허락조건을 명확하게 나타내어야 합니다.

    l 저작권자로부터 별도의 허가를 받으면 이러한 조건들은 적용되지 않습니다.

    저작권법에 따른 이용자의 권리는 위의 내용에 의하여 영향을 받지 않습니다.

    이것은 이용허락규약(Legal Code)을 이해하기 쉽게 요약한 것입니다.

    Disclaimer

    저작자표시. 귀하는 원저작자를 표시하여야 합니다.

    http://creativecommons.org/licenses/by/2.0/kr/legalcodehttp://creativecommons.org/licenses/by/2.0/kr/

  • 理學博士學位 請求論文

    A Dissertation for Doctor of Philosophy

    아이속사졸린 유도체들의 PTP1B 억제 및 비만 억제효과

    와 4,4'-메틸렌비스(5-(퓨란-2-일)-2-하이드록시벤조산)의 생

    물학적 평가

    Isoxazolone Derivatives as PTP1B Inhibitors with Anti-

    Obesity Effects and the Biological Evaluation of 4,4'-

    Methylenebis(5-(furan-2-yl)-2-hydroxybenzoic acid).

    2011 년 8 월

    仁荷大學校 大學院 化學科 (化學專攻)

    Department of Chemistry, Graduate School

    Inha University

    Bhooshan Kafle

  • 理學博士學位 請求論文

    A Dissertation for Doctor of Philosophy

    아이속사졸린 유도체들의 PTP1B 억제 및 비만 억제효과

    와 4,4'-메틸렌비스(5-(퓨란-2-일)-2-하이드록시벤조산)의 생

    물학적 평가

    Isoxazolone Derivatives as PTP1B Inhibitors with Anti-

    Obesity Effects and the Biological Evaluation of 4,4'-

    Methylenebis(5-(furan-2-yl)-2-hydroxybenzoic acid).

    2011 년 8 월

    指導敎授 趙 亨 鎭

    이 論文을 博士學位 論文으로 提出함

    仁荷大學校 大學院 化學科 (化學專攻)

    Department of Chemistry, Graduate School

    Inha University

    Bhooshan Kafle

  • Dedication

    There is elixir in knowledge.

    This dissertation is dedicated to my father late Netra Prashad Kafle

    (Masters in Sanskrit grammar, Sampurnananda University, Varanasi, India, a

    high school teacher of Sanskrit grammar). He started his formal education

    when he was 22. He bunked several lunch and dinner in his student life due

    to lack of money but did not surrender with poverty and continued his

    education. He was a very good teacher. Even in vacation he was surrounded

    with students at home. He rarely enjoyed his vacation. He was physically

    disabled. But mentally, he was abnormally strong.

    Buwa (father), the god did not let you to wait till my graduation but I hope

    your soul is able to see this dissertation.

  • i

    ACKNOWLEDGEMENTS

    I would like to extend my heartfelt gratitude to Prof. Dr. Hyeongjin Cho, who trained,

    encouraged and challenged me throughout my Ph.D. He never accepted less than my best

    efforts. In fact, whatever the results I obtained during my Ph.D. and written in this

    dissertation are creations of Professor Hyeongjin Cho. Beside the science he also trained me

    for the ultimate condition and made me ultimate survivor. Where ever I will that doesn’t

    matter. I will strive to imply the knowledge that I learned with him. Thank you Prof. Cho.

    I would like to acknowledge to my thesis reviewers, Prof. Lee Keun Hyeung, Prof. Soh Jae-

    Won Prof. Choi Eunmi, and Prof. Kim Taehyun for constructive and excellent advices after

    detailed review of this dissertation.

    I am deeply indebted to my senior Dr. Bharat Raj Bhattarai who introduced me in this lab

    and trained me to play with chemicals, enzymes, bacteria and animals. I always missed him

    during every hard time that I faced due to social and other circumstances.

    I owe my sincerest thanks to Dr. Suja Shreshtha, Dr. Chuda Raj Lohani, Lok Nath Neupane,

    Mandira Neupane, Dr Ranjan KC, Dr Suravi Baral, Dr Devi Basnet, Prabha Basnet, Dr Shila

    Maskey, Dr Mrigendra Bir Karmacharya, Binika Hada, Sher Bahadur Rawal, Basanti Rawal,

    Thakur Prasad Subedi, Deegendra Khadka, Dr Nilkanth G.Aher, Dr Thirupati, Dr Vivek

    Thapa, Lee Jun and other Nepalese and Indian friends.

    A special thanks to my Korean friends Kim Joung Min, Choi Unchal, Yang Mi Hwa, Jang

    Su Jang , Mrs. You, Ms Lee, Miso Park, Mr lee and other Korean friends.

    Most especially to my mother Na Kareshwari Kafle, my son Aagam Kafle, my brother

    Alankar Kafle and my wife Anita Kafle, my brother in law & sisters Durga & Bani Guragain,

    Bhim & Benu Khatiwada and Om & Karuna Wasti, my father in law and mother in law

    Kashinath & Mina Baral. Words alone cannot express what I owe them for their

    encouragement and whose patient love enabled me to complete this dissertation.

    I want to acknowledge all the professors and staffs of Inha University for everything, Jung–

    Seok International Scholarship, Brain Korea (BK 21) program and KRF of the Ministry of

    Education, Government of Korea for providing financial supports during my doctoral

    studies and researches. I am indebted to Institute of Engineering, Tribhuban University for

    providing study leave.

    Finally, no one is perfect and alone in this world; this graduate dissertation was not possible

    without a supporting cast. It’s impossible to mention everyone’s name, so if you are missing,

    please forgive me. It is not intentional.

  • ii

    CONTENTS

    Acknowledgement i

    List of figures ix

    List of tables xii

    List of schemes xiii

    List of abbreviations xiv

    Abstract

    In English xvii

    In Korean xviii

    TABLE OF CONTENTS

    1. General Introduction 1

    1.1 Phosphorylation and dephosphorylation 1

    1.2 Phosphatases 1

    1.3 Importance of phosphatase 2

    1.4 Protein Tyrosin Phosphatase 1B 3

    1.5 Landmarks in PTP1B research 3

    1.6 General mechanism of dephosphorylation of substrate proteins by

    PTP1B 5

    1.7 Validation of PTP1B as a therapeutic target 6

    1.8 Some important class of PTP1B inhibitors found in literature 7

    1.8.1 Vanadate (VO4-3

    ) as a non-specific phosphatase inhibitor 7

    1.8.2 DFMP pTyr mimetics 8

    1.8.3 Nonhydrolyzable pTyr mimetics 9

    1.8.3.1 Nonhydrolyzable DFMP pTyr mimetics 9

  • iii

    1.8.3.2 Carboxylic acid pTyr mimetics 11

    1.8.3.3 TDZ pTyr mimetics 13

    1.8.3.4 Novel pTyr mimetics 15

    1.9 PTP1B inhibitors reported from our lab 16

    1.10 Present work 23

    1.11 Objectives of this study 24

    2.

    Synthesis and Biological Evaluation of Isoxazol-5(4H)-one

    Derivatives as a New Family of PTP1B Inhibitors with Anti-

    Obesity Effects

    25

    2.1 Introduction 25

    2.2 Result and discussion 27

    2.2.1 Synthesis of lead compound (Z)-4-(2-(2-chlorobenzyloxy)

    benzylidene)-3-phenylisoxazol-5(4H)-one (L4)

    28

    2.2.2 Synthesis of a small library of compounds L1-L6 29

    2.2.2.1 Synthesis of aldehyde precursors (A & K-O) for small library

    compounds 29

    2.2.2.2 Precursors for small library compounds 29

    2.2.2.3 Synthesis of small library compounds 30

    2.2.2.4 Small library compounds 31

    2.2.3 In vitro results of small library compounds 31

    2.2.4 Chemical library synthesis 32

    2.2.4.1 Synthesis of seven isoxazol-5(4H)-one precursors for chemical

    library synthesis

    32

  • iv

    2.2.4.2 Synthesis of aldehyde precursor (A-F and H) for chemical library

    synthesis 33

    2.2.4.3 Synthesis of aldehydes precursors (G) for chemical library synthesis 34

    2.2.4.4 Commercially available aldehydes precursor (I and J) for Chemical

    Library Synthesis

    35

    2.2.4.5 Chemical library synthesis 37

    2.2.4.6 Solubility determination of precursors for chemical library synthesis 37

    2.2.4.7 Synthesis chemical library compounds 38

    2.2.4.8 Nature of chemical library compounds 39

    2.2.5 In vitro results of library compounds 42

    2.2.5.1 Inhibitory potency of library compounds against PTP1B 42

    2.2.5.2 Selectivity of most potent compound C3 against other PTPs 43

    2.2.5.3 Enzyme kinetic experiments 44

    2.2.6 Animal experiment 46

    2.2.6.1 Toxicity test of compounds C3 on mice 46

    2.2.6.1.1 Animal and diet selection for toxicity 46

    2.2.6.1.2 Feeding 46

    2.2.6.1.3 Drug dose, gavages preparation and administration 47

    2.2.6.1.4 Effect of compounds C3 on (SIC: ICR) mice 47

    2.2.6.1.5 Carcass of the mice at 6 days of C3 treatment 49

    2.2.6.1.6 Conclusion 51

    2.2.6.2 Effect of compounds C3 on diet induced diabetic mice 51

    2.2.6.2.1 External appearance and body weight gain of mice after obesity

    development period

    51

  • v

    2.2.6.2.2 Body weight at 4 weeks of C3 treatment 52

    2.2.6.2.3 Body weight gain during 4 weeks of C3 treatment 53

    2.2.6.2.4 Food intake 54

    2.2.6.2.5 Feed efficiency 56

    2.2.6.2.6 Intraperitoneal glucose tolerance test (IPGTT) 56

    2.2.6.2.7 Carcass and liver appearance at 4 weeks C3 treatment 58

    2.2.6.2.8 Liver, kidneys and lungs weights 59

    2.2.6.2.9 Epididymal and retroperitoneal fat pad weights 60

    2.2.6.2.10 Blood analysis 61

    2.2.7 Chemical sub-library I compounds 63

    2.2.7.1 Synthesized chemical sub-library I compounds 64

    2.2.7.2 Inhibitory potency of chemical sub-library I compounds against

    PTP1B

    65

    2.2.8 Chemical sub-library II 65

    2.2.8.1 Chemical sub-library II compounds 65

    2.2.8.2 Inhibitory potency of chemical sub-library II compounds against

    PTP1B

    66

    2.3 Experimental section 68

    2.3.1 Materials and methods for chemical synthesis 68

    2.3.2 Synthesis of library compound precursors 69

    2.3.2.1 Synthesis of aldehyde precursors 69

    2.3.2.1.1 General procedure for the preparation of (A – E, H and K-O) 69

    2.3.2.1.2 Procedure for the preparation of (F) 69

  • vi

    2.3.2.1.3 Procedure for the preparation of (G) 69

    2.3.2.2 Synthesis of isoxazol-5(4H)-one precursors 72

    2.3.2.2.1 General procedures for synthesis of β-hydroxy esters 72

    2.3.2.2.2 General procedures for synthesis of β-keto esters 75

    2.3.2.2.3 General procedure for the synthesis of isoxazol-5(4H)-one 78

    2.3.2.3 Library synthesis procedure: 81

    2.3.3 In vitro studies 87

    2.3.3.1 Materials 87

    2.3.3.2 IC50 determination: 87

    2.3.3.3 LB plot analysis 88

    2.3.4 In vivo studies 89

    2.3.4.1 Toxicity test of compounds C3 on mice 89

    2.3.4.1.1 Animal and diet selection for toxicity 89

    2.3.4.1.2 Feeding 89

    2.3.4.1.3 Drug dose, gavages preparation and administration 89

    2.3.4.2 Biological effect of C3 in diet-induced obese mice: 90

    2.3.4.2.1 Animals and diets 90

    2.3.4.2.2 Feeding 90

    2.3.4.2.3 Drug administration 90

    2.3.4.2.4 Body weight and food intake 91

    2.3.4.2.5 Intraperitoneal glucose tolerance test 91

    2.3.4.2.6 Blood and tissue collection 91

  • vii

    2.3.4.2.7 Triglyceride, total cholesterol and free fatty acid analysis 92

    2.3.4.2.8 Statistical analysis 92

    2.4 Summary and Conclusions 93

    3. Biological Evaluation of 4,4'-Methylenebis(5-(furan-2-yl)-2-

    hydroxybenzoic acid) 95

    3.1 Introduction 95

    3.2 Result and discussion 98

    3.2.1 Synthesis of difuranyl methylenedisalicylic acid 98

    3.2.2 Animal experiment 99

    3.2.2.1 Toxicity test of compounds SA37 on mice 99

    3.2.2.1.1 Animal and diet selection for toxicity 99

    3.2.2.1.2 Feeding 99

    3.2.2.1.3 Drug dose, gavages preparation and administration 99

    3.2.2.1.4 Effect of compounds SA37 on SLC: ICR Mice 100

    3.2.2.1.5 Body weight after 6 days of SA37 treatment 101

    3.2.2.1.6 Carcass of the mice at 6 days of SA37 treatment 101

    3.2.2.1.7 Conclusion 103

    3.2.2.2 Effect of compounds SA37 on diet induced diabetic mice 103

    3.2.2.2.1 External appearance and body weight gain of mice after obesity

    development period

    104

    3.2.2.2.2 Body weight after 4 weeks of SA37 treatment 105

    3.2.2.2.3 Body weight gain during 4 weeks of SA37 treatment 107

  • viii

    3.2.2.2.4 Food intake 108

    3.2.2.2.5 Feed efficiency 109

    3.2.2.2.6 Intraperitoneal glucose tolerance test (IPGTT) 110

    3.2.2.2.7 Carcass appearance at 4 weeks of SA37 treatment 111

    3.2.2.2.8 Liver, kidneys and lungs weights 112

    3.2.2.2.9 Epididymal and retroperitoneal fat pad weights 113

    3.2.2.2.10 Blood analysis 114

    3.3 Experimental 115

    3.3.1 Materials and methods for chemical synthesis 115

    3.3.2 Synthesis of chemical compounds 116

    3.3.3 Materials and methods for animal experiment 119

    3.3.3.1 Animals and diets 119

    3.3.3.2 Feeding 119

    3.3.3.3 Drug administration 119

    3.3.3.4 Body weight and food intake 120

    3.3.3.5 Intraperitoneal glucose tolerance test 120

    3.3.3.6 Blood and tissue collection 120

    3.3.3.7 Triglyceride, total cholesterol and free fatty acid analysis 121

    3.3.3.8 Statistical analysis 122

    3.4 Summary and conclusions 122

  • ix

    Figure List of figures Page

    1-1 Protein phosphorylation and dephosphorylation 1

    1-2 Catalytic mechanism of PTP1B 5

    1-3 Bis(maltolato)oxovanadium(IV) compound 8

    1-4 Peptide ligand with two DFMP 9

    1-5 Nonhydrolyzable DFMP pTyr mimetics 10

    1-6 Carboxylic acid pTyr mimetics 11

    1-7 TDZ pTyr mimetics 13

    1-8 Some novel pTyr mimetics 15

    1-9 Some potent PTP1B inhibitors reported from our lab 18

    1-10 Novel PTP1B inhibitors identified from virtual screenin 19

    1-11 Two most potent PTP1B inhibitors from TZD-series 20

    1-12 Most potent PTP1B inhibitors from barbituric acid-serirs (52) and

    analogue compound of virtual hit 42 (53)

    21

    2-1 Some PTP1B ihibitors 27

    2-2 Precursors for small library compounds 30

    2-3 Small library compounds 31

    2-4 Commercially available aldehydes precursor for Chemical Library

    Synthesis

    35

    2-5 Precursors for chemical library synthesis 36

    2-6 Solubility of precursors for chemical library compounds 38

    2-7 Precipitation of chemical library compounds at room temperature 40

  • x

    2-8 Chemical library compounds 41

    2-9 Lineweaver burk plot analysis for PTP1B catalyzed reaction in presence

    of C3

    45

    2-10 Physical appearance of control mouse and C3 treated mouse 48

    2-11 Carcass of the mice at 6 days of C3 treatment 49

    2-12 Liver and other organs appearance at 6 days of C3 treatment 50

    2-13 Appearance of mice after obesity development period 52

    2-14 Appearance of mice drug feeding period 53

    2-15 Body weight gain during 4 weeks C3 treatment 54

    2-16 Cumulative food intake during 4 weeks of C3 treatment 55

    2-17 Feed efficiency in mice 56

    2-18 Intraperitoneal glucose tolerance test (IPGTT) 57

    2-19 Carcass of Mice after C3 treatment 58

    2-20 Effect of C3 in Liver, Kidneys and Lungs Weights after 4 Weeks of

    Treatment

    59

    2-21 Effect of C3 in Epididymal and Retroperitoneal Fat Pad Weights after 4

    Weeks of Treatment

    60

    2-22 Effect of C3 in Serum Concentration of (A) Total Cholesterol and

    Triglyceride; and (B) NEFA Levels after 4 Weeks of Treatment

    62

    2-23 Chemical Sub Library I Compounds 64

    2-24 Chemical Sub Library II Compounds 66

    3-1 Some MDSA derivatives 96

    3-2 Physical appearance of HFD-control and SA37-treated mouse 100

    3-3 Carcass of the mice after 6 days of SA37 treatment 102

  • xi

    3-4 The appearance of organs after 6 days of SA37 treatment 103

    3-5 Appearance of mice after 8 weeks of obesity development period 105

    3-6 Appearance of mice after drug feeding period 106

    3-7 Body weight gain during 4 weeks of SA37-treatment 107

    3-8 Cumulative food intake during 4 weeks of SA37 treatment 108

    3-9 Feed efficiency in mice 109

    3-10 Intraperitoneal glucose tolerance test (IPGTT) 110

    3-11 Carcass of HFD control and SA37-treated mice 111

    3-12 Effect of SA37 in liver, kidneys and lungs weights after 4 weeks of

    treatment

    112

    3-13 Effect of SA37 in epididymal and retroperitoneal fat pad weights after 4

    weeks of treatment

    113

    3-14 Effect of SA37 in serum concentration of total cholesterol and

    triglyceride; and NEFA levels

    114

  • xii

    Table List of tables Page

    1-1 IC50 or Ki

    of carboxylic acid pTyr mimetics 12

    1-2 IC50 or Ki of TDZ pTyr mimetics 14

    1-3 IC50 or Ki of some novel pTyr mimetics 16

    2-1 Inhibitory effect of small library compounds against PTP1B 32

    2-2 Nature of chemical library compounds 40

    2-3 Percentage of inhibition by 10 µM solution of the library compounds A1-

    J7 against PTP 1B 42

    2-4 Inhibitory potency of compounds A3, B3, C2, C3, C5, C8 against PTP1B 43

    2-5 Inhibitory effect of compound C3 on various PTPs 44

    2-6 Body weight after 6 days of C3 treatment 48

    2-7 Body weight gain during obesity development Period 52

    2-8 Body weight after 4 weeks of C3 treatment 53

    2-9 Inhibitory potency of chemical sub library I compounds against PTP1B 65

    2-10 Inhibitory potency of chemical sub library II compounds against PTP1B 67

    2-11 IC50 of chemical sub-library II compounds against PTP1B 68

    3-1 IC50 values of disalicylic acid derivatives against PTP1B and IKK β 97

    3-2 Body weight after 6 days of SA37 treatment 101

    3-3 Body weight gain during 8 weeks of obesity development period 104

    3-4 Body weight after 4 weeks of SA37 treatment 106

  • xiii

    Scheme List of scheme Page

    2-1 Synthesis of (Z) 4(2(2 chlorobenzyloxy)benzylidene) 3 phenylisoxazol-

    5(4H)-one (Lead Compound)

    28

    2-2 Synthesis of aldehydes precursors for small librarycompounds 29

    2-3 Synthesis of small library compounds 30

    2-4 Synthesis of substituted 3 phenyliisoxazol-5(4H)-one derivative 33

    2-5 Synthesis of aldehydes precursors for chemical library synthesis 34

    2-6 Synthesis of aldehydes precursors (G) for chemical library synthesis 35

    2-7 Chemical library synthesis 37

    2-8 Synthesis of substituted 3 phenylisoxazol-5(4H)-one derivative 63

    2-9 Hydrolysis of C7 to C3ix 64

    3-1 Synthesis of difuranyl methylenedisalicylic acid 98

  • xiv

    List of abbreviations

    ACS Acyl-CoA synthetase

    ACOD Acyl-CoA oxidase

    ATP Adenosine triphosphate

    BSA Bovine serum albumin

    BW Body Weight

    13 C NMR Carbon-13 nuclear magnetic resonance

    Chemical shift

    DAG Diacylglycerol

    DMSO Dimethyl sulfoxide

    DTT Dithiothreitol

    E.coli Escherechia coli

    EDTA Ethylenediaminetetraacetic acid

    FFA Free fatty acids

    kg; g; mg Kilogram; gram; milligram

    HFD High fat diet

    1H NMR Proton nuclear magnetic resonance

    Hepes (N-[2-hydroxyethyl]piperazine-N’-[ethanesulfonic acid]

    IC50

    Half-maximal inhibitory concentration

    IκB Inhibitor of NFκB

    IKK IκB kinase

    IL-1 β Interleukin -1 β

  • xv

    IPGTT Intraperitoneal glucose tolerance test

    IR Insulin receptor

    IRS Insulin receptor substrate

    JAK2 Janus kinase 2

    LFD Low fat diet

    mL; L Milliliter; microliter

    M; mM; M Molar; millimolar; micromolar

    MHz Megahertz

    mmol Millimole

    NEFA Non-esterified free fatty acids

    nm Nanometer

    NFκB Nuclear factor κB

    pNPP p-Nitrophenyl phosphate

    PTP Protein tyrosine phosphatase

    PTP1B Protein tyrosine phosphatase 1B

    PTK Protein tyrosine kinase

    pTyr Phosphotyrosyl

    SHP-1 Src homology 2 (SH2) domain-containing PTPase

    TC-PTP T-cell protein tyrosine phosphatase

    T-Cho Total Cholesterol

    TG Triglyceride

    TLC Thin-layer chromatography

  • xvi

    TNF- α Tumor necrosis factor-α yeast protein tyrosine posphatase 1

    VHR Vaccinia H1-related phosphatase

    TZD 2, 4- Thiazolidinediones

    YPTP1 Yeast Protein tyrosine phosphatase

  • xvii

    Abstract

    In developing inhibitors of therapeutic target enzymes, significant time and effort are

    committed to the preparation of large numbers of compounds. In an effort to develop a

    potent inhibitor of PTP1B as an anti-obesity and/or anti-diabetic agent, we constructed an

    isoxazolone chemical library using a simplified procedure that circumvents tedious work up

    and purification steps. The 10 7 isoxazolone derivatives were synthesized by coupling the

    two halves of the target compounds. When mixed and heated in test tubes, the precursors

    produced the reaction products as precipitates. After brief washing, the products were pure

    enough to be used for enzymatic experiments. With the precursors for the coupling reactions

    prepared, the 10 7 library compounds could be prepared in a day using the present

    protocol. The library compounds thus obtained were examined for their inhibitory activities

    against PTP1B. Among them, compound C3 was the most potent inhibitor of PTP1B with

    an IC50 of 2.3 μM. The in vivo effect of C3 was also examined in an obesity-prone mouse

    strain. Diet-induced obese/diabetic mice were divided into two groups and each group was

    fed high fat diet (HFD) or HFD + C3 for 4 weeks. The group of C3-fed mice gained

    significantly less weight compared to the HFD-fed control group during the 4 weeks of the

    drug feeding period. In contrast to the anti-obesity effect of C3, no difference was observed

    in the glycemic control of the HFD and HFD + C3 mice groups.

  • xviii

    요 약

    의약개발시 표적 표적효소에 대한 억제제 개발과정에서 다수의 화합물들을

    합성하는데 상당한 시간과 노력이 소모된다. 비만 및 당뇨 치료제로 사용될 수

    있는 PTP1B 억제제 개발을 위하여 옥사졸론 화합물 라이브러리를 제작하였는데

    반응 후 처리와 정제과정을 생략할 수 있는 방법을 사용하였다. 표적화합물들의

    반쪽에 해당하는 두 개의 전구체들을 축합하여 10 7 옥사졸론 유도체들을

    합성하였는데, 전구체들을 시험관 내에서 혼합 가열하면 생성물이 침전형태로

    얻어졌다. 약간의 용매로 세척하는 것만으로 효소 실험에 사용하기에 충분한

    순도의 화합물을 얻을 수 있었다. 축합반응을 위한 전구체들이 준비된 상태에서

    10 7 라이브러리를 하루 동안에 제작할 수 있었다. 라이브러리 화합물들에

    대하여 PTP1B 억제활성을 측정한 결과 가장 강력한 억제제는 화합물 C3 로 IC50

    값은 2.3 μM 이었다. 비만이 되기 쉬운 생쥐 모델을 이용하여 화합물 C3 의 생체

    내 효과를 실험하였다. 생쥐들에게 고지방식을 먹여 비만/당뇨를 유발한 뒤

    생쥐들을 두 그룹으로 나누고 각각 고지방식 또는 고지방식 + C3 를 4 주간

    먹였다. 화합물 C3 투여 그룹은 고지방식만 먹은 그룹에 비해 체중증가가

    현저하게 감소하였다. 화합물 C3 에 의한 혈당조절효과는 관찰되지 않았다.

  • 1

    CHAPTER I

    1. General Introduction

    1.1. Phosphorylation and dephosphorylation

    Kinases and phosphatases are counteracting partners in regulating signaling

    responses. The kinases control the amplitude of a signaling response, and

    phosphatases play a role in controlling the rate and duration of the response.1

    The diversity and complexity of the PTP and PTK are comparable. Similar to

    PTK, PTP are equally important in controlling the signaling pathways in

    both negative and positive manner. Furthermore, biochemical and genetic

    studies indicate PTP play a key role in the regulation of many physiological

    processes in various mammalian tissues and cells.2,3

    Figure 1-1. Protein phosphorylation and dephosphorylation

    1.2. Phosphatases

    One hundred and seven PTP genes were found in human genome, and 81 of

    them encode active phosphatase.4 All the PTPs are characterized by a

    conserved active site sequence (H/V)C(X)5R(S/T), term as PTP signature

    motif, in which the cysteine residue functions as a nucleophile and is

    essential for catalysis.5 PTPs can be divided into three subclasses: dual-

  • 2

    specificity PTPs (DSP-PTPs), low molecular weight PTPs (LMW-PTPs) and

    classical tyrosine-specific PTPs.

    Dual specificity PTPs (DSP-PTPs) are capable of dephosphorylating serine

    and threonine residues as well as tyrosine residues. LMW-PTPs constitute a

    family of 18 kDa enzymes with specificity primarily towards

    phosphotyrosine.

    The classical tyrosine-specific PTPs can be further divided in two

    subfamilies: receptor-like PTPs and cytosolic PTPs. The receptor-like PTP

    contain a transmembrane domain, an extracellular receptor like domain and

    one or two PTP catalytic intracellular domains. The catalytic activities of

    PTP reside in membrane-proximal (D1) PTP domain except in PTPα6, while

    the membrane-distal domain (D2) is almost inactive and is considered to

    play a regulatory role.

    Cytosolic PTPs contain a single catalytic domain and flanking regions with

    putative roles in the regulation of catalytic activity, protein-protein

    interactions and subcellular targeting.7 PTP1B is an example of cytosolic

    PTPs, that contains a carboxy-terminal endoplasmic reticulum-targeting

    domain. The most prominent substrates for PTP1B is the insulin receptor (IR)

    and insulin receptor substrate (IRS) proteins in skeletal muscle and liver.8,9

    1.3. Importance of phosphatases

    About 4% of the ‘druggable genome’ is thought to be phosphatases.10

    Some

    of the validated phosphatases as a drug target are PTP1B, 8,11,12,13,14,15

    SHP-1, SHP-2,16,17

    CD45,18

    PTP-α, PTP-ε, Prl-3, Cdc25,19,20

    PTP-β (VE-

    PTP), PTP-ε,21

    PTP-σ, GLEPP-1(PTP-oc), and PTP-ε.22,23

    Among PTPs,

    PTP1B is the most exploited PTP, and recently validated as a therapeutic

    target for obesity and diabetes.

  • 3

    1.4. Protein Tyrosin Phosphatase 1B

    PTP1B is the first PTP to be purified from human placental tissue.24

    PTP1B

    is widely expressed cytosolic enzyme and is primarily localized in the

    endoplasmic reticulum (ER) via a cleavable proline-rich C-terminal

    segment.25

    PTP1B is a 50 kD PTP that consists of 435 amino acids.

    Residues 30-278 comprise catalytic domain and 35 carboxyl terminal

    residues target enzyme to the cytosolic face of the endoplasmic reticulum.26

    Most of the structural features of PTP1B such as P-loop (His-Cys-Ser-Ala-

    Gly-Ile-Gly-Arg) and WPD loop are common in other PTPs.27

    The active

    site of PTP1B is defined by a loop of eight amino acid residues (214-221)

    and are important for catalysis. The cysteine residue (Cys215) is critical for

    catalytic activity. Second phosphotyrosine binding site has been discovered

    adjacent to the catalytic site.28

    Identification of the second phosphotyrosine

    binding site provided an additional opportunity for the design of more potent

    and specific inhibitors.28

    1.5. Landmarks in PTP1B research29

    (Yip S. C. et al, 2010):

    1988 - Purification and characterization of PTP1B from human placenta.30,31

    1990-Cloning of hPTP1B cDNA, Microinjection in Xenopus laevis

    oocytes.32,33,34,27

    1992 - ER targeting domain identified, the effect of PTP1B on Neu- and V-

    src transformed fibroblasts.25

    1993 - Serine phosphorylation of PTP1B, Calpain-induced cleavage of the

    PTP1B C-terminal tail.35,36

    1994 - Crystal structure of hPTP1B (1–321 at 2.8 A°), PTP1B over-

  • 4

    expression in human breast & ovarian cancers.27

    1995 - Osmotic loading of neutralizing antibody.37

    1996 - Characterization of SH3 domain-containing PTP1B binding partners.

    38

    1997 - Development of substrate-trapping mutant: PTP1B-D181A.39

    1998 - Suppression of transformation.40

    1999 - Ptp1b knockout mice generated.8

    2000 - Positive role in c-Src activation in cancer cells.41

    2001 - ROS regulation of PTP1B identified TYK2 and JAK2 as substrates.

    42,43

    2002 - PTP1B attenuates leptin signaling, FRET imaging of receptor

    dephosphorylation.

    2003 - Structure of oxidized PTP1B.44

    2004 - Positive role in Ras signaling, Muscle-specific Ptp1b knockout mice

    generated.45,46,47

    2005 - Crystal structure of non-catalytic binding to IR.48

    2006-Brain-specific Ptp1b knockout mice generated.8

    2007 - SUMO regulation of PTP1B, MMTV-Erbb2; Ptp1b-/-

    mouse models

    generated.49,50,51

    2008 - Quantitative SILAC proteomics of PTP1B substrates.52

    2009 - Src activation in ErbB2-mediated transformation in 3D culture, Liver-

    specific Ptp1b knockout mice generated.53,54

  • 5

    1.6. General mechanism of dephosphorylation of substrate proteins by

    PTP1B

    The phosphotyrosine of substrate proteins binds deeply in the catalytic site of

    phosphate-binding loop (P-loop). The binding of phosphotyrosine are

    mediated by residues 214-221 (His, Cys, Ser, Ala, Gly, Ile, Gly, Arg) of

    PTP1B. The WPD (tryptophan, proline, aspartic acid) loop closes down onto

    the substrate and Asp181 acts as a general acid catalyst. The phosphate is

    cleaved from phosphotryosine residue, allowing dephoshorylated substrate to

    diffuse from the active site and water to replace it. General base catalyzed

    hydrolysis of the resultant phosphocysteine by Asp181 regenerates the active

    form of phosphatase and completes the catalytic cycle.55

    O

    Enzyme

    P

    O

    -O

    -O

    pTyr substrate

    Asp

    OHO

    + HO

    Hydrolysed substrate

    S PCys

    O

    O-O

    -

    HO

    Ser

    OH

    H

    Asp

    O-

    O

    O

    GlnH2N

    Thiophosphoryl enzymeintermediate

    Enzyme

    + P-O-O

    OH

    O

    215

    181

    222

    181

    262

    Cys S-

    215

    Cys S P

    O

    O-

    -O

    215

    Cys S-

    215

    A

    B

    HO

    Ser 222

    HO

    Ser 222

    HO

    Ser 222

    Asp

    OHO

    181

    Asp

    O-

    O

    181

    Thiophosphoryl enzymeintermediate

    Figure 1-2. Catalytic mechanism of PTP1B56

  • 6

    Entire PTPs share a common catalytic mechanism. (A) Formation of

    thiophosphoryl enzyme intermediate: thiolate anion from the active site

    cysteine attacks phosphate of the substrate and forms a thiophosphoryl

    enzyme intermediate. Expulsion of dephosphorylated substrate is aided by

    the general acid (Asp) of the enzyme.

    (B) Hydrolysis of thiophosphoryl enzyme intermediate: A water molecule

    activated by the general base (Asp) attacks the intermediate. The glutamine-

    262 positions the attacking water molecule. The hydrogen bond with the

    serine-222 compensates the negative charge developed on the S atom of the

    active site cysteine.

    1.7. Validation of PTP1B as a therapeutic target

    The validation of PTP1B as therapeutic target for diabetes and obesity are

    supported by growing number of evidences collected from several

    biochemical, pharmacological and genetic studies. Two research groups

    independently reported that PTP1B knockout mice displayed a phenotype

    strongly suggestive of a role in insulin and leptin signaling.8,11

    The PTP1B

    deficient mice showed enhanced insulin sensitivity, lower plasma glucose

    and insulin levels, and resistance to weight gain compared to control mice

    with high fat diets. The PTP1B deficient mice also exhibited normal

    development and longevity. In contrast, TC-PTP deficient mice died at 3-5

    weeks of age. The death of TC-PTP knockout mice was due to impaired B

    cell and T cell functions. Even though, TC-PTP is the most homologous

    phosphatase to PTP1B.57

    Several studies on antisense oligionucleotide,

    overexpression in vitro, human single nucleotide polymorphisms, and

    observations of mutations within the human PTP1B gene sequence were

  • 7

    conducted to further validate the role of PTP1B as a therapeutic target.58,59

    Xie, L. et al. reported that small-molecule inhibitors of PTP1B can work

    synergistically with insulin to increase insulin signaling and augment insulin-

    stimulated glucose uptake.60

    In another approach, pretreatment of leptin-

    resistant rats with a potent and selective PTP1B inhibitor results in a marked

    improvement in leptin-dependent suppression of food intake.61

    On the basis

    of these evidences, PTP1B is considered as one of the validated biological

    targets for non-insulin dependent diabetes and obesity.

    1.8. Some important PTP1B inhibitors found in literature

    Among PTPs, PTP1B is the most exploited phosphatase as a drug target.

    Several screenings and structural studies were conducted to develop specific

    PTP1B inhibitors. Almost all the research groups faced two major hurdles in

    the development of potent and selective PTP1B inhibitors as a drug, 1)

    highly conserved active site and 2) the regulation of multiple signaling

    pathways by a single PTP as well as the regulation of single pathway by

    diverse PTPs. To overcome aforementioned obstacles, different research

    groups gleaned more biological roles and structural features of PTPs. On the

    basis of available information, several research groups designed synthesized

    and reported several inhibitors that target PTP1B.

    1.8.1. Vanadate (VO4-3

    ) as a non-specific phosphatase inhibitor

    In proper oxidation state, Vanadium is highly similar to phosphate.62

    It can

    enter into tight covalent complexes within the PTP catalytic site, and can

    serve as an effective PTP inhibitor.63,64

    Vanadate (VO4-3

    ) was found to be a

    non-specific phosphatase inhibitor acting as insulin mimics.65 , 66

    Bis(maltolato)oxovanadium(IV) compound 1, has been reported as a potent

  • 8

    PTP1B inhibitor with a blood glucose lowering effect in diabetic animal

    models.67

    Figure 1-3. Bis(maltolato)oxovanadium(IV) compound

    1.8.2. DFMP pTyr mimetics

    The pTyr moiety binds deeply into the phosphatase active site. It is

    responsible for majority of binding energy to PTP1B. Thus, several small

    molecule PTP1B inhibitors with non-hydrolyzable or poorly hydrolyzable

    pTyr mimics were reported. Zhang (1997) reported a highly potent (PTP1B

    Ki = 2.4 nM) and selective (TC-PTP 10-fold selectivity) peptide ligand 2

    with two DFMP groups.28

    Unfortunately, the highly polar nature of the bis-

    anionic DFMPs and peptide scaffold provided compounds with poor

    physiochemical properties i.e. poor cell permeability and low oral

    bioavailability.

  • 9

    Figure 1-4. Peptide ligand with two DFMP

    1.8.3. Nonhydrolyzable pTyr mimetics

    As peptide backbone is not good for the development of effective drugs, the

    focus has mainly been on the development of non-peptide low molecular

    weight inhibitors. Combs A.P. (2010) classified PTP1B inhibitor into four

    classes and are summarized hereafter.55

    1.8.3.1. Nonhydrolyzable DFMP pTyr mimetics (Combs A.P., 2010)

  • 10

    Figure 1-5. Nonhydrolyzable DFMP pTyr mimetics

    Several approaches of non-peptide scaffolds with DFMP group have been

    trailed by various pharmaceutical companies. Merk Frosst reported three

    promising PTP1B inhibitor classes: arylketone DFMP 3 (IC50 = 120 nM),

    benzotriazole DFMP 4 (IC50 = 5 nM), and naphthyl DFMP 5 (IC50 = 120

    nM).68,69 ,70

    These inhibitor classes demonstrated modest degree of cell

    permeability and oral bioavailability in rodents. However bis-anionic DFMP

    demonstrated less desirable pharmacokinetics in higher species. Compounds

    3 and 5 demonstrated an antidiabetic effect when tested in an oral glucose

    tolerance test (oGTT) in diet induced obese (DIO) mice. A sulfonamide

    scaffold bearing the DFMP 6 (IC50 = 28 nM) and a novel ketophosphonate

    pTyr mimetic 7 (IC50 = 600 nM) as PTP1B inhibitors were identified.71

    However, selectivity report for compound 6 and 7 do not exist.

  • 11

    1.8.3.2. Carboxylic acid pTyr mimetics

    Figure 1-6. Carboxylic acid pTyr mimetics

  • 12

    Table 1-1. IC50 or Ki

    of carboxylic acid pTyr mimetics

    Compound

    No

    IC50 or Ki in µM PTP1B

    Selectivity

    with respect

    to TC-PTP

    Research

    Group PTP1B TC-PTP

    8 0.018 0.065

    3.6 Abbott

    72

    9 9.0 182

    20 Abbott

    73

    10 8.4 ˃200 ˃23 Abbott

    52

    11 2.1 ˃30 ˃15 Abbott

    74

    12 0.081 NA NA

    Novo

    Nordisk75

    13 3.2 NA NA Kyorin

    Pharma76

    14 0.016 NA NA Inst. of

    Pharm. Dis.77

    15 0.004 0.005

    1 Wyeth

    78

    Highly potent dicarboxylic acid containing PTP1B inhibitors were reported

    by different group. But these inhibitors have poor membrane permeability.

    When one of the carboxylic acid moieties was eliminated to improve the

  • 13

    membrane permeability, the resulting compounds were found to be 100-1000

    fold less potent or converted in to extremely hydrophobic compounds that

    may result in nonspecific inhibition.

    1.8.3.3. TDZ pTyr mimetics

    Figure 1-7. TDZ pTyr mimetics

  • 14

    Table 1-2. IC50 or Ki of TDZ pTyr mimetics

    Compound

    No

    IC50 or Ki in µM PTP1B

    Selectivity

    with respect

    to TC-PTP

    Research

    Group PTP1B TC-PTP

    16 1600 NA NA AstraZeneca79

    17 2.5 NA NA AstraZeneca58

    18 0.14 0.15 1 Incyte80

    19 0.032 0.025 0.8 Incyte81

    20 0.27 0.25 1 Incyte82

    21 0.023 0.025 1 Incyte83

    22 4.3 NA NA Incyte

    84

    23 0.104 NA NA Novartis85

    24 0.126 NA NA Novartis86

    25 0.080 NA NA Novartis87

    26 0.3 NA NA Novartis88

    27 0.13 NA NA Novartis77

  • 15

    TDZ and (S)-IZD five-membered heterocyclic pTyr mimetics were reported

    by four independent research groups. These small molecule inhibitors that

    incorporate the TDZ and (S)-IZD pTyr mimetics were the most potent pTyr

    mimetics class of PTP1B inhibitors with better membrane permeability then

    carboxylic acid and DFMP containing inhibitors. But, still lack sufficient

    cellular penetration for in vivo efficacy and oral bioavailability.

    1.8.3.4. Novel pTyr mimetics

    Figure 1-8. Some Novel pTyr mimetics

  • 16

    Table 1-3. IC50 or Ki of some pTyr mimetics

    Compound

    No

    IC50 in µM PTP1B

    Selectivity

    with respect

    to TC-PTP

    Research

    Group PTP1B TC-PTP

    28 2.5 NA NA Sugen89

    29 4.8 9.4 1.9 Proct. &

    Gamble90

    30 0.07 Na NA Proct. &

    Gamble91

    31 8 NA NA Taylor et al92

    32 4.1 5.3 1.3 Tremblay et

    al93

    The sulfamic acid pTyr mimetic obtained from high throughput screening are

    low molecular weight compounds. On optimization these compounds shows

    extremely good inhibitory potency against PTP1B, but the membrane

    permeability of compounds containing this pharmacophore were not found in

    literature.

    1.9. PTP1B inhibitors reported from our lab

    As an effort to develop potent and selective PTP1B inhibitors, our lab

  • 17

    reported a novel series, formylchromone derivatives,94

    disalicylic acid

    derivatives56

    and pyrogallol derivatives95

    .

    The most potent compound from formylchromone derivatives 33 with IC50 =

    1µM, was irreversible inhibitor of PTP1B with good selectivity among

    several PTPases including TC-PTP.

    MDSA derivatives were synthesized and examined for the inhibitory activity

    against diverse PTPs. Among those, three MDSA derivatives 34, 35 and 36

    demonstrated about 10-fold selectivity over TC-PTP and two of the

    compounds, 34 and 36 inhibited PTP1B reversibly. Oral administration of

    compound 34 and 36 to DIO mice significantly reduced body weight gain

    compared with that of the untreated HF control group without suppressing

    food intake.56

    Our lab further modified disalicylic acid compounds to 37 (IC50 = 5 µM) and

    38 (IC50 = 0.5 µM) improved PTP1B inhibitory effect. Compound 37

    showed glucose level lowering effect and improved glucose tolerance in

    HFD-induced diabetic mice.96

    Non-phosphorous small molecule derivatives of pyrogallol with one

    carboxyllic acid group showed low IC50 value upto 2 µM and Ki value of 1.1

    µM against PTP1B. The most potent compound 39 of pyrogallol series was

    7-fold selective for PTP1B than TC-PTP. Oral administration the most potent

    compound 39 of this series exhibited glucose lowering effect and improved

    glucose tolerance in DIO mice without any overt toxicity.97

  • 18

    Figure 1-9. Some potent PTP1B inhibitors reported from our lab

    In order to find potent inhibitors of PTP1B, we screened a drug library called

    ‘The Prestwick Chemical Library® ’

    that contains 1120 small molecules, 90%

    of which are drugs and 10% are bioactive alkaloids or related compound.98

    This library contains a limited number of highly diverse drug molecules for

    which bioavailability and toxicity studies have already been performed and

    proven safety in humans. Thus, the initial hits of this library can be used as a

    starting point for drug optimization in medicinal chemistry.26

    We identified 9

    novel inhibitors of PTP1B by applying a computer-aided drug design

    protocol involving the structure-based virtual screening with docking

    simulations under consideration of the effects of ligand solvation in the

    scoring function. These inhibitors are structurally diverse and reveal potency

    with IC50 values ranging from 10 to 50 µM.

  • 19

    Figure 1-10. Novel PTP1B inhibitors identified from virtual screening

  • 20

    Compound 45, was found to be a competitive inhibitor of PTP1B. Several

    variations in substitutions in ortho-, meta- and para- directions from the

    TZD moiety were synthesized and evaluated for their PTP1B inhibitory

    activity. Most of the synthesized compounds exhibited low µM IC50 values

    against PTP1B. Two of the most potent compounds of different series,

    compound 50 and 51 were tested for their in vivo antihyperglycemic and

    antiobesity effects.

    Figure 1-11. Two most potent PTP1B inhibitors from TZD-series

    Treatment of two of the most potent compounds 50 and 51 of that series to

    the HFD-induced diabetic mice controlled the HFD-induced body weight

    gain and exhibited an improved glucose tolerance effect. The serum

    triglyceride, total cholesterol and free fatty acids levels were found

    significantly lowered.

    Compound 46 was found as a non-competitive inhibitor. We designed and

    synthesized a series of barbituric acid derivative to investigate the potency,

    mode of inhibition, steady-state kinetic experiments of PTP1B and YPTP1

    were performed for compound 52. The mode of inhibition was determined

  • 21

    by the Lineweaver-Burk plot analysis of the results of the kinetic

    experiments. The compound 52 inhibited PTP1B and YPTP1 in a

    noncompetitive and a mixed-type noncompetitive fashion, respectively.

    These results indicate binding site for the compound 52 is different from that

    for the substrate. The noncompetitive inhibition of PTP1B might be

    explained by the possible binding of the barbituric acid moiety in the

    secondary aryl phosphate-binding site, present near the active site of PTP1B.

    Figure 1-12. Most potent PTP1B inhibitors from barbituric acid-serirs (52)

    and analogue compound of virtual hit 42 (53)

    On the other hand, the mixed-type noncompetitive inhibition of YPTP1 by

    52 could hardly be explained on the same basis, because no aryl phosphate-

    binding site distinct from the active site has been identified on YPTP1.

    Nondiscriminative inhibition of the three structurally diverse PTPs by 52

    also implicates the possible similarity of the inhibitor binding sites on this

    PTPs. Explanation of these apparently conflicting observations and

    considerations requires additional studies including X-ray crystallographic

  • 22

    structure determination of the enzyme-inhibitor complex. Enzyme-inhibitor

    interaction yet to be explained at the molecular level, the barbiturate moiety

    was proven to be a promising scaffold for the design of PTP inhibitors.

    To improve the inhibitory potency of barbituric acid derivatives we modified

    the structure of compound 46 and di-substituted barbituric acid compounds

    were synthesized. The work is in progress in our lab.

    We synthesized compound 53 as an analogue of compound 42. The

    inhibitory potency and kinetic study of the compound 53 revealed that it’s a

    promising competitive inhibitor of PTP1B. Further modification of

    compound 42 is in progress.

  • 23

    1.10. Present work

    PTP1B is considered as one of the validated biological targets for non-

    insulin dependent diabetes and obesity. Targeting PTP1B for drug discovery

    is challenging because of the highly conserved and positively charged active-

    site pocket. Potent and selective PTP1B inhibitors, with minimum charge

    density, good pharmacokinetics and pharmacodynamics can be considered as

    a drug.

    Our present work is based on compound 44 (a compound with isoxazol-

    5(4H)-one moiety). The compound was obtained from virtual screening.

  • 24

    1.11. Objectives of this study

    Numerous researches have been performed for the development of selective

    PTP1B inhibitors possessing beneficial effects for diabetes and obesity.

    However, clinically usable drugs have not been developed yet. Aimed for the

    development of PTP1B inhibitors with therapeutic effects, this study focused

    on the synthesis and the biological evaluation of isoxazol-5(4H)-one

    derivatives. Main objectives of the present work are summarized below.

    1. To synthesize a chemical library with a isoxazol-5(4H)-one moiety as

    a common scaffold.

    2. To evaluate their inhibitory potency against PTP1B and the selectivity

    profile of the most potent compound against various PTPases.

    3. To study the inhibition pattern of a most potent compound by kinetic

    experiments.

    4. To examine the in vivo effect of the most potent PTP1B inhibitor in a

    diet-induced obese/diabetic mouse model system.

  • 25

    CHAPTER – II

    2. Synthesis and Biological Evaluation of Isoxazol-5(4H)-one Derivatives

    as a New Family of PTP1B Inhibitors with Anti-Obesity Effects

    2.1. Introduction

    The rapid increase in the prevalence of obesity throughout the world poses a

    serious health threat in modern society.99,100,101

    Obesity enhances the risk of

    associated morbidities, including diabetes, hypertension, dyslipidemia,

    ischaemic heart disease, and even cancer.102

    Existing therapies for obesity

    are limited in number and effectiveness. Only two drugs, orlistat and

    sibutramine, are currently approved for the long-term treatment of obesity.

    These drugs have limitations in their use due to adverse side effects or

    limited efficacy.103

    Another drug, rimonabant, recently developed as an

    appetite suppressant was approved in 2006 in the European Union. However,

    rimonabant was found to increase the risk of adverse psychiatric effects,

    including serious depression and suicide, and its use was suspended in

    2008.104

    To circumvent the problems of the current drugs, numerous studies have

    been devoted to the identification of novel therapeutic targets and significant

    progress has been made.105,106,107

    Among these progresses, inhibition of

    PTP1B was recognized as a promising therapeutic strategy for the

    management of both obesity and diabetes.108

    Genetic ablation of PTP1B in

    mice suppressed weight gain and maintained insulin sensitivity upon feeding

    a high fat diet (HFD). Wild-type mice, on the other hand, gained weight and

    became insulin resistant. Furthermore, PTP1B-depleted mice were healthy,

    without worrisome traits such as enhancement of mitogenic signaling.

  • 26

    Inhibition or reduction of the cellular abundance of PTP1B in mice also

    resulted in increased sensitivity to leptin and insulin, and has been shown to

    exhibit a protective effect against diet-induced obesity.8

    Numerous active site-directed PTP1B inhibitors have been reported, many of

    them with pharmacophores that mimic phosphotyrosine residues.109 , 110

    Among those, Ertiprotafib (Figure 2-1) had progressed to clinical trials but it

    was discontinued in the second phase due to unwanted side effects and

    insufficient efficacy.111

    A noncompetitive PTP1B inhibitor, trodusquemine

    (Figure 2-1), has recently proceeded to the early stages of clinical trials with

    promising preclinical results as both an appetite suppressant and a

    hypoglycemic and hypocholestrolemic agent.112

    First-in-class drug targeting

    PTP1B is yet to be developed and intensive research is underway to develop

    a potent and selective PTP1B inhibitor with anti-obesity and/or

    hypoglycemic effect.

    In an effort to find potent inhibitors of PTP1B, we previously performed a

    virtual screening with docking simulations and identified nine compounds

    with half-maximal inhibitory concentrations (IC50) lower than 50 μM toward

    PTP1B. Among those, compound L4 (Figure 2-1) with an IC50 value 22 μM

    was chosen as a lead compound for this study to develop potent PTP1B

    inhibitors.

  • 27

    Figure-2-1: Some PTP1B inhibitors.

    2.2. Result and discussion

    To obtain a large number of drug-like molecules is one of the preliminary but

    an important step in a drug discovery process. An efficient synthetic strategy

    is required for the synthesis of large number of molecules with minimum

    time and efforts. Diverse techniques have been developed to construct

    chemical libraries such as polymer-supported synthesis, multi component

    one pot condensation, and combinatorial synthesis. In the construction of a

    chemical library, it would be better if a single reaction step is available with

    a simple work-up and purification procedure.

    With the construction of a chemical library in mind, compound L4 with IC50

    22 μM against PTP1B is selected as lead compound. On kinetic study, the

    compound was found to be a competitive inhibitor of PTP1B. We designed

  • 28

    and synthesized a structurally diverse chemical library with an isoxazol-

    5(4H)-one moiety as a common scaffold.

    2.2.1. Synthesis of lead compound (Z)-4-(2-(2-

    chlorobenzyloxy)benzylidene)-3-phenylisoxazol-5(4H)-one (L4)

    The precursor, 3-phenylisoxazol-5(4H)-one, was synthesized from

    benzaldehyde via β-hydoxy ester and β-keto ester. And to prepare aldehyde

    precursor 2-(2-chlorobenzyloxy)benzaldehyde commercially available 2-

    hydroxy benzaldehydes were subjected to benzylation with the 1-chloro-2-

    (chloromethyl)benzene in presence of K2CO3. Coupling 3-phenyl isoxazol-

    5(4H)-one with 2-(2-chlorobenzyloxy)benzaldehyde, we got the lead

    compound.(Scheme2-1).

    Scheme 2-1. Synthesis of lead compound (Z)-4-(2-(2-

    chlorobenzyloxy)benzylidene)-3-phenylisoxazol-5(4H)-one: Reagents and

    conditions: (a) Zn/NH4Cl, rt, 1 h; (b) MnO2/DCM, rt, 24 h, (c) NH2OH·HCl,

    pyridine, EtOH, 65 ºC, (d) K2CO3, DMF, 90 ºC, (e) isopropanol, 65 ºC, 4 h.

    The synthetic strategy for the synthesis of lead compound was used for the

  • 29

    preparation of a small library of compounds L1-L6.

    2.2.2. Synthesis of a small library of compounds L1-L6

    2.2.2.1. Synthesis of aldehyde precursors (A & K-O) for small library

    compounds

    To prepare the ortho or meta or para-substituted derivatives A & K-O,

    commercially available 2 or 3 or 4-hydroxybenzaldehydes were subjected to

    benzylation with appropriate benzylchlorides in presence of K2CO3. (Scheme

    2-2)

    Scheme 2-2: Synthesis of aldehyde precursors (A & K-O) for small library

    compounds: Reagents and Conditions: (a) K2CO3, DMF, 90 ºC, 3-6 h

    2.2.2.2. Precursors for small library compounds

    Six aldehyde precursors and 3-phenylisoxazol-5(4H)-one derivatives were

  • 30

    synthesized to set up the reaction conditions for chemical library synthesis

    and to find the preliminary inhibitory potency trend.

    Figure 2-2: Precursors for small library compounds.

    2.2.2.3. Synthesis of small library compounds

    To check the potency trend of isoxazol-5(4H)-one derivatives against PTP1B,

    we synthesized two series of test compounds, L1-L3 and L4-L6, (Fig. 2-3)

    bearing a benzyloxy or a chlorobenzyloxy substituent at the ortho, meta, and

    para positions respectively on the ring A. Scheme 2-3.

    Scheme 2-3. Synthesis of small library compounds: Reagents and

  • 31

    Conditions: (a) isopropanol, 65 ºC, 4 h

    2.2.2.4. Small library compounds

    Figure 2-3: Small library compounds

    2.2.3. In vitro results of small library compounds

    With the construction of a chemical library in mind, we synthesized two

    series of test compounds, L1-L3 and L4-L6, bearing a benzyloxy or a

    chlorobenzyloxy substituent at the ortho, meta, and para positions

    respectively on the ring A of the 3,4-diarylisoxazol-5-one derivatives. These

  • 32

    compounds, when examined for their inhibitory potency against the enzyme

    activity of PTP1B, exhibited different levels of potency in enzyme inhibition

    with IC50 values ranging from 15 to 102 μM (Table 2-1).

    Table 2-1. Inhibitory effect of small library compounds against PTP1B

    Compd. Mol. Wt IC50(µM)a Compd. Mol. Wt IC50 (µM)

    a

    L1 355.39 102±10 L4 389.83 22±2

    L2 355.39 71±4 L5 389.83 25±1

    L3 355.39 43±3 L6 389.83 15±1

    [a] Data expressed as means ± standard deviations of two experiments. The

    kinetic data are analyzed using the GraFit 5.0 program (Erithacus Software).

    Notably, the assay result revealed that para-isomers L3 and L6 were the

    most potent within the two groups, L1-L3 and L4-L6. Based on this

    observation, a chemical library was designed with structural variations on the

    A and B rings of this series of compounds, with the substituent on the A ring

    being fixed at the para position.

    2.2.4. Chemical library synthesis

    Ten aldehyde precursors and seven isoxazol-5(4H)-one precursors were

    synthesized as synthetic scheme 2-4, 2-5 and 2-6.

    2.2.4.1. Synthesis of seven isoxazol-5(4H)-one precursors for chemical

    library synthesis

    To prepare β-hydroxy ester derivative, mixture of appropriate commercially

  • 33

    available aldehyde and ethyl bromoacetate were ground with Zn/NH4Cl. The

    reactions were carried out in solvent free condition for liquid aldehydes.

    Minimum volume of THF (Technical Grade) was used for the solid

    aldehydes. The β-hydroxy ester derivatives were oxidized to β-keto esters by

    MnO2 catalyzed oxidation reaction. On pyridine catalyzed cyclisation, β-keto

    esters converted into corresponding substituted 3-phenylisoxazol-5(4H)-one.

    (Scheme 2-4).

    Scheme 2-4: Synthesis of substituted 3-phenylisoxazol-5(4H)-one

    derivative: Reagents and conditions: (a) Zn/NH4Cl, rt, 5 min – 2 h; (b)

    MnO2/DCM, rt, 24 h, (c) NH2OH·HCl, pyridine, EtOH, 65 ºC.

    2.2.4.2. Synthesis of aldehyde precursor (A-F and H) for chemical

    library synthesis

    4-hydroxybenzaldehydes were subjected to benzylation with the appropriate

  • 34

    benzylchlorides in presence of K2CO3. To prepare the p-tosyl derivative F

    commercially available 4-hydroxybenzaldehydes were subjected to

    tosylation with p-toluenesulfonyl chloride in presence of DIEPA. (Scheme 2-

    5)

    Scheme2-5: Synthesis of aldehyde precursors for chemical library Synthesis:

    Reagents and conditions: (a) K2CO3, DMF, 90 ºC, 3-6 h (b) DIEPA, DMF, 0

    ºC to rt.

    2.2.4.3. Synthesis of aldehyde precursor (G) for chemical library

    synthesis

    To convert the benzonitrile to tetrazol, a solution of 4-(4-formylphenoxy)

  • 35

    benzonitrile, NaN3 and Et3N·HCl in toluene was refluxed for 6 h.

    Scheme 2-6: Synthesis of aldehyde precursor (G) for chemical library

    synthesis: Reagents and conditions: (a) NaN3, Et3N·HCl, 4-(4-

    formylphenoxy) benzonitrile, and toluene, reflux for 6 h.

    2.2.4.4. Commercially available aldehydes precursor (I and J) for

    Chemical Library Synthesis

    4-(5-pyrimidinyl)benzaldehyde (I) and 5-(3-chloro-4-methoxyphenyl)-2-

    furaldehyde (J) were Purchased from Aldrich.

    Figure 2-4: Commercially available aldehyde precursors (I and J) for

    Chemical Library Synthesis:

  • 36

    Figure 2-5: Precursors for chemical library synthesis

  • 37

    2.2.4.5. Chemical library synthesis

    In vitro data obtained from small chemical library hints that the para

    substituent on the A ring might be better for further investigations. We

    designed and synthesized seventeen precursors. Eight aldehyde precursors

    (A–H) were synthesized. Two aldehyde precursors I and J were purchased

    from Aldrich. Seven substituted 3-phenylisoxazol-5(4H)-one (1-7)

    derivatives were synthesized. Table 2-5.

    The aldehyde precursors were coupled with isoxazol-5(4H)-one precursors

    by catalyst free Knoevenagel type condensation reaction.

    Scheme 2-7. Chemical library synthesis: Reagents and Conditions: (a)

    isopropanol, 65 ºC, and 4 h.

    2.2.4.6. Solubility determination of precursors for chemical library

    synthesis

    The solubility of the aldehydes and isoxazol-5(4H)-one precursors was

    determined by dissolving 0.05 mmol of aldehydes (A–J) and substituted 3-

    phenylisoxazol-5(4H)-one (1–7) in a minimum volume of reaction solvent

    isopropanol at reaction temperature 65 ºC. (Fig 2-6)

  • 38

    Figure 2-6: Solubility of precursors for chemical library compounds

    2.2.4.7. Synthesis chemical library compounds

    The strategy for the library synthesis is shown in (Scheme 2-7). The

    condensation reaction of (A-J) and (1-7) was accomplished without catalyst

    by heating the reaction mixture at 65 ºC. In this study, all the condensation

    reactions proceeded in the absence of piperidine or any other catalysts.

    Furthermore, the product precipitated on cooling of the reaction mixture,

    which eliminated work up process and saved the time and effort for the

    isolation of the desired product. This feature is one of the conspicuous

    virtues in the library synthesis for the present study.

    We developed a protocol that uses test tubes as reaction vessels and

  • 39

    eliminates the aqueous work up step to minimize the time and effort required

    for the preparation of a chemical library of 70 compounds,. Briefly, 70 test

    tubes (10 mL capacity) were set in a 10 × 7 array. Solutions of benzaldehyde

    derivatives (A–J) and isoxazol-5(4H)-one derivatives (1–7) (0.05 mmole

    each) in isopropanol were placed in glass test tubes in 10 × 7 combinations.

    Following heating for 4 h at 65 ºC in a dry heating block and cooling to room

    temperature, the reaction products had separated out as precipitates in most

    of the reaction tubes. In some cases where no precipitate had formed, the

    addition of hexane (0.5 mL) and cooling to -50 ºC effectively induced

    precipitation. Removal of the solvent and washing the precipitate with

    isopropanol (0.5 mL) afforded a solid product which showed essentially a

    single spot in thin layer chromatography (TLC) analysis. Even though the

    products can be further purified by recrystallization or column

    chromatography, the precipitates were used as such for enzyme experiments.

    2.2.4.8. Nature of chemical library compounds

    After four hour reaction at 65 ºC, the reaction mixtures were cooled to room

    temperature. Most of the reaction products were obtained as precipitate as

    shown in Figure below.

  • 40

    Table 2-2: Nature of chemical library compounds

    P = The product precipitate on cooling to room temperature

    S = The product is soluble in reaction solvent even on cooling to room

    temperature. Precipitation induced by adding hexane and cooling at -50 ºC.

    Figure 2-7: Precipitation of chemical library compounds at room

    temperature

  • 41

    Figure 2-8: Chemical library compounds

  • 42

    2.2.5. In vitro results of library compounds

    2.2.5.1. Inhibitory potency of library compounds against PTP1B

    Crude products were harvested from chemical library synthesis. 100 µM

    solution of each product were prepared and assayed against PTP1B. The

    final concentration of assayed solution was 10 µM.

    Table 2-3. Percentage of Inhibition by 10 µM solution of the library

    compounds (A1-J7) against PTP 1B

    1 2 3 4 5 6 8

    A 27.8 76.4 97.8 13.5 64.2 16.7 26.7

    B 47.0 38.0 100.0 42.2 52.2 20.6 42.7

    C 67.1 98.3 97.8 86.1 94.4 78.3 94.4

    D 21.4 1.7 27.4 NI NI NI NI

    E NI 35.1 57.0 0.0 7.8 0.5 NI

    F 10.7 33.5 85.5 17.6 NI 36.1 27.1

    G NI 27.3 34.6 87.4 25.0 31.5 27.1

    H NI NI 38.5 NI NI NI NI

    I NI NI 18.4 NI NI NI NI

    J 33.9 63.2 83.8 NI 70.6 4.1 4.0

  • 43

    NI = No Inhibition

    Eighteen compounds out of seventy showed >50% inhibition for PTP1B

    enzyme activity. Six compounds inhibited the PTP1B activity by >90%.

    Compounds at column 3 and row C exhibited higher potency compared to

    other columns and rows. IC50 values are shown as numbers for the

    compounds that inhibit >90% PTP1B enzyme activity.

    Table 2-4. Inhibitory potency of compounds A3, B3, C2, C3, C5, C8 against

    PTP1B

    Compd. Mol. Wt IC50(µM)a Compd. Mol. Wt IC50 (µM)

    a

    A3 457.83 2.5±1.3 C3 507.38 2.3±0.0

    B3 457.83 3.9±1.1 C5 516.47 4.2±0.0

    C2 518.28 2.4±0.1 C7 497.42 7.2±0.5

    aData expressed as the mean standard deviations of two experiments.

    2.2.5.2. Selectivity of most potent compound C3 against other PTPs

    The inhibitory activity of C3 against other PTPs was tested in order to

    examine the PTP1B selectivity of C3, the results of which are summarized in

    Table 2-5. Compound C3 demonstrated a 10-fold greater selectivity over

    TC-PTP, the most homologous with PTP1B among the human PTPs, and a

    2.9-fold greater selectivity over the catalytic domain of SHP-1 (SHP-1cat).

    Compound C3 showed a 12-fold greater selectivity for PTP1B versus VHR,

    a dual specificity PTP. YPTP1, a yeast PTP, was also tested to compare the

    PTPs of different origin, and it exhibited 4.6-fold selectivity.

  • 44

    Table 2-5. Inhibitory effect of compound C3 on various PTPs

    Compound PTP1B TCPTP SHP1-cat YPTP1 VHR

    C3 2.3 ± 0 24 ± 3 6.7 ± 1 11 ± 0 27 ± 1

    IC50 (μM), Values are means ± standard deviations of two experiments. The

    kinetic data were analyzed using the GraFit 5.0 program (Erithacus

    Software).

    2.2.5.3. Enzyme kinetic experiments

    To investigate the mode of inhibition of C3, the enzyme activity of PTP1B

    was determined with varying the concentration of the substrate, p-

    nitrophenyl phosphate (pNPP). Lineweaver-Burk plot analysis of the results

    by plotting the reciprocal of the enzyme reaction rates versus 1/[pNPP]

    revealed the competitive pattern of inhibition, which indicates that C3 binds

    to the active site of the enzyme.

  • 45

    Figure 2-9: Lineweaver-Burk Plot Analysis for PTP1B-catalyzed Reaction

    in Presence of C3: Hydrolysis of pNPP by PTP1B was measured in absence

    (○), or presence of 10 μM (●), 20 μM (□) and 30 μM (■) C3.

    We successfully developed a simple and efficient library synthesis protocol

    for the condensation of aldehydes and isoxazol-5(4H)-one derivatives. The

    Knoevenagel condensation was accomplished in the absence of catalyst. By

    employing this library synthesis protocol, a number of isoxazol-5(4H)-one

    molecule were synthesized and assayed without purification against enzymes.

    The IC50 value obtained from crude products and purified products were

    similar. The crude products were TLC pure and all the NMR data of the

  • 46

    randomly selected compounds were essentially same before and after

    purification. These result distinctly verified our library synthesis protocol.

    The in vitro result revealed that, the compound C3 was the best inhibitor

    among the 70 library compounds, exhibiting the lowest IC50 value of 2.3 μM

    against PTP1B.

    2.2.6. Animal experiment

    2.2.6.1. Toxicity test of compounds C3 on mice

    The most potent PTP1B inhibitor C3 was selected for the evaluation of in

    vivo efficacy as anti-obesity and/or hypoglycemic agent, to avoid the risk of

    animal death. The toxic effect of C3 was tested with 2 male, 5-week-old (30-

    35 g) (SIC: ICR) mice. Two mice were kept as DMSO control.

    2.2.6.1.1. Animal and diet selection for toxicity

    Male, 4-week-old (18-20 g) (SIC: ICR) mice were purchased from Japan

    SLC, OHHARA breeding branch and individually caged at 25 2 C. The

    12 h light/dark cycle was maintained and food and water were supplied ad

    libitum.

    The experimental diets were obtained from Research Diets (New Brunswick,

    NJ). Low fat diet (LFD, D12450B) contained 10% calories from fat, 70%

    calories from carbohydrate and 20% calories from protein. All the foods

    were low fat diet and in pellet form.

    2.2.6.1.2. Feeding

    All six mice (SIC: ICR) were provided with low fat diet during 7 days of

    acclimatization period. Then they were weighed and divided in to two groups.

  • 47

    During the toxicity testing period (6 days), all the mice were deprived of

    food for 6 hours and of water for 2 hours before the drug administration.

    After 1 hour of drug administration, food and water were supplied ad libitum.

    2.2.6.1.3. Drug dose, gavages preparation and administration

    Drug dose for toxicity test was fixed as 400mg of C3/kg body weight of

    mouse for 6 days. To maintain this drug dose, 120 mg of C3 was dissolved

    in 3 mL of DMSO. The DMSO solution of C3 was administrated by needle

    (shaft length 2 inch, ball diameter 21/4

    mm, curved) as, 10 µL/gm body

    weight of mouse i.e. 1 % of body weight of mouse.

    2.2.6.1.4. Effect of compounds C3 on (SIC: ICR) mice

    The mice were found highly stressed for ~6 h of drug administration but

    slowly normalized. The external appearance perineum was found normal the

    next day of the drug administration.

  • 48

    Figure 2-10: Physical appearance of control mouse and C3-treated mouse.

    Table 2-6. Body weight after 6 days of C3 treatment:

    During 6 days of toxicity testing period, the body weight gain was 4 g for the

    DMSO control group, compared to 2 g for the C3-treated group.

    Initial Weight (g) Final Weight (g)

    DMSO Control 34.1± 0.3 38.10±1.9

    DMSO + C3 31.65±0.6 33.73±1.5

  • 49

    2.2.6.1.5. Carcass of the mice at 6 days of C3 treatment

    Figure 2-11: Carcass of the mice after 6 days of C3-treatment

    At the last day of experiment, overnight fasted mice were anaesthetized with

    physical dislocation and the mice were dissected to observe the organs. All

    the organs were found normal on visual inspection except the liver which

    was abnormally brown spotted, in C3- treated mouse.

  • 50

    Figure 2-12: Liver and other organs appearance after 6 days of C3-treatment

  • 51

    2.2.6.1.6. Conclusion: Even though the toxicity test was done in small group

    of normal mice the result revealed that the mice can survive with C3. The

    brown spots were seen in liver may be the side effect of overdose.

    2.2.6.2. Effect of compound C3 on diet induced diabetic mice

    C3 is the most potent inhibitor of PTP1B among the chemical library

    compound. We tested the selectivity and nature of inhibition of compound

    C3. The compound C3 was found ten times selective with TC-PTP and

    competitive inhibitor. The toxicity test of C3 was performed and found to be

    safe for animal experiment. With the reference of these in vitro and in vivo

    results, C3 was chosen for determining their antiobesity or

    antihyperglycemic effect in HFD-induced diabetic mice (C57BL/6J Jms Slc

    male). After 1-week of acclimatization, the mice were divided into three

    groups; two groups of which were fed HFD for further 8 weeks to develop

    the HFD induced diabetes/obesity and the another one group was provided

    with LFD to serve as a lean control group. After 8 weeks on HFD, one of the

    groups was continued on HFD an obese control group whereas the other

    group was provided with HFD + C3 for further 4 weeks.

    2.2.6.2.1. External appearance and body weight gain of mice after

    obesity development period:

    During the obesity development period, body weight gain was 10 g for the

    HFD-fed mouse groups, compared to 5 g for the LFD-fed group. The LFD-

    fed lean control mice gained much lower body weight compared to the HFD-

    fed mice and the two groups were clearly different in external appearances.

    (Figure 2-13 and Table 2-7).

  • 52

    Table 2-7. Body weight gain during obesity development Period

    Initial Weight (g) Final Weight (g)

    HFD-Fed Groups LFD-Fed Groups

    18.00 ±0.12 28.47 ±0.44 23.70 ± 0.29

    Figure 2-13: Appearance of mice after obesity development period

    2.2.6.2.2. Body weight after 4-weeks of C3 Treatment

    No significant difference in body weights between HFD-fed control group

  • 53

    and C3-treated group was observed during and after 4 weeks of the

    compounds treatment period (Table 2-8).

    Table 2-8. Body weight after 4 weeks of C3 treatment:

    Initial Weight (g) Final Weight (g)

    HFD 28.55 ± 1.32 36.05 ±1.56

    HFD + C3 28.18 ±0.81 34.1 ±1.14

    LFD 23.70 ± 0.29 26.26 ±0.41

    2.2.6.2.3. Body weight gain during 4-weeks of C3 treatment

    Figure 2-14: Appearance of mice after drug feeding period

  • 54

    Figure 2-15. Body weight gain during 4 weeks C3 treatment

    C3 (0.1% w/w) in HFD were mixed well and provided with food. Each

    points represents the mean value ± SEM; n = 8/group. Mice provided with

    LFD served as a reference group of lean control. Significance was calculated

    by One-way ANOVA, where, ** represents p < 0.05.

    There was significant difference in body weight gain between HFD control

    group and C3-treated group mice beginning on day 12 and remaining for the

    most of the duration of the study. (Figure 2-15).

    2.2.6.2.4. Food intake

    There was a slight difference in food intake in C3-treated mice group and

  • 55

    LFD control mice group as compare to the HFD control mice group. But,

    there was no significantly difference in cumulative food intake between HFD

    control and C3-treated mice groups during 4 weeks of treatment period.

    (Figure 2-16.)

    Figure 2-16: Cumulative food intake during 4 weeks of C3 treatment

    C3 (0.1% w/w) in HFD were mixed well and provided with food. Each bar

    represents the cumulative mean value ± SEM; n = 8/group

    There was no significantly difference in cumulative food intake between

    HFD control and C3-treated mice groups during 4 weeks of treatment period.

    (Figure 2-16.)

  • 56

    2.2.6.2.5. Feed Efficiency

    The ratio of weight gain to calories consumed, were described in terms of

    feed efficiency. And the body weight gains per calories consumed were

    calculated for the HFD control, C3-treated and LFD control groups. (Figure

    2-17). Feed efficiency of C3-treated group was significantly lower compared

    to HFD control group. This result revealed that, the body weight gain in C3-

    treated group was due to high energy expenditure.

    Figure 2-17. Feed efficiency in mice: Significance was calculated by One-

    way ANOVA, where, ** represents p < 0.05.

    2.2.6.2.6. Intraperitoneal Glucose Tolerance Test (IPGTT)

    At the end of 4 weeks of C3-feeding period, fasting blood glucose level of

  • 57

    all the groups was measured i.e. 0 min (Figure 2-18). Then extra glucose

    was loaded by i.p. injection.

    Figure 2-18. Intraperitoneal glucose tolerance test (IPGTT)

    Mice were fasted for 8 hrs starting from the beginning of light cycle. Just

    before the intraperitoneal injection of glucose (1 g/kg BW), the blood was

    withdrawn from the tip of the tail and base line glucose level was measured.

    Then, after injection of the glucose, blood glucose levels were measured at

    given times. All values are the mean values ± SEM; n= 8/group. Significance

    was calculated by One-way ANOVA, where, ** represents p < 0.05.

    At the end of the 4 weeks of drug-feeding period, the fasting glucose level of

    the test compounds-fed groups was similar as the HFD control group

  • 58

    (Figure 2-18, 0 min) After injection of extra glucose, there were no any

    significantly faster decrease in blood glucose concentration in C3-treated

    group compared to the HFD control group.

    2.2.6.2.7. Carcass appearance at 4-wks of C3 treatment

    No any visual differences were seen in organs of the C3-treated mice and

    HFD control mice. The fat pad deposits in HFD control groups were seen

    significantly higher compared to HFD + C3 group after dissection of mice at

    the end of experiment (Figure 2-19).

    Figure 2-19. Carcass of mice after C3 treatment

    There was no visual difference in liver of C3-treated mice and control group.

  • 59

    2.2.6.2.8. Liver, kidneys and lungs weights

    Figure 2-20. Effect of C3 in liver, kidneys and lungs weights after 4 weeks

    of treatment: C3 (0.1% w/w) in HFD were mixed well and provided with

    food. All values are the mean values ± SEM; n = 8/group. Mice provided

    with LFD served as a reference group of lean control. Significance was

    calculated by One-way ANOVA.

    The liver weights of C3-treated group of mice were significantly higher

    compared to HFD and LFD control groups. But there was no significant

    difference in lung and kidney weights between HFD control and C3-treated

    mice groups.

  • 60

    2.2.6.2.9. Epididymal and retroperitoneal fat pad weights

    Figure 2-21. Effect of C3 in epididymal and retroperitoneal fat pad weights

    after 4 weeks of treatment

    C3 (0.1% w/w) in HFD were mixed well and provided with food. All values

    are the mean values ± SEM; n = 8/group. Mice provided with LFD served as

    a reference group of lean control. Significance was calculated by One-way

    ANOVA, where, ** represents p < 0.05.

    There was no significant difference in Epididymal fat pad deposition in C3-

    treated mice. But there was significant difference in retroperitoneal fat pad

    deposition in C3 treated mice compared to HFD control (Figure 2-21).

  • 61

    2.2.6.2.10. Blood analysis

  • 62

    Figure 2-22. Effect of C3 in serum concentration of (a) total cholesterol and

    triglyceride; and (b) nefa levels after 4 weeks of treatment

    C3 (0.1% w/w) in HFD were mixed well and provided with food. Data

    presented are the mean values ± SEM; n = 8/group. Mice provided with LFD

    served as a reference group of lean control. Significance was calculated by

    One-way ANOVA, where, ** represents p < 0.05.

    No significant difference of total cholesterol, triglyceride and non-esterified

    free fatty acids were found in serum of the C3-treated mice compared to

    HFD control mice when tested after overnight deprivation of the food

    (Figure 2-22).

  • 63

    2.2.7. Chemical sub-library I compounds

    Compound C3 exhibited highest inhibitory potency against PTP1B among

    the chemical library compounds. We fixed compound C as an aldehyde

    precursor. Eight new isoxazol-5(4H)-one precursors were synthesized.

    Chemical sub-library I compounds were synthesized by using same synthetic

    strategy as library synthesis. And the library compound C7 was hydrolyzed

    to get C3ix compound. Their inhibitory potency was tested against PTP1B.

    Scheme 2-8. Synthesis of substituted 3-phenyl isoxazol-5(4H)-one

    derivatives. Reagents and conditions: (a) Zn/NH4Cl, rt, 1h; (b) MnO2/DCM,

    rt, 24 h, (c) Acetone, Johns’ reagent (d) NH2OH·HCl, pyridine, EtOH, 65

    ºC

  • 64

    Scheme 2-9. Hydrolysis of C7 to C3ix: Reagents and conditions: (a) 1,4-

    dioxane, 1M, NaOH, 100 ºC.

    2.2.7.1. Synthesized chemical sub-library I compounds

    Figure 2-23. Chemical sub-library I compounds

  • 65

    2.2.7.2. Inhibitory potency of chemical sub-library I compounds against

    PTP1B:

    Table 2-9. Inhibitory potency of Chemical sub library I compounds against

    PTP1B

    Compd. Mol. Wt IC50(µM)a Compd. Mol. Wt IC50 (µM)

    a

    C3i 523.38 3.2±0.7 C3vi 455.38 12.5±4.0

    C3ii 523.38 2.7±0.0 C3vii 609.57 1.5±0.2

    C3iii

    C3iv

    C3v

    541.37

    541.37

    541.37

    4.0±0.1

    2.3±0.3

    6.8±0.6

    C3viii

    C3ix

    599.48

    483.39

    0.7±0.1

    18.6±1.1

    aData expressed as the mean standard deviations of two experiments.

    Compound C3vii and C3viii exhibited higher potency compared to other

    compound.

    2.2.8. Chemical sub-library II

    2.2.8.1. Chemical sub-library II compounds

    To cross check the potency trend against PTP1B. Isoxazol-5(4H)-one

    precursor 3vii and 3viii were selected to couple with aldehyde precursors of

    library synthesis and their inhibitory potency was tested against PTP1B.

  • 66

    Figure 2-24. Chemical sub-library II compounds

    2.2.8.2. Inhibitory potency of chemical sub-library II compounds against

    PTP1B

    Percentage of Inhibition of chemical sub-library compounds A3vii-J3vii and

  • 67

    A3viii- J3viii against PTP 1B by 10µM solution.

    Compd. Mol. Wt Inhibition% Compd. Mol. Wt Inhibition%

    A3vii 560.02 98 A3viii 549.92 100

    B3vii 560.02 97 B3viii 549.92 100

    D3vii 526.56 71 D3viii 516.47 75

    E3vii 570.57 80 E3viii 560.48 88

    F3vii 589.64 78 F3viii 579.54 89

    G3vii 579.58 74 G3viii 569.49 70

    H3vii 533.59 83 H3viii 523.50 87

    I3vii 497.52 53 I3viii 487.43 63

    J3vii 549.98 87 J3viii 539.89 99

    Table 2-10. Inhibitory potency of Chemical sub library II compounds against

    PTP1B : IC50 values are shown for the compounds that inhibit >90% PTP1B

    enzyme activity.

  • 68

    Table 2-11. IC50 of chemical sub-library II compounds against PTP1B

    Compd. Mol. Wt IC50(µM)a Compd. Mol. Wt IC50(µM)

    a

    A3vii 560.02 3.3±0.4 A3viii 549.92 1.1±0.0

    B3vii 560.02 4.0±1.3 B3viii 549.92 1.6±0.1

    J3viii 539.89 1.3±0.1

    aData expressed as the mean standard deviations of two experiments.

    2.3. Experimental section

    2.3.1. Materials and methods for chemical synthesis

    1H and

    13C NMR spectra were recorded on a Varian Inova 400 (400 MHz) or

    Varian Vnmrs 400 (400 MHz) spectrometer using CDCl3 or DMSO-d6 as a

    solvent. Chemical shifts are reported in unit of parts per million (ppm) from

    tetramethylsilane with the solvent resonance as the internal standard (CHCl3:

    δ = 7.26 ppm for 1H, CDCl3: δ = 77.0 ppm for

    13C). Data are reported as

    follows: chemical shift, multiplicity (s; singlet, d; doublet, t; triplet, q;

    quartet, m; multiplet, br; broad), coupling constants (Hz) and integration. All

    reactions were monitored by thin-layer chromatography using E. Merck

    silica gel plates (60F-254) pre-coated plates (0.25 mm). TLC visualization

    was done with UV light and/or 5% ethanolic p-anisaldehyde. IR spectra were

    recorded on a Bruker Vertex 80V spectrophotometer. All reagents were

    purchased from Aldrich (St. Louis, U.S.A.), Sigma (St. Louis, U.S.A.), or

    TCI (Tokyo, Japan) and used as received.

  • 69

    2.3.2. Synthesis of library compound precursors

    2.3.2.1. Synthesis of aldehyde precursors

    2.3.2.1.1. General Procedure for the preparation of (A – E, H and K-O)

    A solution of appropriate hydroxybenzaldehyde (5.0 mmol), appropriate

    benzylchlorides and K2CO3 (12 mmol) in dry DMF (10 mL) was heated to

    90ºC. After 3 – 6 h, the reaction mixture was cooled to r.t. and added into a

    mixture of 0.1 M HCl (60 mL) and EtOAc (30 mL). The aqueous layer was

    separated and extracted with EtOAc (30 mL × 2). Combined organic layers

    were washed with H2O (20 mL × 3) and saturated NaCl (20 mL), dried in

    (anh. Na2SO4), filtered and concentrated. The crude product was purified by

    column chromatography on silica gel using Hexane/EtOAc mixture as a

    solvent system.

    2.3.2.1.2. Procedure for the preparation of (F)

    4-methylbenzene-1-sulfonyl chloride (5.5 mmol) and DIEPA (6.5 mmol)

    were used instead of benzylchlorides and K2CO3 and the reaction was carried

    out at 0 ºC to room temperature.

    2.3.2.1.3. Procedure for the preparation of (G)

    A solution of 4-(4-formylphenoxy) benzonitrile (0.447 g, 2.0 mmol), NaN3

    (0.390 g, 6.0 mmol,) and Et3N·HCl (0.823 g 6.0 mmol) in toluene (3 mL)

    was refluxed for 6 h. The reaction mixture was cooled to rt. and poured into

    1 M HCl (25 mL). It was extracted with EtOAc (30 mL × 3). The organic

    layers were combined, washed with 1 M HCl (10 mL × 2) and saturated

    NaCl (20 mL) successively, dried in anh. Na2SO4, filtered, and concentrated.

    The crude product was used as such for next reaction.

  • 70

    4-(2-chlorobenzyloxy)benzaldehyde (A): White solid, 92% yield; Rf = 0.46

    (hexane/ethyl acetate, 4:1); m.p. 54-55 ºC; 1H NMR (200 MHz, CDCl3): δ

    5.31 (s, 2H), 7.15 (d, J = 8.8 Hz, 2H), 7.31-7.37 (m, 2H), 7.45- 7.61 (m, 2H),

    7.91 (d, J = 8.8 Hz, 2H), 9.95 (s, 1H, CHO).

    4-(4-chlorobenzyloxy)benzaldehyde (B): White solid, 89% yield; Rf = 0.35

    (hexane/ethyl acetate, 4:1); m.p. 73-74 ºC; 1H NMR (200 MHz, CDCl3): δ

    5.16 (s, 2H), 7.1 (d, J = 8.8 Hz, 2H), 7.42 (brs, 4H), 7.89 (d, J = 8.8 Hz, 2H),

    9.93 (s, 1H, CHO).

    4-(4-(trifluoromethoxy)benzyloxy)benzaldehyde (C): White solid, 96%

    yield; Rf = 0.46 (hexane/ethyl acetate, 2:1); m.p. 45-46 ºC; 1H NMR (200

    MHz, CDCl3): δ 5.20 (s, 2H), 7.12 (d, J = 8.8 Hz, 2H), 7.31 (d, J = 8.0 Hz,

    2H), 7.52 (d, J = 8.0 Hz, 2H), 7.91 (d, J = 8.8 Hz, 2H), 9.95 (s, 1H, CHO).

    4-(pyridin-3-ylmethoxy)benzaldehyde (D): Light Yellow solid, 55% yield;

    Rf = 0.25 (hexane/ethyl acetate, 1:4); m.p. 71-72 ºC; 1H NMR (200 MHz,

    CDCl3): δ 5.20 (s, 2H), 7.12 (d, J = 7.6 Hz, 2H), 7.36-7.42 (m, 1H), 7.81-

    7.92 (m, 3H), 8.65 (d, J = 4.6 Hz, 1H), 8.74 (brs, 1H), 9.93 ppm (s, 1H,

    CHO).

    4-(4-nitrobenzyloxy)benzaldehyde (E): Light Yellow solid, 53% yield; Rf =

    0.18 (hexane/ethyl acetate, 4:1); m.p. 110-111 ºC; 1H NMR (400 MHz,

    CDCl3): δ 5.27 (s, 2H), 7.09 (d, J = 8.8 Hz, 2H), 7.62 (d, J = 8.4 Hz, 2H),

    7.87 (d, J = 8.8 Hz, 2H), 8.27 (d, J = 8.4 Hz, 2H), 9.90 ppm (s, 1H, CHO).

    4-Tosyloxy-benzaldehyde (F): White solid, 94% yield; m.p. 73-74 ºC; Rf =

    0.33 (hexane/ethyl acetate, 4:1); 1H NMR (200 MHz, CDCl3): δ 2.50 (s, 3H),

    7.21 (d, J = 8.6 Hz, 2H), 7.37 (d, J = 7.8 Hz, 2H), 7.76 (d, J = 7.8 Hz, 2H),

    7.88 (d, J = 8.6 Hz, 2H), 10.01 ppm (s, 1H, CHO).

  • 71

    4-(4-(1H-tetrazol-5-yl)phenoxy)benzaldehyde (G): Cream color solid 93%

    yield; Rf = 0.14 (ethyl acetate/MeOH, 19:1); m.p. 179-180 ºC; 1H NMR (400