195
Ref. code: 25595729320217IWP ANTI-CHOLANGIOCARCINOMA AND TOXICITY EVALUATION OF Atractylodes lancea (Thunb.) DC. AND Kaempferia galanga Linn. HERBAL EXTRACT PREPARATIONS FOR DRUG DEVELOPMENT BY MR. ASMARE AMUAMUTA LIMAENEH A DISSERTATION SUBMITTED IN PARTIAL FULFILLMENT OF THE REQUIREMENTS FOR THE DEGREE OF DOCTOR PHILOSOPHY (BIOCLINICAL SCIENCES) CHULABHORN INTERNATIONAL COLLEGE OF MEDICINE THAMMASAT UNIVERSITY ACADEMIC YEAR 2016 COPYRIGHT OF THAMMASAT UNIVERSITY

Anti-cholangiocarcinoma and toxicity evaluation of ...ethesisarchive.library.tu.ac.th/thesis/2016/TU_2016...6.1 Preparation and qual ity control of the plant extract and CMC oral pharmaceutical

  • Upload
    others

  • View
    8

  • Download
    0

Embed Size (px)

Citation preview

  • Ref. code: 25595729320217IWP

    ANTI-CHOLANGIOCARCINOMA AND TOXICITY

    EVALUATION OF Atractylodes lancea (Thunb.) DC. AND

    Kaempferia galanga Linn. HERBAL EXTRACT

    PREPARATIONS FOR DRUG DEVELOPMENT

    BY

    MR. ASMARE AMUAMUTA LIMAENEH

    A DISSERTATION SUBMITTED IN PARTIAL FULFILLMENT

    OF THE REQUIREMENTS FOR THE DEGREE OF DOCTOR

    PHILOSOPHY (BIOCLINICAL SCIENCES)

    CHULABHORN INTERNATIONAL COLLEGE OF MEDICINE

    THAMMASAT UNIVERSITY

    ACADEMIC YEAR 2016

    COPYRIGHT OF THAMMASAT UNIVERSITY

  • Ref. code: 25595729320217IWP

    ANTI-CHOLANGIOCARCINOMA AND TOXICITY

    EVALUATION OF Atractylodes lancea (Thunb.) DC. AND

    Kaempferia galanga Linn. HERBAL EXTRACT

    PREPARATIONS FOR DRUG DEVELOPMENT

    BY

    MR. ASMARE AMUAMUTA LIMAENEH

    A DISSERTATION SUBMITTED IN PARTIAL FULFILLMENT

    OF THE REQUIREMENTS FOR THE DEGREE OF DOCTOR

    OF PHILOSOPHY (BIOCLINICAL SCIENCES)

    CHULABHORN INTERNATIONAL COLLEGE OF MEDICINE

    THAMMASAT UNIVERSITY

    ACADEMIC YEAR 2016

    COPYRIGHT OF THAMMASAT UNIVERSITY

  • Ref. code: 25595729320217IWP

    (1)

    Dissertation Title

    ANTI-CHOLANGIOCARCINOMA AND

    TOXICITY EVALUATION OF Atractylodes

    lancea ( Thunb. ) DC. AND Kaempferia galanga

    Linn. HERBAL EXTRACT PREPARATIONS

    FOR DRUG DEVELOPMENT

    Author

    Mr.Asmare Amuamuta Limaeneh

    Degree

    Doctor of Philosophy (Bioclinical Sciences)

    Major Field/Faculty/University

    Chulabhorn International College of Medicine

    Thammasat University

    Dissertation Advisor

    Professor Kesara Na-Bangchang, PhD

    Academic Year

    2016

    ASTRACT

    Cholangiocarcinoma (CCA) is an important public health problem in

    many tropical and subtropical parts of the world particularly Southeast Asian

    countries. Chemotherapy against CCA is largely ineffective and discovery and

    development of effective chemotherapeutics is urgently needed. The aim of the

    research dissertation was to evaluate the toxicity and anti-CCA activity of the prepared

    CMC (chemistry manufacturing and control) oral formulation of the crude ethanolic

    extract of Atractylodes lancea (Thunb.) DC. rhizomes (AL) for further first-in-human

    clinical evaluation. In addition, the toxicity and anti-CCA activity of the crude

  • Ref. code: 25595729320217IWP

    (2)

    ethanolic extract of Kaempferia galanga Linn. rhizomes (KG) was also investigated.

    Quality control of the CMC oral pharmaceutical formulation of AL rhizome extract

    was performed using HPLC based on the identification of the marker compound

    atracylodin (with content of 1.14 + 0.09 mg/g or 0.114% w/w). Quality control of the

    crude ethanolic extract of KG rhizome was performed using HPLC based on the

    identification of the marker compound ethyl-p- methoxycinnamate (with peak area of

    94.09% of total content of the extract preparation).

    Based on the results of acute and subchronic (90 days repeated oral

    dosing) toxicity evaluation in Wistar rats, the maximum tolerated dose (MTD) of the

    CMC oral pharmaceutical formulation of the AL crude ethanolic extract was at least

    5,000 mg/kg body weight. This formulation can be used in the next step of product

    development for further first-in-human study. Nevertheless, further studies including

    chronic toxicity and genotoxicity of the extract formulation are necessary to ensure

    tolerability of the product in human uses for treatment of CCA. The KG rhizome

    extract exhibited moderate to high cytotoxic activity against human CCA CL-6 cell

    line. Results of the toxicity study in mice (acute and subacute toxicity testing)

    suggested the MTD of the KG extract of 1,000 mg/kg body weight. In CCA (CL-6)-

    xenografted nude mouse model, the KG rhizome extract exhibited significant anti-

    CCA activity at the maximum dose level of 1,000 mg/kg body weight as shown by

    TGI of 58.41%, prolongation of survival time until 62 (53.2-71.8) days, and inhibition

    of lung metastasis by 33% compared with the untreated control mice. Preparation of

    the CMC oral pharmaceutical formulation of the KG rhizome extract is needed before

    further studies in humans. Further studies including subchronic toxicity and other

    pharmacological activities with the extract should be further investigated for

    determining the oral safety dose in an effort for developing an effective drug option

  • Ref. code: 25595729320217IWP

    (3)

    against CCA from this potential herbal agent.

    Keywords: Cholangiocarcinoma, Atractylodes lancea (Thunb.) DC., Kaempferia

    galanga Linn., cytotoxicity, CL-6, toxicity.

  • Ref. code: 25595729320217IWP

    (4)

    ACKNOWLEDGEMENTS

    First of all, I would like to express my special thanks and appreciation to

    my advisor, Prof. Dr. Kesara Na-Bangchang for her generosity, valuable guidance and

    encouragement since the beginning to the end of my study and ultimately made what I

    thought impossible to be possible.

    I also want to express my sincere gratitude for my co-advisor Asst. Prof.

    Dr. Tullayakorn Plengsuriyakarn for his enormous guidance, understanding and

    encouragement throughout my study not only as a mentor but also as a friend.

    I would also like to thank my advisor committee members, Prof. Dr. Juntra

    Karbwang, Asst. Prof. Dr. Wanna Chaijaroenkul, and Dr. Mayuri Tarasuk for their

    valuable guidances.

    My deep appreciation also goes to Dr. Thunyatorn Yimsoo, Dr. Werayut

    Yingmema, Mr. Noppadon Suttirak, and Mr. Noppanan Kotsaouppara and all other

    staff members there for their professional guidance and compassionate help during my

    long time work and study at the Laboratory Animal Center, Thammasat University.

    I wish to express my sincere gratitude also to Thammasat University (TU)

    particularly Chulabhorn International College of Medicine (CICM) and Center of

    Excellence in Pharmacology and Molecular Biology of Malaria and

    Cholangiocarcinoma, National Research University Project of Thailand (NRU) and

    Office of Higher Education Commission of Thailand for funding me the scholarship

    grants and support for the completion of this study.

    I would also like to thank my home University (Bahir Dar University,

    College of Medicine and Heath Sciences) for allowing me to pursue my Ph.D. study

  • Ref. code: 25595729320217IWP

    (5)

    leave and all the favorable supports and encouragement I received from it throughout.

    CICM staff members, my classmates, TU community and all Thai

    individuals I met also deserve my appreciation and heartfelt thanks for every kind of

    support and lessons I gained from them during my study and stay in Thailand.

    I would like to owe a special thanks to my dear father Mr. Amuamuta

    Limeneh and my dear mother Mrs. Tangut Birhanae and also my gratitude to my

    entire family (brothers, sisters and relatives), who have made valuable support, love,

    and prayers throughout my entire life that makes me constantly feel alive. Helen

    Endale, my love partner, also deserve my special thanks for her best wishes,

    encouragement and trust on me.

    Finally, it is my wish to forward my appreciation and gratitude to all of

    my friends and anyone involved and contributed for the success of this work and to

    my life.

    Mr. Asmare Amuamuta Limeneh

  • Ref. code: 25595729320217IWP

    (6)

    TABLE OF CONTENTS

    Page

    ABSTRACT (1)

    ACKNOWLEDGEMENTS (4)

    TABLE OF CONTENTS (6)

    LIST OF TABLES (11)

    LIST OF FIGURES (14)

    LIST OF ABBREVIATIONS (18)

    CHAPTER 1 INTRODUCTION 1

    CHAPTER 2 REVIEW OF LITERATURE 5

    2.1 Cholangiocarcinoma (CCA) and treatment approaches 5

    2.2 Screening methods for development of anticancer and anti-CCA

    drugs and/or phytochemicals

    11

    2.2.1 In vitro methods for screening anticancer compounds 11

    2.2.2 In situ models 12

    2.2.3 In vivo methods for screening anticancer compounds (screening

    using animal models)

    12

    2.3 Medicinal plants with anti-CCA activities 19

    2.4 Description of the study plants 20

    2.4.1 Atractylodes lancea (Thunb.) DC. 20

    2.4.1.1 Plant description and medicinal properties 20

    2.4.1.2 Phytochemistry 23

    2.4.1.3 Safety and toxicity 23

    2.4.2 Kaempferia galanga Linn. 25

    2.4.2.1 Plant description and medicinal properties 25

    2.4.2.2 Phytochemistry 29

  • Ref. code: 25595729320217IWP

    (7)

    2.4.2.3 Safety and toxicity 30

    CHAPTER 3 OBJECTIVES 35

    3.1 General objective 35

    3.2 Specific objectives 35

    CHAPTER 4 RESEARCH METHODOLOGY 37

    4.1. Reagents and cell lines 37

    4.2 Preparation of test materials 37

    4.2.1 Chemistry, manufacturing and control (CMC) oral

    pharmaceutical formulation of Atractylodes lancea (Thunb.) DC.

    37

    4.2.2 Kaempferia galanga Linn. 38

    4.3 Identification of marker compounds 41

    4.3.1 CMC oral pharmaceutical formulation of Atractylodes

    lancea (Thunb.) DC.

    41

    4.3.2 Standardization of the crude ethanolic extract of Kaempferia

    galanga Linn. rhizomes

    42

    4.4 Animal stocks, handling and ethical approval 42

    4.5 Toxicity evaluation of the CMC oral pharmaceutical formulation

    of the crude ethanolic extract of Atractylodes lancea (Thunb.) DC.

    rhizomes

    45

    4.5.1 Acute toxicity evaluation 45

    4.5.2 Subchronic toxicity evaluation 45

    4.6 Cytotoxicity, toxicity testing and anti-CCA activity evaluation of

    the crude ethanolic extract of K. galanga Linn. rhizomes

    47

    4.6.1 Cytotoxic activity evaluation 47

    4.6.2 Toxicity testing of K. galanga Linn. Extract 48

    4.6.3 Anti-CCA activity of K. galanga Linn. extract in CCA-

    xenografted BALB/c nude mice

    50

    4.6.3.1Tumor growth inhibition 53

  • Ref. code: 25595729320217IWP

    (8)

    4.6.3.2 Survival time and metastasis rate 53

    4.7. Data management and analysis 54

    CHAPTER 5 RESULTS 55

    5.1 Preparation of the test materials and identification of marker

    compounds

    55

    5.1.1 CMC oral pharmaceutical formulation of Atractylodes

    lancea (Thunb.) DC. extract

    55

    5.1.2 Standardization of the crude ethanolic extract of Kaempferia

    galanga Linn. rhizomes

    55

    5.2 Toxicity evaluation of the CMC oral pharmaceutical formulation

    of the crude ethanolic extract of Atractylodes lancea (Thunb.) DC.

    rhizomes

    60

    5.2.1 Acute toxicity test 60

    5.2.1.1 Clinical signs, mortality, and body weight changes 60

    5.2.1.2 Autopsy and histopathological findings 60

    5.2.2 Subchronic toxicity test 62

    5.2.2.1 Clinical signs, mortality, and body weight changes 62

    5.2.2.2 Autopsy and histopathology findings 62

    5.2.2.3 Hematology and serum biochemical evaluation 63

    5.3 Cytotoxicity, toxicity testing and anti-CCA activity evaluation for

    the crude ethanolic extract of K. galanga Linn. rhizomes

    69

    5.3.1 Cytotoxic activity the ethanolic extract of K. galanga Linn.

    rhizomes against CL-6

    69

  • Ref. code: 25595729320217IWP

    (9)

    5.3.2 Toxicity evaluation of the ethanolic extract of K. galanga Linn.

    rhizomes

    70

    5.3.2.1 Acute toxicity test 70

    5.3.2.2 Subacute toxicity test 76

    5.3.3 Anti-CCA activity of the crude ethanolic extract of K. galanga

    Linn. in CCA-xenografted BALB/c nude mice

    91

    5.3.3.1 Tumor growth inhibition 91

    5.3.3.2 Survival time and tumor metastasis 91

    CHAPTER 6 DISCUSSION 100

    6.1 Preparation and quality control of the plant extract and CMC oral

    pharmaceutical formulation

    100

    6.1.1 CMC oral pharmaceutical formulation of the crude ethanolic

    extract of A. lancea (Thunb.) DC. rhizomes

    100

    6.1.2 Crude ethanolic extract of K. galanga Linn. 100

    6.2 Toxicity evaluation of the CMC oral pharmaceutical formulation

    of the crude ethanolic extract of A. lancea (Thunb.) DC. rhizomes

    101

    6.3 Cytotoxicity, toxicity testing and anti-CCA activity evaluation of

    the crude ethanolic extract of K. galanga Linn. rhizomes

    102

    6.3.1 Cytotoxic activity evaluation 102

    6.3.2 Toxicity testing of K. galanga Linn. rhizome extract 103

    6.3.3 Anti-CCA activity of the crude ethanolic extract of K. galanga

    Linn. rhizome in CCA-xenografted BALB/c nude mice

    105

    CHAPTER 7 CONCLUSIONS AND RECOMMENDATIONS 107

    7.1 Toxicity evaluation of the CMC oral pharmaceutical formulation 107

  • Ref. code: 25595729320217IWP

    (10)

    of the crude ethanolic extract of A. lancea (Thunb.) DC. rhizomes

    7.2 Cytotoxicity, toxicity testing and anti-CCA activity evaluation of

    the crude ethanolic extract of K. galanga Linn. rhizomes

    107

    REFERENCES 110

    APPENDICES 120

    Appendix A 120

    Appendix B 121

    Appendix C 122

    Appendix D 122

    Appendix E 123

    Appendix F 123

    Appendix G 124

    Appendix H 125

    Appendix I 126

    Appendix J 131

    Appendix K 133

    Appendix L 140

    Appendix M 149

    Appendix N 152

    Appendix O 154

    Appendix P 164

    BIOGRAPHY 168

  • Ref. code: 25595729320217IWP

    (11)

    LIST OF TABLES

    Tables Page

    1. Animal models in current use for screening and development of

    cancer chemo preventive agents

    17

    2. Phytochemical test results detected in different extracts of K.

    galanga Linn. rhizomes

    31

    3. Volatile oil components, retention time and peak area (%) of K.

    galanga Linn. oil

    33

    4. Summary of possible mechanisms of action associated with the

    bioactive constituents of K. galanga Linn. for the specific

    pharmacological activities

    34

    5. Experimental design of the anti-CCA activity evaluation of the

    KG extract in comparison with control groups (vehicle control

    and 5-FU) in CCA-xenografted BALB/c nude mice

    52

    6. Percent yield (w/w) of the crude KG rhizome extract using

    different extraction methods

    58

    7. Haematological parameters of male and female rats in the

    subchronic toxicity evaluation of the AL extract formulation in

    comparison with control (fed with 20% tween-80)

    65

    8. Haematological parameters of female rats in the subchronic

    toxicity evaluation of the AL extract formulation in comparison

    with control (fed with 20% tween-80)

    66

    9. Serum biochemistry parameters of male rats in the subchronic

    toxicity evaluation of the AL extract formulation in comparison

    with control (fed with 20% tween-80)

    67

  • Ref. code: 25595729320217IWP

    (12)

    Tables Page

    10. Serum biochemistry parameters of female rats in the subchronic

    toxicity evaluation of the AL extract formulation in comparison

    with control (fed with 20% tween-80)

    68

    11. Cytotoxic activities expressed as the IC50 values of the crude

    ethanolic extract of KG rhizomes and 5-FU in CL-6 and OUMS

    cell lines at in vito

    71

    12. Vital organ weights of male and female mice following treatment

    with 5,000 mg/kg body weight of the KG extract and control

    group in the acute toxicity evaluation

    74

    13. Survival of mice receiving the oral dose administration of KG

    extract & 20% tween-80 (control group) in the acute and subacute

    toxicity evaluation

    79

    14. The body weights (in grams) of male and female ICR mice in

    subacute toxicity test of the KG extract at the three dose levels,

    i.e.,5,000, 3,000, and 1,000 mg/kg body weight

    81

    15. Median (95% CI) organ weights (in grams) as median (95% CI)

    of mice in subacute toxicity test of the KG extract at 5,000, 3,000,

    and 1,000 mg/kg body weight dose levels

    82

    16. Haematological parameters of male mice receiving the KG extract

    at high (5,000 mg/kg body weight), medium (3,000 mg/kg body

    weight), and low (1,000 mg/kg body weight) and 20% tween-80

    (control) in the subacute toxicity test

    87

    17. Haematological parameters of female mice receiving the KG

    extract at high (5,000 mg/kg body weight), medium (3,000 mg/kg

    body weight), and low (1,000 mg/kg body weight) and 20%

    tween-80 (control) in the subacute toxicity test

    88

  • Ref. code: 25595729320217IWP

    (13)

    Tables Page

    18. Serum biochemistry parameters of male mice receiving the KG

    extract at high (5,000 mg/kg body weight), medium (3,000 mg/kg

    body weight), and low (1,000 mg/kg body weight) and 20%

    tween-80 (control) in the subacute toxicity test

    89

    19. Serum biochemistry parameters of female mice receiving the KG

    extract at high (5,000 mg/kg body weight), medium (3,000 mg/kg

    body weight), and low (1,000 mg/kg body weight) and 20%

    tween-80 (control) in the subacute toxicity test

    90

    20. Representative tumors of the CCA (CL-6)-xenografted nude mice

    following treatment with KG extract at low (100 mg/kg body

    weight), medium (500 mg/kg body weight), and high (1,000

    mg/kg body weight) dose levels, 5-FU (reference control: 40

    mg/kg body beight) and 20% tween-80 (untreated control)

    95

    21. Representative tumor metastases at autopsy of the CCA (CL-6)-

    xenografted nude mice following treatment with KG extract at

    low (1,000 mg/kg body weight), medium (3,000 mg/kg body

    weight), and high (5,000 mg/kg body weight) dose levels, 5-FU

    (reference control: 40 mg/kg body weight) and 20% tween-80

    (untreated control)

    98

    22. Summary of the comparison of toxicity and anti-CCA activity

    findings for AL rhizome extract and its CMC oral pharmaceutical

    formulation

    109

  • Ref. code: 25595729320217IWP

    (14)

    LIST OF FIGURES

    Figures Page

    1. Worldwide annual incidence (per 100,000) of liver cancer in

    males (1995)

    6

    2. Age-standardized incidence rates of liver cancers world wide per

    100,000 in males

    8

    3. Representative some liver fluke (O. viverrini) recovered

    specimens

    9

    4. Life cycle of O. viverrini (A), and the freshwater fish species,

    Puntius orphoides (B)

    10

    5. General scheme for uptake and pharmacokinetics of dietary

    phytochemicals

    13

    6. Contemporary preclinical drug development cascade 14

    7. Screening procedure using patient derived tumors for the

    establishment of in vitro and in vivo models

    18

    8. Atracylodes lancea (Thunb.) DC. leaves (a) and its rhizomes (b) 22

    9. The chemical structures of major components of A. lancea

    (Thunb.) DC. rhizome extract

    24

    10. (A) Flower, (B) fresh rhizomes, (C) dried rhizomes, and (D)

    sprouting plant from rhizomes of K. galanga Linn .

    28

    11. Chemical structures of some important phytoconstituents

    isolated from K. galanga Linn. extract

    32

    12. A diagram showing extract preparation using 7 day maceration of

    the KG rhizomes

    40

  • Ref. code: 25595729320217IWP

    (15)

    Figures

    13. Overall schematic study design of the toxicity testing and anti-

    CCA evaluation of the crude ethanolic extract of K. galanga Linn.

    rhizomes (KG) and CMC oral pharmaceutical formulation of the

    crude ethanolic extract of A. lancea (Thunb.) DC. rhizomes (AL).

    Page

    44

    14. HPLC chromatograms of (A) A. lancea (Thunb.) DC. ethanolic

    extract and (B) atractylodin standard

    56

    15. Characteristic appearance of the KG ethanolic extract after

    concentration

    57

    16. HPLC chromatograms of (A) standard ethyl-p-

    methoxycinnamate (EPMC) and (B) the ethanolic extract of KG

    rhizomes

    59

    17. Median body weight (g) in male and female rats (n=10 for each

    group) following a single oral dose of the CMC oral formulation

    of AL rhizome extract at the highest dose level of 5,000 mg/kg

    body weight

    61

    18. Median body weight changes (in grams) of (A) male and (B)

    female rats following repeated 90 days oral dosing of the CMC

    formulation of the AL rhizome extract at 5,000, 3,000, 1,000

    mg/kg body weight and 20% tween-80 (control group)

    64

    19. Changes in body weights (g) of male and female mice (n=10 for

    each group) following a single oral dose of 5,000 mg/kg body

    weight of the KG rhizome extract and 20% tween-80 (control

    group) for acute toxicity evaluation

    20. The net body weight gains (in grams) of male (A) and female (B)

    mice on the 15th day of treatment with KG at the highest dose of

    5,000mg/kg body weight and 20% tween-80 (control group) in

    the acute toxicity evaluation

    72

    73

  • Ref. code: 25595729320217IWP

    (16)

    Figures Page

    21. Representative haematoxylin-eosin staining of various vital

    organs (liver, spleen, heart, lungs, and kidneys) collected at

    autopsy from mice (males and females) treated with the KG

    extract at the high dose level (5,000 mg/kg body weight) and 20%

    tween-80 (untreated control mice) in the acute toxicity evaluation

    75

    22. Time to death (days) and survival curves for male and female

    mice during subacute toxicity evaluation of the KG extract

    80

    23. Representative haematoxylin-eosin staining of various vital organs

    and tissues collected at autopsy from male mice in the KG-treated

    mice at high (5,000 mg/kg body weight), medium (3,000 mg/kg

    body weight), and low (1,000 mg/kg body weight) doses and 20%

    tween-80 (control) in the subacute toxicity testing

    84

    24. Representative haematoxylin-eosin staining of various vital

    organs and tissues collected at autopsy from female mice in the

    KG-treated mice at high (5,000 mg/kg body weight), medium

    (3,000 mg/kg body weight), and low (1,000 mg/kg body weight)

    doses and 20% tween-80 (control) in the subacute toxicity testing

    86

    25. Anti-CCA activity and tumor volume progression (mm3) of the

    CCA (CL6)-xenografted nude mice following treatment with the

    KG extract at low (100 mg/kg body weight), medium (500 mg/kg

    body weight), and high (1,000 mg/kg body weight) dose levels, 5-

    FU (reference control: 40 mg/kg body weight) and 20% tween-80

    (untreated control) during the investigation period

    93

  • Ref. code: 25595729320217IWP

    (17)

    Figures Page

    26. Tumor volumes (mm3) of the CCA (CL-6)-xenografted nude mice

    at the end of treatment (on day 31st) with KG extract at low (100

    mg/kg body weight), medium (500 mg/kg body weight), and

    high (1,000 mg/kg body weight) dose levels, 5-FU (reference

    control: 40 mg/kg body weight) and 20% tween-80 (untreated

    control)

    94

    27. Median survival time (days) of the CCA (CL-6)-xenografted nude

    mice following treatment with the KG extract at low (1,000

    mg/kg body weight), medium (3,000 mg/kg body weight), and

    high (5,000 mg/kg body weight) dose levels, 5-FU (reference

    control: 40 mg/kg body weight) and 20% tween-80 (untreated

    control) during the investigation period

    96

    28. Gross appearance of the representative primary CCA tumor at

    autopsy in a CCA (CL-6)-xenografted nude mouse

    97

    29. Representative primary CCA tumors and lung metastases at

    autopsy of the CCA (CL-6)-xenografted nude mice following

    treatment with KG extract at low (1,000 mg/kg body weight),

    medium (3,000 mg/kg body weight), and high (5,000 mg/kg body

    weight) dose levels, 5-FU(reference control: 40 mg/kg body

    weight) and 20% tween-80 (untreated control)

    99

  • Ref. code: 25595729320217IWP

    (18)

    LIST OF ABBREVIATIONS

    Symbols/Abbreviations Terms

    AL Atractylodes lancea (Thunb.) D.C.

    ALP

    ALT

    AST

    AVMA

    Alkaline phosphatase

    Alanine aminotransferase

    Aspartate aminotransferase

    American Veterinary Medical Association

    BUN

    BW

    Blood urea nitrogen

    Body weight

    oC

    CA19-9

    CAM

    Degree celsius

    Carbohydrate antigen 19-9

    Complementary and alternative medicine

    CCA Cholangiocarcinoma

    CEA

    CI

    CICM

    Carcinoembryonic antigen

    Confidence interval

    Chulabhorn International College of

    Medicine

    CL-6 Cholangiocarcinoma cell line

  • Ref. code: 25595729320217IWP

    (19)

    CNS

    CO2

    COX

    CYP450

    Central nervous system

    Carbon dioxide

    Cyclooxygenase

    Cytochrome P450

    dH2O Distilled water

    DMEM

    DMN

    DMSO

    EC

    EDTA

    EPMC

    Dulbecco's Modified Eagle's Medium

    Dimethyl nitrosamine

    Dimethyl sulfoxide

    Ethyl cinnamate

    Ethylene diamine tetraacetic acid

    Ethyl-p-methoxycinnamate

    FBS Fetal bovine serum

    FDA Food and Drug Administration

    5-FU

    GAE

    g/dl

    H

    Hb

    5-Fluorouracil

    Galic acid equivalent

    Gram per deciliter

    Hour

    Hemoglobin

  • Ref. code: 25595729320217IWP

    (20)

    HCC

    HCF

    HCT

    H & E

    HDL

    HepG2

    Hepatocellular carcinoma

    Human cardiac fibroblast

    Hematocrit

    Hematoxylin and eosin

    High density lipoproteins

    Hepatocarcinoma cell line

    HPLC

    HTS

    High performance liquid chromatography

    High through-put assays

    IC50

    ICR

    Inhibitory concentration by 50%

    Imprinting control region

    ID50 Inhibitory dilution by 50%

    KG Kaempferia galanga Linn.

    LD50

    LDL

    Lethal dose concentration by 50%

    Low density lipoproteins

    µg

    µl

    MCV

    MCH

    MCHC

    Microgram

    Microliter

    Mean corpuscular volume

    Mean corpuscular hemoglobin

    Mean corpuscular hemoglobin

    concentration

  • Ref. code: 25595729320217IWP

    (21)

    mEq/L Milliequivalent per liter

    mm Millimeter

    MPV

    MST

    Mean platelet volume

    Mean survival time

    MTD Maximum tolerated dose

    MTT

    NBF

    Nm

    Methyl thiazoldiphenyl tetrazolium

    Neutral buffered formalin

    Nanometer

    NOAEL

    NSS

    No observed adverse effect level

    Normal saline solution

    OD Optical density

    OECD

    Organization for Economic Co-operation

    and Development

    % Percent

    PBS

    PCT

    PDW

    Pg

    PK

    PD

    Phosphate buffered saline

    Plateletcrit

    Platelet distribution width

    Picogram

    Pharmacokinetics

    Pharmacodynamics

  • Ref. code: 25595729320217IWP

    (22)

    RBC

    RDW

    Red blood cell (s)

    Red cell distribution width

    Rpm Revolutions per minute

    RPMI

    SC

    SD

    SE

    Roswell Park Memorial Institute

    Subcutaneous

    Standard deviation

    Standard error

    TGI

    TM

    TNF-α

    TPC

    TU

    Tumor growth inhibition

    Traditional medicine

    Tumor necrosis factor-α

    Total phenolic content

    Thammasat University

    TV

    U/l

    Tumor volume

    International unites per liter

    v/v

    WBC

    w/w

    WHO

    Volume by volume

    White blood cell (s)

    Weight by weight

    World Health Organization

  • Ref. code: 25595729320217IWP

    1

    CHAPTER 1

    INTRODUCTION

    Cholangiocarcinoma (CCA) is an important public health problem in

    several parts of the world and in particular in Asia including Thailand. CCA or

    adenocarcinoma of the bile ducts arises from the epithelial cells of bile ducts

    anywhere along the intrahepatic and extra hepatic biliary tree excluding the papilla of

    Vater and the gall bladder (Mosconia et al., 2009). The highest prevalence of CCA in

    Northeast of Thailand is associated to the consumption of improperly cooked and

    fermented or preserved cyprinid fish species food which contains the fluke,

    Opisthorchis viverrini (Dholwani et al., 2008; Haswell-Elkins et al., 1992;

    Khuhaprema and Srivatanakul, 2008; Singh and Facciuto, 2012). In other Asian

    countries like China, the parasite known as Clonorchis sinensis is the main cause of

    CCA (WHO, 2003).

    The major challenge for CCA control and treatment is the lack of early

    diagnosis and multidrug or radio-resistant nature of CCA tumor (Na-Bangchang and

    Karbwang, 2014; Namwat et al., 2008). This requires an ongoing and urgent need for

    the discovery and development of effective alternative diagnostic tools and

    chemotherapeutics. Even though the clinical response rate is low and the recurrence

    rate is extremely high, surgical resection of detectable tumors and combination

    therapy with standard chemotherapeutic agents including 5-fluorouracil (5-FU) leads

    to an improvement in the 5-year survival rate in the present therapeutic approaches

    (Lee et al., 2004; Prabhleen and Todd, 2005). The promising therapeutic options in

    many types of cancers including CCA is the use of combination therapies of standard

    treatments and in conjunction with alternative therapy with dietary phytochemicals

    (Vapiwala et al., 2006). There is a pressing need of drug discovery research for new

    alternative drugs against cancers in general and CCA in particular.

    Plants have formed the basis of traditional medicine (TM) systems in

    many parts of the world which have been used for thousands of years and the use of

    plant-based therapeutic systems continues to play an essential role in health care

  • Ref. code: 25595729320217IWP

    2

    system (Zia-Ul-Haq et al., 2013). The use of plant-derived material or preparation

    with therapeutic or other human health benefits which contains either raw or

    processed ingredients of one or more plants and in some conditions, materials of

    inorganic or animal origin may also be added is referred as herbal medicine (WHO,

    1993). Approximately 80 % of the population in developing countries rely on TM for

    their primary health care (Hostesttmann and Marston, 2002; Zia-Ul-Haq et al., 2013).

    In China, the use of traditional herbal preparations account for 30 to 50% of the

    medicines (WHO, 2003). In developed countries, therapeutic approaches of TM also

    known as complementary and alternative medicine (CAM), also plays an important

    role in the health care system of 20% of the population. In addition to such therapeutic

    options, TM has also afforded a rich source of remedies or drugs with diverse

    chemical structures and bioactivities against several health disorders including cancer

    and several modern anticancer drugs approved by FDA (including vinblastine,

    vincristine, etoposide, teniposide, paclitaxel, vinorelbine, docetaxel, topotecan, and

    irinotecan) have been developed from plant sources (Dholwani et al.,2008; Na-

    Bangchang and Karbwang, 2014; Schwikkard and van Heerden, 2002; Sudhanshu et

    al., 2003).

    Thailand is a country which is rich in a wide range of tropical habitats,

    remarkable biodiversity, and uses traditional medicines for treatment of various

    infectious illnesses and chronic diseases including cancer (Subchareon, 1998;

    Thiengsusuk et al., 2013). Ethnopharmacological studies of many candidate medicinal

    plants or herbal formulations commonly used in Thai TM practice for treatment of

    various diseases have been screened for their anti-CCA activities (Mahavorasirikul et

    al., 2010; Na-Bangchang and Karbwang, 2014; Plengsuriyakarn et al., 2012).The

    extracts from seven plant species (Atractylodes lancea (Thunb.) DC., Kaempferia

    galanga Linn., Zingiber officinale Roscoe., Piper chaba Linn., Mesua ferrea Linn.,

    Ligusticum sinense Oliv. cv. Chuanxiong, Mimusops elengi Linn.) and one folklore

    recipe (Pra-Sa-Prao-Yhai) exhibited promising activity against CL-6 cell line with

    IC50 (concentration that inhibits cell growth by 50%) values reported less than

    50μg/ml (Mahavorasirikul et al., 2010). Among the above extract and recipe

    preparations (including Atractylodes lancea (Thunb.) DC., Kaempferia galanga Linn.,

  • Ref. code: 25595729320217IWP

    3

    Zingiber officinale Roscoe., Piper chaba Linn., Mesua ferrea Linn., and Pra-Sa-Prao-

    Yhai recipe), potent cytotoxic activity were found with mean IC50 values of 24.09,

    37.36, 34.26, 40.74, 48.23 and 44.12 μg/ml, respectively. The crude ethanol extract

    from A. lancea (Thunb.) DC. showed a potent activity against CL-6 cell lines (IC50 =

    24.09 + 3.40 μg/ml and selectivity index = 8.6) and HepG2 cell line (IC50 = 76.68 ±

    15.94 and SI = 2.7) at in vitro (Mahavorasirikul et al., 2010). In a similar protocol

    (Mahavorasirikul et al., 2010), K. galanga Linn. extract showed cytotoxic activity

    against CL-6 cell lines (mean + SD IC50 = 37.36 ± 3.98μg/ml and selectivity index =

    2.9) and against HepG2 cell line (mean + SD IC50 =115.47 ± 26.23 and SI= 0.9).

    Moreover, the anti-CCA activity and safety of the crude ethanolic extract of A. lancea

    (Thunb.) DC. rhizomes was also demonstrated CCA-xenografted nude mice at

    different dose levels (1,000, 3,000, and 5,000 mg/kg body weight) (Plengsuriyakarn et

    al., 2012) that showed significant reduction in tumor size, prolongation of survival

    time, and inhibition of lung metastasis as compared with the standard drug (5-FU) and

    the untreated control.

    Upon reviewing the current literature, there has been little evidence or

    report found especially on the in vivo toxicity and anti-CCA activity of K. galanga

    Linn.. On the other hand, previous in vitro and in vivo studies conducted on the crude

    ethanol extract of A. lancea (Thunb.) DC. rhizomes indicated its potential role for

    treatment of CCA and its safety as described above (Mahavorasirikul et al., 2010;

    Plengsuriyakarn et al., 2012). In hamster model, the ethanolic extract of A. lancea

    (Thunb.) DC. showed satisfactory safety and a significant anti-CCA activity

    particularly at the highest dose level of 5,000 mg/kg body weight compared with the

    standard drug 5-FU and untreated control animal groups (Plengsuriyakarn et al.,

    2015). Moreover, the in vivo anti-CCA study (Plengsuriyakarn et al., 2015) conducted

    on CCA-induced mice using PET-CT imaging following treatment with high dose β-

    eudesmol (100 mg/kg body weight for 30 days), the bioactive compound isolated

    from A. lancea (Thumb.) DC. rhizomes showed significant reduction in tumor size

    and lung metastasis. The survival time of mice was prolonged by 64.4% compared

    with the untreated control. Based on such preliminary screening results (in vitro and

  • Ref. code: 25595729320217IWP

    4

    in vivo) of various plants used in traditional medicine against CCA in Asia, including

    Thailand, there is a need to select and further study the toxicity (safety) and efficacy

    of some promising candidate herbal agent preparations including A. lancea (Thunb.)

    DC. formulation and K. galanga Linn. extract in an effort for new drug discovery and

    development option.

  • Ref. code: 25595729320217IWP

    5

    CHAPTER 2

    REVIEW OF LITERATURE

    2.1. Cholangiocarcinoma (CCA) and treatment approaches

    Hepatocellular carcinoma (HCC) is the most frequent and important of

    liver cancers which poses a significant disease burden in many parts of the world,

    particularly in Africa and Asia (WHO, 2003). In high incidence regions or countries

    of HCC, chronic infection with hepatitis B virus (HBV) is its principal underlying

    cause and in countries like Japan, it’s associated with high prevalence of hepatitis C

    infection. On the other hand, liver cirrhosis associated to chronic alcohol abuse is a

    major cause of HCC in the Western or developed countries. Unlike HCC,

    cholangiocarcinoma (CCA) has a different geographical distribution, with peak

    incidences in Asia especially in the Northeast of Thailand (WHO, 2003).

    CCA is classified into three major groups, i.e., intrahepatic, perihilar, and

    distal extra hepatic and generally CCA accounts for approximately 15% of liver

    cancers worldwide (Nakanuma et al., 2003). Intrahepatic type of CCA arises from any

    portion of the intrahepatic bile duct epithelium or intrahepatic small bile ducts, as well

    as from the right and left hepatic ducts at or near their junction called hilar CCA

    which is considered an extrahepatic lesion (Nakanuma et al., 2003). Intrahepatic CCA

    is the most common case of CCA in Thailand and the disease in the area has been

    definitely related to chronic infestation with Opisthorchis viverrini (O. viverrini)

    (Sripa et al., 2011; Watanapa and Watanapa, 2002). According to the World Health

    Organization reports in 1994 (WHO, 1994), the age standardized incidence of

    intrahepatic CCA in Khonkaen region of Thailand was 88 per 100,000 in males and

    37 per 100,000 in females (Figure 2). Approximately 90% of the confirmed cases of

    liver cancers were intrahepatic CCA, and almost all the CCA cases were found to be

    related to chronic O. viverrini infection in the area of this study.

  • Ref. code: 25595729320217IWP

    6

    Figure 1 Worldwide annual incidence (per 100,000) of liver cancer in males (1995).

    Numbers on the map indicate regional average values (WHO, 2003).

  • Ref. code: 25595729320217IWP

    7

    O. viverrini (OV) infections is pathologically associated to a number of

    hepatobiliary diseases including cholangitis, obstructive jaundice, hepatomegaly,

    cholecystitis, and cholelithiasis are associated with (Harinasuta et al., 1984). Both

    experimental and epidemiological evidences strongly indicated that chronic infection

    with OV liver fluke as the etiology of CCA (Elkins et al., 1996; Haswell-Elkins et al.,

    1992). Moreover, the pathological consequences of OV infection (i.e., epithelial

    desquamation, inflammation, epithelial hyperplasia, goblet cell metaplasia,

    adenomatous hyperplasia, and periductal fibrosis) appear to be similar in both human

    and animals (Riganti et al., 1989).

    In addition to limitations of therapeutic options, early detection of CCA is

    also a major problem for controlling CCA. Among the serum tumor markers,

    carcinoembryonic antigen (CEA) and carbohydrate antigen 19-9 (CA19-9) are used as

    candidate biomarkers for assessment and monitoring during and after the treatment of

    several gastrointestinal cancers. At present, surgical resection of detectable tumors

    leads to an improvement in the 5-year survival rate and additional chemotherapy with

    standard drugs including 5- fluorouracil (5-FU) has been indicated to improve local

    control and prolong survival time of patients (Prabhleen and Todd, 2005). However,

    the recurrence rate is extremely high, with a 5-year survival rate of less than 40%

    even with operable tumors (Sirica, 2005; Thongprasert, 2005). Other reports also

    indicated that chemotherapeutic treatment of CCA is largely ineffective; and

    treatment with 5- FU always produces low clinical response rates (Lee et al., 2004;

    Patt et al., 2001; Thongprasert, 2005). Moreover, in severe stages of CCA, metastatic

    tumors in the lungs develop by spreading from the liver origin through the

    bloodstream or lymphatic system, which is one of the main determinants of therapy

    and patients with such metastatic disease (inoperable CCA) should be considered for

    systemic chemotherapy with gemcitabine and cisplatin (Namwat et al., 2008).

    Generally, CCA is considered to be a multidrug and radio-resistant tumor that still

    requires new approaches of treatments.

  • Ref. code: 25595729320217IWP

    8

    Figure 2 Age-standardized incidence rates of liver cancers worldwide per 100,000 in

    males (WHO, 1994).

  • Ref. code: 25595729320217IWP

    9

    Figure 3 Representative liver fluke (Opisthorchis viverrini) recovered specimens.

    (A) An adult specimen of O. viverrini recovered from a patient after chemotherapy

    and purgation, (B) A metacercaria of O. viverrini detected in a freshwater fish Puntius

    orphoides, and (C) A young adult of O. viverrini recovered after an experimentally

    infected hamster with the metacercariae in Cambodia (Sohn et al., 2012).

  • Ref. code: 25595729320217IWP

    10

    (A)

    (B)

    Figure 4 Life cycle of (A) O. viverrini, and (B) the freshwater fish species Puntius

    orphoides (Sohn et al., 2012).

  • Ref. code: 25595729320217IWP

    11

    The growing trend and promising activity to improve survival of cancer

    patients including CCA, is to combine conventional therapy such as advanced surgical

    techniques in combination with chemotherapy and alternative chemotherapeutic

    options from dietary phytochemicals (Vapiwala et al., 2006). In general,

    chemotherapy with plant-derived compounds or dietary phytochemicals has emerged

    as an accessible and alternative promising approach to cancer control and

    management in many countries (Surh, 2003). In such efforts to develop new and

    alternative drugs against CCA and other cancers from natural products, an ongoing

    preclinical research is required and should be conducted to evaluate the safety,

    efficacy and essential pharmacokinetic aspects of the test substances.

    2. 2. Screening methods for development of anticancer and anti-CCA drugs

    and/or phytochemicals

    To combat the problem of resistance, newer drugs are urgently required

    and an unprecedented number of anticancer discovery and development projects are

    now underway throughout the world. Its recognized that most of the methods for

    anticancer screening and investigating the principal mechanisms of liberation,

    absorption, distribution, metabolism and elimination (LADME) have originated from

    the pharmaceutical research/new drug development sector, but the same procedure

    may be applied and optimized for phytochemicals as well (Birgit and Gary, 2004).

    Several methods including in vitro, in situ and in vivo screening bioassay methods

    described below are routinely used for anticancer drug development evaluations

    (Birgit and Gary, 2004).

    2.2.1 In vitro methods for screening anticancer candidates

    Researchers use cell-free and cell-based screenings (e.g. artificial

    membranes, cells, tissues, and organ cultures) to study a broad range of endogenous

    processes outside the body under controlled and standard conditions (Birgit and Gary,

    2004). In the majority of cases, either cellular or target-based high-throughput assays

    (HTS) are performed before in vivo evaluation of potential anticancer drugs/test

  • Ref. code: 25595729320217IWP

    12

    compounds. Permanent human tumor cell lines (their immortal nature and

    reproducible growth behavior) makes cell-based screening assays in cancer research

    are suitable test systems (Birgit and Gary, 2004). The cell-based assay formats can be

    performed in 96- to 384-well plates but the detection method (which could depend on

    the cell number used and desired sensitivity) is of critical importance and the earliest

    broadly used detection method (growth inhibition assay) is the methyl thiazoldiphenyl

    tetrazolium (MTT) assay (Mosmann, 1983).

    2.2.2 In situ models

    In situ models such as continuous, single-pass perfusion in animals, and

    humans, provide a powerful research tool for the investigation of intestinal transport

    and metabolism of test compounds (Birgit and Gary, 2004).

    2.2.3 In vivo methods for screening anticancer compounds (screening

    using animal models)

    It is difficult to study complex interactions in simplistic models like in

    vitro and only in vivo models provide the opportunity to integrate the dynamic

    components of the mesenteric blood circulation, the mucous layer, and all other

    factors that can influence ADME (absorption, distribution, metabolism, and excretion)

    and the subsequent pharmacodynamic effects (Birgit and Gary, 2004) (Figure 5).

    In general, compounds effective in in vitro screening tests are taken up for

    in vivo evaluation with the appropriate animal models/organisms (Figure 6). Despite

    the fact that the in vivo methods are expensive and time consuming, rodent xenograft

    models are the current traditional preclinical test-bed and form an excellent model for

    initial basic pharmacologic research and safety investigations (Malaney et al, 2014;

    Peldschus and Ittrich, 2014) and currently even for clinical therapeutic response

    studies (Burger and Fiebig, 2014).

  • Ref. code: 25595729320217IWP

    13

    Figure 5 General scheme for uptake and pharmacokinetics of dietary phytochemicals

    (Birgit and Gary, 2004).

  • Ref. code: 25595729320217IWP

    14

    Figure 6 Contemporary preclinical drug development cascade (Burger and Fiebig,

    2014; Rudek et al., 2014).

  • Ref. code: 25595729320217IWP

    15

    Animal based (in vivo) bioassays afford a strategic framework for

    evaluating evidence of agent efficacy, and also serve to generate valuable dose-

    response, toxicity, and pharmacokinetic data which are required prior to phase I

    clinical safety testing (Steele et al., 2005). It has been indicated that some key

    elements are necessary in choosing the ideal animal model for cancer chemotherapy

    or chemoprevention testing; (1) the animal model should bear relevance to human

    cancers (including in terms of specific organ sites and in producing cancerous lesions

    of similar pathology); (2) the animal model should have relevant intermediate lesions

    that simulate the human cancer process both histologically and molecularly; (3) the

    genetic abnormalities of these lesions should resemble those of humans; (4) the model

    should be capable of producing a consistent tumor burden of greater than 80% lesions

    within a reasonable period of time (less than 6 months); and (5) the carcinogen or

    genetic defect used to produce cancer should bear relevance to that encountered by

    humans (Steele et al., 2005).

    Research and development of better animal models is ongoing, although

    no current animal model is considered ideal or best (Steele et al., 2005) and a review

    of some of the currently used animal models for cancer chemoprevention efficacy

    testing is presented in Table 1.

    The Freiburg Experience: The Freiburg xenograft panel is derived

    directly from patient explants instead of established from permanent human tumor

    cell line material (Figure 7) (Fiebig et al., 2004). In this panel, comparison of the

    efficacy of a standard drug or drug combinations in patients and their tumors in nude

    mice, a total of 21 patients reached a remission and the same result was observed in

    19 tumors growing as xenografts (Fiebig et al., 2004). Moreover, 59 mice did not

    respond to treatment and the same result was found in 57 mice. Overall, xenografts

    provided a correct prediction for resistance in 97 % (57/59) and for tumor

    responsiveness in 90 % (19/21).

    For ethical and other pragmatic reasons, the most frequently used animal

    models for in vivo profiling is the rat (Kararli, 1995). However, interspecies

    differences between humans and animals would mean that the endpoints of such

  • Ref. code: 25595729320217IWP

    16

    studies may not reflect the human situation. Several factors attributed to differences in

    anatomy and physiology which include gut length and motility, amount and

    composition of gastrointestinal microflora, enterohepatic circulations, as well as

    intestinal transit time may all have profound effects on pharmacokinetic and

    pharmacodynamic activities of a test compound in different species (Birgit and Gary,

    2004).

    In general, at dietary levels, food constituents including phytochemicals

    are recognized as safe. As a result, human studies of dietary phytochemicals at dietary

    doses should always be considered as the method of choice, which provides a great

    advantage over drug studies. The disadvantage and challenge is the complexity of the

    food matrix that needs to be studied to simulate dietary conditions, the low

    concentrations of most phytochemicals in food, and the complexity of the human

    organism. Considering such aspects, it is necessary to address the gaps in knowledge

    on the processes of liberation, absorption, distribution, metabolism, and elimination

    (LADME) and complement human in vivo data with appropriate in vitro data obtained

    by studies on human tissues, cells, or subcellular fractions and animal in vivo data

    (Birgit and Gary, 2004).

  • Ref. code: 25595729320217IWP

    17

    Table 1 Animal models in current use for screening and development of cancer

    chemo preventive agents (Steele et al., 2005).

  • Ref. code: 25595729320217IWP

    18

    Figure 7 Screening procedure using patient derived tumors for the establishment of in

    vitro and in vivo models (Fiebig et al., 2004).

  • Ref. code: 25595729320217IWP

    19

    2.3. Medicinal plants with anti-CCA activities

    Treatment of malignancies by using herbal medicine in Asian countries

    has been used since early times (Efferth et al., 2007) and TM is also commonly used

    as an alternative treatment for cancers by Thai peoples (Subchareon, 1998). It is

    recognized that considerable researches have been performed on these plants to

    treatment of cancers, and some plant products have been marketed as anticancer drugs

    based on the traditional uses and scientific evidences (Mahavorasirikul et al., 2010;

    Na-Bangchang and Karbwang, 2014).

    Numerous cancer research for chemotherapeutic potential of medicinal

    plants have been carried out in an effort to discover new therapeutic agents that lack

    the toxic effects associated with current cancer therapeutic agents and it is known that

    several conventional anticancer drugs approved by the FDA and being used in

    patients with different types of cancers are derived from plants (Dholwani et al.,

    2008). These include vinblastine, vincristine, etoposide, teniposide, paclitaxel,

    vinorelbine, docetaxel, topotecan, and irinotecan. Several Thai traditional folklores

    have been shown to possess anticancer activities in various human cancerous cell

    lines with some promising candidates (Itharat et al., 2004; Mahavorasirikul et al.,

    2010; Prayong et al., 2008).

    Specific ethnobotanical and in vitro investigations of the candidate

    medicinal plants or herbal formulations which are commonly used in Thai traditional

    medicine for their anti-CCA activity screening (Mahavorasirikul et al., 2010). The

    extracts from seven plant species, i.e., Atractylodes lancea (Thunb.) DC., Kaempferia

    galangal Linn., Zingiber officinale, Piper chaba, Mesua ferrea, Ligusticum sinense,

    Mimusops elengi, and one folklore recipe Pra-Sa-Prao-Yhai were shown to exhibit

    promising activities against the human CCA cell line (CL-6) with IC50 (concentration

    that inhibits cell growth by 50%) values of less than 50 μg/ml (Mahavorasirikul et al.,

    2010). The corresponding mean IC50 values for these test agents were 24.09, 37.36,

    34.26, 40.74, 48.23 and 44.12 μg/ml, respectively. The crude ethanol extract from

  • Ref. code: 25595729320217IWP

    20

    Atractylodes lancea (Thunb.) showed the most potent activity with selectivity index

    (SI) of 8.6. In addition, its activity against the cancer cell HepG2 was also potent with

    mean IC50 of 76.68 μg/ml and SI of 2.7 (Mahavorasirikul et al., 2010). Kaempferia

    galanga Linn. extract also showed cytotoxic activity against CL-6 cell line with mean

    IC50 of 37.36 μg/ml and SI of 2.9; its mean IC50 and SI values for the HepG2 cell were

    115.47 μg/ml and 0.9, respectively.

    Another ethnopharmacological study conducted in the United States also

    confirmed the anticancer activity of A. lancea (Thunb.) DC. in murine neuroblastoma

    cells originally derived from a spontaneous malignant tumor and its activity was

    considered moderate to strong with LC50 (50% lethal concentration) of 0.70 mg/ml

    (Mazzio and Soliman, 2010). Moreover, the anti-CCA activity of A. lancea (Thunb.)

    DC. extract and its active compound known as β–eudesmol were also demonstrated at

    in vivo in CCA-xenografted nude mice (Plengsuriyakarn et al., 2012) and in hamster

    model (Plengsuriyakarn et al.,2015). Such results attract researchers’ attentions to

    further investigate the safety anticancer property of both K. galanga Linn. extract and

    A. lancea (Thunb.) DC. extract preparations.

    2.4 Description of the study plants

    2.4.1 Atractylodes lancea (Thunb.) DC.

    2.4.1.1 Plant description and medicinal properties

    Atractylodes lancea (Thunb.) DC., belonging to the Compositae

    Family, also commonly known as Khoad- Kha- Mao (Thailand) and So- Jitsu (Japan), is

    widely distributed in Asia. It is known as “Cang Zhu” in traditional Chinese medicine

    (Florae et al., 2005). It has long been used as crude drugs to treat a lot of diseases

    such as rheumatic diseases, digestive disorders, night blindness, and influenza (Xiao,

    2002).

  • Ref. code: 25595729320217IWP

    21

    A. lancea (Thunb.) DC. has been used as diuretic and gastro-intestinal

    (against stomachic damage and dyspepsia) drugs in Japanese (Kitajima et al., 2003)

    and Korean (Tatsuta and Iishi, 1993) traditional medicines. In Thai traditional

    medicine, the dried rhizome of A. lancea (Thunb.) DC. has been used to treat fever

    and the common cold, and to relieve gastro-intestinal symptoms such as dyspepsia,

    flatulence, nausea, and noninfectious diarrhea (Qian et al., 2006).

    Ethnopharmacological studies also suggest as the A. lancea (Thunb.)

    DC. extract exhibits anti-inflammatory and antimicrobial activities as well as activities

    on the nervous, cardiovascular, and gastro-intestinal systems (Koonrungsesomboon et

    al., 2014). Results of cytotoxic testing in the human CCA cell line CL-6 demonstrated

    potent activity of A. lancea (Thunb.) DC. crude extract (IC50 = 24.09 μg/ml) and most

    selective against CCA (SI= 8.6) (Mahavorasirikul et al., 2010). The crude ethanolic

    extract of A.lancea (Thunb.) DC. and its bioactive compound (β-eudesmol)

    demonstrated significant reduction of tumor size, tumor metastasis inhibition, and

    increased survival time when compared with standard drug 5-FU and untreated

    control in OV/DMN-induced CCA hamster model (Plengsuriyakarn et al., 2015) and

    CCA-xenografted mice (Plengsuriyakarn et al., 2012). The in vivo anti-CCA study

    conducted in CCA-xenografted nude mice indicated significant reduction in tumor

    size and lung metastasis by 91.6% (of baseline) and 95% (of total lung mass),

    respectively following treatment with high dose β-eudesmol (100 mg/kg body weight

    for 30 days), one of the bioactive compounds isolated from A. lancea (Thunb.) DC.,

    by using PET-CT imaging study (Plengsuriyakarn et al., 2015). Moreover, the

    survival time of mice was prolonged by 64.4% compared with the untreated control.

    The pharmacokinetics of the radiolabeled β-eudesmol was also evaluated in healthy

    and CCA-xenografted nude mice showing the systemic clearance of the compound

    was rapid (particularly during the first 60 min) and the compound was distributed to

    the vital organs following oral and intravenous administrations (Plengsuriyakarn et

    al., 2015).

  • Ref. code: 25595729320217IWP

    22

    (A)

    (B)

    Figure 8 (A) Atracylodes lancea (Thunb.) DC. leaves and (B) its rhizomes (Subhuti,

    2003).

  • Ref. code: 25595729320217IWP

    23

    2.4.1.2 Phytochemistry

    The major essential oil constituents and sesquiterpenoid glycosides of

    A. lancea (Thunb.) DC. rhizomes (Figure 9) which exhibit several pharmacological

    functions include β-eudesmol, hinesol, atractylone, and atractylodin. β-eudesmol

    accounts for about 6% of the total yield, while atractylodin and atractylone account

    for about 14 and 9% of the total yield, respectively, following activity guided

    fractionation (Ji et al., 2001; Ouyang et al., 2012; Zhou et al., 2012). The essential

    oils of A. lancea (Thunb.) DC. rhizomes possess anti-inflammatory and anti-ulcer

    properties, scavenging activity on trichloromethyl (CCl3) radical, inhibitory activity

    on lipid peroxidation and xanthine oxides, inhibitory activity on tert-butyl

    hydroperoxide-induced cytotoxicity and lipid peroxidation in primary culture of rat

    hepatocytes, and inhibitory activity on the growth of esophageal carcinoma cells and

    tumor cells (Yang et al., 2002).

    2.4.1.3 Safety and toxicity

    The maximum tolerated dose of A. lancea (Thunb.) DC. rhizomes

    extract was 5,000 mg/kg body weight in three species (hamsters and mice) and no

    significant toxicity except stomach irritation and general CNS depressant signs

    (reduced alertness and locomotion and diminished response to touch and balance)

    were observed following acute and subacute toxicity studies (Plengsuriyakarn et al.,

    2012; Plengsuriyakarn et al., 2015).

  • Ref. code: 25595729320217IWP

    24

    β-Eudesmol Atractylodin

    Atractylone Hinesol

    Figure 9 The chemical structures of major components of A. lancea (Thunb.) DC.

    rhizome extract (Ji et al., 2001).

  • Ref. code: 25595729320217IWP

    25

    2.4.2 Kaempferia galanga Linn.

    2.4.2.1 Plant description and medicinal properties

    K. galanga Linn. is one of the plants in Zingiberaceae family and a

    genus of rhizomatous herbs distributed in the tropics and sub tropics of Asia and

    Africa, being cultivated for its aromatic rhizomes (Geetha et al., 1997). Since the

    rhizomes of this plant contain volatile oils and other important compounds of

    enormous medicinal values, they are very demanding to the traditional health care

    practitioner and is used as traditional medicinal plant in many countries (Mohammad,

    2008). It is well known for its action as remedy for inflammation and various ethno

    botanical studies indicated that its rhizome extract is used to relieve pain and various

    disorders around Southeast Asia (Kanjanapothi et al., 2004; Mohammad, 2008).

    (1) Anti-inflammatory and analgesic activity

    Aqueous extracts of K. galanga Linn. (leaves and rhizomes)

    administered at different doses (30, 100, and 300 mg/kg body weight) to rats

    produced significant anti-inflammatory and analgesic effects in hot plate and formalin

    tests (Sulaiman et al., 2008). An oral dose of 200 mg/kg body weight of KG rhizome

    extract produced anti-nociceptive effect more potent than aspirin at the dose of 100

    mg/kg body weight, but lesser than morphine at the dose of 5mg/kg body weight, in

    another study (Ridtitid et al., 2008). The capacity of the extracts to block abdominal

    constriction demonstrated in the hot plate and formalin tests (Sulaiman et al., 2008)

    indicates that analgesic activity of A. lancea (Thunb.) DC. rhizomes involves both

    central (opioid receptors), and peripheral (cyclooxygenase, COX) mechanisms.

    A study conducted using n-hexane rhizome extract of K. galanga

    Linn. in acetic induced writhing test in mice demonstrated the highest analgesic effect

    (62.31% inhibition) and the chloroform extract showed moderate analgesic activity

    (38.19% inhibition) as compared with the activity observed by the positive control

    diclofenac sodium (63.82% inhibition) (Chowdhur et al., 2014).

    (2) Antimicribioal activity

    K. galanga Linn. extract has also been demonstrated to exhibit

    antimicrobial activity against a number of organisms including Staphylococcus

  • Ref. code: 25595729320217IWP

    26

    aureus, Streptococcus pyogenes, Candida albicans, Escheriachia coli, Klebsiella

    pneumonia, Salmonella typhi, Seratia marcescens, Vibrios cholera, Vibrio

    sparahaemolyticus, Enterococcus faecalis, and Pseudomonas aeruginosa with

    minimum inhibitory concentration (MIC) of 0.81, 3.25, 25, >6.5, > 6.5, >6.5, >6.5,

    >6.5, >6.5,1.625 and >6.5 μg/ml, respectively (Mekseepralard et al., 2010). Moderate

    antibacterial activity (including gram positive and gram negative bacteria) has also

    been demonstrated by another study (Dash et al., 2014) with different K. galanga

    Linn. extracts as compared with the standard drug ciprofloxacin (5 μg/disc).

    Significant antibacterial and antifungal properties were also

    demonstrated by the rhizome extract of K. galanga Linn. by using different solvents

    (ethanol, methanol, and aqueous). The ethanolic extract showed potent activity

    against Staphylococcus aureus, Aspergillus niger, A. flavus, A. fumigatus and

    Candida albicans with mean + SD inhibition zone of 21.3±0.08, 16.4±0.45,

    15.3±0.36, 14.0±0.48, 12.2±0.45 mm, respectively (Kochuthressia et al., 2012). On

    the other hand, the methanolic rhizome extract showed potent activity against

    Aspergillus niger (inhibition zone of 14.2±0.26 mm).

    (3)Anti-oxidant activity

    Anti-oxidant activity of K. galanga Linn. extract was also observed

    in various studies (Chan et al., 2008; Mekseepralard et al., 2010). The total phenolic

    content (TPC) of ethanolic extract was found to be 146 mg galic acid equivalent

    (GAE)/100 g from the leaves of K. galanga Linn. and 57 mg GAE/100g from the

    rhizomes of K.galanga Linn. (Chan et al., 2008). The antioxidant activities of the

    leave and rhizome extracts were 77 mg ascorbic acid/100 g and 17 mg ascorbic

    acid/100 g, respectively, and this anti-oxidant activity was further reduced by drying

    which was in turn prevented if the plant was subjected to freeze-drying (Chan et al.,

    2008). However, it was reported in another study that the volatile oil extract from K.

    galanga Linn. was inactive for antioxidant activity with IC50 >100 µg/ml (Tewtrakul

    et al., 2005).

    (4) Anti-hyperglycemic effects

    Different solvent extracts of K. galanga Linn. were shown to exhibit

    antihyperglycemic activity in in vivo models. The crude methanolic extract and

  • Ref. code: 25595729320217IWP

    27

    chloroform soluble fractions of K. galanga Linn. extract (200 mg/kg body weight)

    showed remarkable blood glucose level reduction (with 61.2 and 89.63 % reduction,

    respectively), which were more prominent than that of standard drug glibenclamide

    (34.78% reduction compared to control) after 30 minutes of glucose loading

    (Chowdhur et al., 2014). The percentage of maximal inhibition of glucose load for

    that of the n-hexane extract was 91.63%, which was also higher than the value

    observed for that of the standard drug glibenclamide (Chowdhur et al., 2014).

    (5) Antineoplastic and apoptotic activity

    K. galanga Linn. extract has also been reported to possess

    antineoplastic activity with inhibitory effect being observed on the tumor-promoting

    stage of neoplasia (Koh, 2009; Vimala et al., 1999). A study conducted with the

    methanolic extract of K.galanga Linn. showed inhibitory activity in 12-O

    tetradecanoyl-phorbol-13-acetate (TPA)-induced activation of Epstein barr virus early

    antigen in Raji cells and this led to the inhibitory activity on tumor promoting stage

    even though the inhibition of this tumor promoting stage was partial (Vimala et al.,

    1999). At the dose of 320 μg/ml, the inhibition observed was 80% and the inhibition

    was even increased up to a maximum of 90% at 640 μg/ml in this study. In a recent

    study (Mahavorasirikul et al., 2010), K. galanga Linn. ethanolic extract showed

    cytotoxic activity against CL-6 and HepG2 cell lines with mean (+SD) IC50 values of

    37.36+3.98 and 115.47+26.23 µg/ml, respectively. Moreover, the methanolic extracts

    of K. galanga Linn. has been shown to exert inhibitory effect on human cardiac

    fibroblast (HCF-7) and human T-cell leukemia (HT-29) cell lines but only at doses

    higher than 250 μg/ml (Kirana et al., 2003).

  • Ref. code: 25595729320217IWP

    28

    (A) (B)

    (C) (D)

    Figure 10 (A) Flower, (B) fresh rhizomes, (C) dried rhizomes, and (D) sprouting

    plant from rhizomes of K. galanga Linn. (Katzer, 2011).

  • Ref. code: 25595729320217IWP

    29

    2.4.2.2 Phytochemistry

    Preliminary phytochemical group tests revealed that different K.

    kalanga Linn. extracts contain carbohydrates, tannins, flavonoids, proteins, steroids,

    alkaloids, and resins (Dash et al., 2014) as summarized in Table 2.

    The major constituents of the volatile oil extracts of K.galanga

    rhizome include cineol, borneol, 3-carene, camphene, kaempferol, kaempferide,

    cinnamaldehyde, p-methoxycinnaamic acid, ethyl cinnamate, and ethyl p-

    methoxycinnamate (Figure 11) (Sutthanont et al., 2010). Ethyl–para-

    methoxycinnamate (EPMC) and ethyl-cinnamate (EC) are found to be the most

    dominant and vital constituents in the methanol and water extracts (Huang et al.,

    2008; Sirisangtragul and Sripanidkulchai, 2011; Tewtrakul et al., 2005). Among the

    volatile oil content obtained from the rhizomes of K. galanga Linn., the highest

    content (peak chromatogram area 31.77%) was EPMC (Tewtrakul et al., 2005)

    (Table 3). A much higher EPMC content (80 mg %) was reported from the

    dichloromethane extract of K. galanga Linn. rhizomes (Sirisangtragul and

    Sripanidkulchai, 2011). About 98.98% of essential oil constituents from K. galanga

    Linn. have been isolated and identified; the remaining 1.11% constituents are still

    unknown (Sutthanont et al., 2010).

    It has been reported that EPMC of K. galanga Linn. is involved in

    many biological activities including anticancer (Liu et al., 2010; Zheng et al., 1993),

    antimicrobial (Kanjanapothi et al., 2004), and anti-monoamine oxidase activities

    (Noro et al., 1983). This chemical constituent has been also found to inhibit

    proliferation of HepG2 cell line in a dose-dependent manner (Liu et al., 2010).

    Mechanistic-based studies using annexin-fluorescein isothiocyanate and propidium

    iodide staining showed an increased early apoptotic population in human

    hepatocellular carcinoma cells. It was proposed that EPMC induced translocation of

    phosphatidyl serine of HepG2 cells to cell surface, resulting in an increase in sub-G

    cell population (Liu et al., 2010). A significant anti-inflammatory potential has also

    been demonstrated by suppressing interleukin-1, tumor necrosis factor-α (TNF-α),

    and angiogenesis by blocking endothelial functions (Umar et al., 2014). EC, EPMC

    and p-methoxycinnamic acid from the methanolic extract of K. galanga Linn. also

  • Ref. code: 25595729320217IWP

    30

    showed larvicidal activity against the second stage larva of dog roundworm, Toxocara

    canis (Kiuchi et al., 1988). The pharmacological activities of various chemical

    constituents identified from K. galanga Linn. including their possible mechanisms of

    action are summarized in Table 4.

    2.4.2.3 Safety and toxicity

    Administration of K. galanga Linn. ethanol extract to rats up to the

    dose level of 5,000 mg/kg body weight (acute toxicity test) neither resulted in

    mortality nor any significant difference in body and organ weights between control

    and test animals (Kanjanapothi et al., 2004). However, in the subacute toxicity testing

    at the dose levels of 25, 50, and 100 mg/kg body weight for 28 days, a slight decrease

    in the lymphocyte count was observed in male rats.

    In a study conducted with the dichloromethane extract of K. galanga

    Linn. (100 mg/kg body weight) and EPMC (120 and 160 mg/kg body weight) in mice

    for 28 consecutive days of subacute toxicity testing, a significant effect on death,

    body or organ weights of mice were not observed in both treated and control groups

    (Sirisangtragul and Sripanidkulchai, 2011). Oral administration with the K. galanga

    Linn. extract and EPMC for evaluation of its effect on hepatic microsomal

    cytochrome P450 (CYP450) enzyme activities demonstrated a reduction in

    microsomal P450 content, whereas CYP1A1 and CYP2B activities were increased but

    no effect was observed on CYP1A2 and CYP3A4 activity (Sirisangtragul and

    Sripanidkulchai, 2011). It has been indicated that more than 50% of conventional

    drugs are metabolized via these CYP isoforms (CYP1A1 and CYP2B) (Anzenbacher

    and Anzenbacherova, 2001) and the results observed by such study (Sirisangtragul

    and Sripanidkulchai, 2011) might suggest certain safety of this plant with regard to

    herbal-drug interaction and human risk assessment. Moreover, the slight reduction of

    CYP2E1 activity was observed (Sirisangtragul and Sripanidkulchai, 2011) which

    could suggest slight anti-carcinogenic activity since CYP2E1 is involved in metabolic

    activation of many low molecular weight toxins and carcinogens (such as carbon

    tetrachloride, chloroform, methylchloride, and vinylchloride) (Anzenbacher and

    Anzenbacherova, 2001).

  • Ref. code: 25595729320217IWP

    31

    Table 2 Phytochemical test results detected in different solvent extracts of K. galanga

    Linn. rhizomes (Dash et al., 2014).

  • Ref. code: 25595729320217IWP

    32

    Ethyl-cinnamate (EC) Ethyl-p-methoxycinnamate (EPMC)

    Kaempferol Kaempferide

    Figure 11 Chemical structures of some important phytoconstituents isolated from K.

    galanga Linn. extract (Sutthanont et al., 2010).

  • Ref. code: 25595729320217IWP

    33

    Table 3 Volatile oil components, retention time and peak area (%) of K. galanga

    Linn. oil (Tewtrakul et al., 2005).

  • Ref. code: 25595729320217IWP

    34

    Table 4 Summary of possible mechanisms of action associated with the bioactive

    constituents of K. galanga Linn. for the specific pharmacological activities.

    Pharmacological

    activity

    Responsible active

    Constituent

    Possible mechanism of

    action

    Mosquito

    repellant,

    larvicidal and

    antimalarial

    activity

    Ethyl p-methoxycinnamate,

    ethyl-cinnamate, 3 carene, 2-

    propionic acid (Kim et al.,

    2008; Sutthanont et al., 2010)

    Destruction of ionic

    regulation in the anal gills

    (Insun et al., 1999)

    Analgesic and

    anti-inflammatory

    …….. Central mechanism involving

    opioid receptors and

    peripheral mechanism

    involving COX

    pathway(Ridtitid et al., 2008;

    Sulaiman et al., 2008)

    Antimicrobial

    activity

    Ethyl-p-methoxycinnamate

    (Kanjanapothi et al., 2004)

    ……..

    Anti-oxidant

    activity

    Total phenolic content and

    flavovoids including luteolin

    and apigenin (Mustafa et al.,

    2010)

    ……..

    Antihyperglycemic

    activity

    …… May probably possess an insulin-like effect or stimulate

    the pancreatic ß-

    cells(Chowdhur et al., 2014)

    Antineoplastic

    activity

    Ethyl-p-methoxycinnamate (Liu

    et al., 2010)

    Translocation of

    phosphatidylserine of Hep G2

    cells to cell surface, resulting

    in an increase in sub-G cell

    population (Liu et al., 2010)

  • Ref. code: 25595729320217IWP

    35

    CHAPTER 3

    OBJECTIVES

    3.1. General objective

    The overall objective of the research dissertation was to evaluate the

    safety/toxicity of the two candidate plants for further development as alternative

    chemotherapeutics for cholangiocarcinoma (CCA). These included the CMC

    (chemistry, manufacturing and control) oral pharmaceutical formulation of the crude

    ethanolic extract of A. lancea (Thunb.) DC. (AL) rhizomes prepared for first-in-human

    clinical study. The safety profile and anti-CCA activity of the crude ethanolic extract

    of this plant has previously been shown in a series of in vitro and in vivo model

    (Mahavorasirikul et al., 2010; Plengsuriyakarn et al., 2012). In addition, anti-CCA

    activity of the crude ethanolic extract of K. galanga Linn. (KG) leaves was initially

    evaluated in vitro models (Mahavorasirikul et al., 2010).

    3.2 Specific objectives

    3.2.1 To evaluate potential of toxicity of the CMC oral pharmaceutical

    formulation of the crude ethanolic extract of AL rhizomes:

    3.2.1.1 To evaluate the acute toxicity of the CMC oral pharmaceutical

    formulation of AL in rats;

    3.2.1.2 To evaluate the subchronic (90 days repeated dosing) toxicity

    of the CMC oral pharmaceutical formulation of AL in rats;

    3.2.2 To evaluate potential cytotoxic activity against CCA, toxicity, and

    anti-CCA activity of the crude ethanolic extract of KG rhizomes:

    3.2.2.1 To standardize the extraction procedure for preparation of the

    crude ethanolic extract of KG rhizomes;

  • Ref. code: 25595729320217IWP

    36

    3.2.2.2 To evaluate cytotoxic activity of the KG rhizome extract in

    CCA cell line (CL-6);

    3.2.2.3 To evaluate potential of toxicity (acute and subacute toxicity)

    of the crude ethanolic extract of KG rhizomes in mice; and

    3.2.2.4 To evaluate the anti-CCA activity of the KG rhizome extract in

    CCA-xenografted BALB/c nude mouse model.

  • Ref. code: 25595729320217IWP

    37

    CHAPTER 4

    RESEARCH METHODOLOGY

    4.1. Reagents and cell lines

    The solvent used for plant extraction (95% ethanol) was commercial

    grade which was obtained from Labscan Asia Co. Ltd. (Bangkok, Thailand). HPLC

    grade methanol and distilled water were obtained from Fisher Scientific

    (Leicestershire, UK). The cell culture reagents including RPMI 1640, DMEM, fetal

    bovine serum (FBS), phosphate buffered saline (PBS), penicillin and streptomycin

    antibiotics were purchased from Gibco Life Technologies (NY, USA). The CCA cell

    line (CL-6) used for both in vitro and in vivo study was kindly provided by Associate

    Professor Adisak Wongkajornsilp, Department of Pharmacology, Faculty of

    Medicine, Siriraj Hospital, Bangkok, Thailand. The normal fibroblast cell line used

    (OUMS) was obtained from JCRB cell bank (Osaka, Japan). Methyl thiazoldiphenyl

    tetrazolium (MTT) reagent was obtained from Life Technologies (CA, USA) and

    dimethyl sulfoxide (DMSO) was obtained from MP Biomedicals (CA, USA).

    Atractylodin (standard compound) and 5-FU were purchased from Wako Pure

    Chemical Industries Ltd. (Osaka, Japan) and tween-80 was purchased from Sigma-

    Aldrich (MO, USA). Neutral buffered formalin (NBF) used for organs/tissue fixation

    was purchased from Bio-Optica (Milano, Italy). Isoflurane used for euthanasia was

    purchased from Minrad Inc. (PA, USA). All other remaining chemicals and reagents

    were purchased from commercial suppliers and were of high purity grade. The

    standard ethyl-p-methoxycinnamate (EPMC) was kindly supplied by Dr. Sumet

    Kongkiatpaibo, Drug Discovery and Development Center, Thammasat University,

    Thailand.

  • Ref. code: 25595729320217IWP

    38

    4.2 Preparation of test materials

    4.2.1 Chemical, manufacturing and control (CMC) oral

    pharmaceutical formulation of Atractylodes lancea (Thunb.) DC.

    The CMC oral pharmaceutical formulation of the crude ethanolic extract

    of AL rhizomes was prepared by Ms. Thananchanok Rattanathada, the Center of

    Excellence in Pharmacology and Toxicology and Molecular Biology of Malaria and

    Cholangiocarcinoma. The AL powder formulation was prepared as AL crude extract

    (from AL rhizomes) mixed with lactose (25% w/w) and the methods used for

    extraction and preparation of the AL formulation included maceration, rotary

    evaporation, and freeze-drying.

    4.2.2 Kaempferia galanga Linn.

    The dried and powdered rhizomes of the plant K. galanga Linn. (KG) were

    obtained from Thailand, Nakhon Pathom Province. Authentication of the plant

    material/rhizome was carried out at the herbarium of the Department of Forestry,

    Bangkok, Thailand, where the herbarium voucher has been kept (Mahavorasirikul et

    al., 2010).

    The plant rhizome powder was extracted by sonication, heating under

    reflux, and maceration (1, 3, and 7 days) using 95 % ethanol to obtain the optimized

    method for preparation of the extraction with high and consistent yield. For the

    sonication method, a sample powder of about 10 g was mixed with 95% ethanol (300

    ml) and sonicated with the ultrasonicator (Meditop, Bangkok, Thailand) for 75

    minutes. The plant rhizomes of about 10 g in 300 ml of 95% ethanol was also used for

    the heating under reflux method in a water bath (MemmertTM, Schwabach, Germany)

    for about 50 minutes according to the previously described methods with modification

    (Khoddami et al., 2013; Sasidharan et al., 2011).

    For the maceration method, the powdered KG rhizomes (10 g) was mixed

    with 300 ml of 95% ethanol in a small glass flask and stored at 25 oC for 1, 3, and 7

    days. Later on, maceration method for 7 days was optimized and about 1,000 g

  • Ref. code: 25595729320217IWP

    39

    powdered rhizomes was added in each three large clean flasks containing 5 liters of

    95% ethanol for 7 days at 25-30 °C with occasional shaking and stirring. The extracts

    were separated first (filtered) with gauze and then through Whatman no. 1 filter paper

    (GE HealthcareTM, Maidstone, England). The residues were remacerated for

    additional 24 h and then filtered as indicated above. The extract solvent from was

    evaporated using a rotary evaporator (HeidolphTM, Schwabach, Germany) and further

    concentrated by heating at 60 oC in a water bath (Builders et al., 2011). The crude

    extracts obtained using different methods were weighed and the median (range)

    percent yields were calculated. The extracts were stored in small glass bottles

    (covered with aluminum foil) at -20 °C until used (Figure 12) to ensure the apparent

    stability and activity (Builders et al., 2011; Sasidharan et al., 2011).

  • Ref. code: 25595729320217IWP

    40

    K. galanga Linn. (KG) rhizomes

    Maceration in 95% of ethanol for 7 days, followed by remaceration for 24 hours

    Filtration through gauze and Whatman no. 1 filter paper

    Concentration with rotary evaporator (at 60oC) and then in a water bath

    Storage in small glass bottles (covered with aluminum foil) at -20 oC until use

    Figure 12 A diagram showing extract preparation using 7 day maceration of the KG rhizomes.

  • Ref. code: 25595729320217IWP

    41

    4.3 Identification of marker compounds

    4.3.1 CMC oral pharmaceutical formulation of Atractylodes lancea

    (Thunb.) DC.

    Atracylodin was used as a marker compound for quality control of the

    preparation process for preparation of the AL extract and CMC oral pharmaceutical

    formulation (Ji et al., 2001; Ouyang et al., 2012). The stock solution was prepared by

    accurately weighing and dissolving the compound in methanol to obtain the final

    concentration of 1,000 µg/ml. Working solutions at desired concentrations were

    prepared by diluting the stock standard solution with methanol. The AL rhizome

    extract CMC oral pharmaceutical formulation sample solution was also prepared by

    accurately weighing 100 mg of each sample and dissolved in methanol (5 ml). To

    enable complete dissolution, each sample was sonicated for 60 min (triplicate each).

    Prior to injection onto the HPLC system, each solution was filtered through a 0.22 µm

    nylon membrane filter. Semi-quantitative determination of atractylodin content of the

    extract CMC oral formulation was performed by using high performance liquid

    chromatography (HPLC) according to the previously described procedures

    (Kongkiatpaiboon et al., 2015). The HPLC system consisted of an Agilent 1260 series

    equipped with a 1260 quaternary pump, 1260 ALS autosampler, 1260 TCC column

    thermostat, and 1260 DAD detector (Agilent Technologies, CA, USA). Sample

    separation was achieved on a Hypersil BDS C18 column (4.6 x 100 mm i.d., 3.5 µm)

    with a C18 guard column (Thermoschientific, MA, USA) and the mobile phase

    consisting of (A) water and (B) methanol using gradient elution: 50% B in A to 100%

    B for 15 min; 100% B for 10 min according to the previously described procedures

    with modification (Kongkiatpaiboon et al., 2015. The column was equilibrated with

    50% B in A for 10 min prior to each analysis and the flow rate was set at 1.0 ml/min

    with controlled temperature at 25C. Sample injection volume was 5 µl and UV

    detection was set at the wavelength of 360 nm for the analysis.

  • Ref. code: 25595729320217IWP

    42

    4.3.2 Standardization of the crude ethanolic extract of Kaempferia

    galanga Linn. rhizomes

    Ethyl-p-methoxycinnamate (EPMC) which is the major bioactive

    chemical constituent of the KG extract (Sirisangtragul and Sripanidkulchai, 2011;

    Tewtrakul et al., 2005) was used as a marker compound for quality control of the

    preparation process of the KG rhizome extract. Semi-quantitative determination of

    EPMC content of the extract was performed using HPLC according to the previously

    described method with modification (Sirisangtragul and Sripanidkulchai, 2011). The

    extract and the standard EMPC were prepared at the concentrations of 20 and 10

    µg/ml, respectively. The HPLC system used consisted of HPLC instrument 1200

    series (Agilent Technologies, CA, USA), and photo diode array detector (PDA)

    detector (Thermoschientific, MA, USA); and sample separation was performed using

    the ThermoscientificTM Hypersil Gold Column (250 x 4.6mm i.d., 5 µm particle size,

    reversed phase C18) with an isocratic mobile phase consisting of a mixture of

    methanol and distilled water (54:46% v/v) at a flow rate of 1 ml/min. Sample

    injection volume was 10 μl , the column temperature was maintained at 250C and UV-

    detection was set at 270 nm for this analysis.

    4.4 Animal stocks, handling and ethical approval

    The toxicity (acute and 90-days subchronic toxicity) testing of the CMC

    oral pharmaceutical formulation of the crude ethanolic extract of AL rhizomes was

    evaluated in Wistar albino (strain Crl:W1 or Han) rats of both genders (6-9 weeks of

    age, and weighing 160-400g). The toxicity testing (acute and subacute toxicity) of the

    crude ethanolic extract of KG rhizomes was evaluated in ICR (Imprinting Control

    Region) male and female mice, 6-7 weeks of age, weighing 26-40 g. The anti-CCA

    activity of the KG rhizome extract was evaluated in BALB/c nude mice of both

    genders (7-8 weeks of age, and weighing 15-24 g). All the animal stocks were

    obtained from the National Laboratory Animal Center, Thailand. Animals were

    housed under standard conditions and acclimatized for abou