Pharmacognostic Investigation of Black Cohosh ( racemosa (L.) Nutt.)

BY

DANIEL S. FABRICANT B.S., University of North Carolina-Chapel Hill, 1997

DISSERTATION

Submitted in partial fulfillment of the requirements for the degree of Doctor of Philosophy in Pharmacognosy in the Graduate College of the University of Illinois at Chicago, 2005 Chicago, Illinois

DEDICATION

This dissertation is dedicated to my family, Wendy, Michael, Rachel, and

Loretta. Thank you for your support, without which this work would not have been possible. ii

ACKNOWLEDGEMENTS

Much gratitude is due to Dr. Guido Pauli who shared lab space,

equipment, philosophy and his enthusiasm. Special thanks to all of those brave

souls who volunteered to serve on my committee: Drs. Norman R. Farnsworth,

major and dissertation advisor, Guido F. Pauli, Judy L. Bolton, Richard B. van

Breemen, Jim Wang and Dean Rosalie Sagraves.

I am indebted to a number of people who have helped make this work

possible. Thanks to Joanna Burdette for her skilled and exhaustive bioassay

work, Cassia Overk, Drs. Vivian Zheng, Jianghua Liu, Rachel Ruhlen and the

project leader, Dr. J.L. Bolton for their help during the bioassay phases of this

research. To Dr. van Breemen and the Botanical Center’s Core C, especially Dr.

Dejan Nikolic, thank you for the wonderful mass spectral work provided herein.

I am very grateful to Drs. Shao-Nong Chen and Linda Lu for all their

exceptional assistance and expansive knowledge with many phytochemical

aspects of this research. Much gratitude is due to Dr. Guido Pauli, the Project

Leader, who shared lab space, equipment, philosophy and his enthusiasm.

I am much obliged to Drs. Dejan Nikolic, David Lankin, Steve Totura,

James Graham, Colleen Piersen, Aleksej Krunic, Eduardo Callegari, Paul W.

Buehler, Chad Haney, Hongjie Zhang, Jose Fausto-Rivero, Amanda Koch and

Wenkui Li for cheerful collaboration along the way.

Thanks to the Department of Medicinal Chemistry and Pharmacognosy,

the Program for Collaborative Research in the Pharmaceutical Sciences, the

ODS/NCCAM/NIH center for Botanical Dietary Supplement Research in iii

Womens’ Health (grant # P50 AT00155) and the office of the Dean, College of

Pharmacy, UIC, for teaching and research assistantships during the course of my studies. Thanks to Dr. Qun Yi Zheng of Pure World Botanicals and to our collaborators at Pharmavite.

Much gratitude is due to NAPRALERT for funding and data that made this work possible, especially Mary Lou Quinn, and Norman R. Farnsworth. Thanks to

Aubrey Neas, Dr. Gwynn Ramsey, Bambi Teague and Keith Langdon at the

National Park Service with their assistance in acquiring material. Thanks to

Drs. Andreas Constantinou, Andrew Mesecar, Steven Swanson and Bernard

Santarsiero for kindly sharing lab space and equipment during the PCR-phase of this research. Thanks to Donna Webster for her hard work with the RAPD-PCR work.

I am truly grateful to my mother, Loretta Mae Fabricant, for her support. I would like to individually thank my brother and sister, Michael and Rachel

Fabricant, for their continuous support, friendship and humor. To Wendy, thanks for keeping me on task, you make me want to be a better person.

Special thanks to the Boss, Professor Norman R. Farnsworth and his better half Priscilla, for their input, patience, kindness and guidance.

iv

PREFACE

This dissertation is concerned with the standardization of a botanical dietary supplement for use in a clinical trial. It is the intent of the author, for this document to be used as a resource for future work with Cimicifuga racemosa extracts and its constituents. As well as to provide a sound review of the chemical, biological and botanical aspects of the literature of C. racemosa. v

TABLE OF CONTENTS

ACKNOWLEDGEMENTS ii PREFACE iv LIST OF SYMBOLS AND ABBREVIATIONS x LIST OF TABLES xiv LIST OF FIGURES xv SUMMARY xviii

1.0 INTRODUCTION AND SCOPE OF STUDY 2

2.0 STATEMENT OF PROBLEM 6

3.0 LITERATURE REVIEW 7

3.1 OF BLACK COHOSH 7

3.1.1 Family 7 3.1.2 Cimicifuga L. 8 3.1.3 Cimicifuga (L.) Nutt. 9

3.2 BLACK COHOSH IN TRADITIONAL MEDICINE 10

3.3 EFFECTS ON CLIMACTERIC SYMPTOMS 11 RELATED TO MENOPAUSE

3.3.1 Conclusion of Clinical Study of Black Cohosh 18 on Climacteric Symptoms Related to Menopause

3.4 PHARMACOLOGY AND BIOCHEMISTRY OF 20 CIMICIFUGA RACEMOSA EXTRACTS

3.4.1 Competitive Estrogen Receptor Binding 21 3.4.2 Receptor Expression 22 3.4.3 Plasma Hormone Levels 23 3.4.4 Hormonal Secretion 24 3.4.5 Osteopenia Inhibition 24 3.4.6 Uterine Weight/Estrous Induction 25 3.4.7 MCF-7 Cell Proliferation Inhibition 25 vi

3.4.8 CNS Effects and Neurotransmitter Binding 26 3.4.9 Miscellaneous 27 3.4.10 Conclusion on Extract Activity 28

3.5 PHYTOCHEMISTRY OF BLACK COHOSH 35

3.5.1 Triterpene Glycosides 36

3.5.2 Phenolic Acid Derivatives 36

3.5.3 Flavonoids 37

3.5.4 Miscellaneous 37

4.0 EXPERIMENTAL 39

4.1 PLANT MATERIAL 39

4.1.1 Procurement of Plant Material 39

4.1.2 RAPD-PCR Plant Identification 39

4.1.3 Microscopic Analysis 41

4.1.3.1 Light Microscopy 41

4.1.3.2 Scanning Electron 41 Microscopic (SEM) Analysis

4.2 CHROMATOGRAPHY 42

4.2.1 Solvent Partition and Fractionation of Black Cohosh 42

4.2.2 Column Chromatography 42

4.2.3 Thin-Layer Chromatography (TLC) 43

4.2.4 High-Performance Liquid Chromatography (HPLC) 44

4.2.4.1 Analytical HPLC 46

4.2.4.2 Semi-preparative HPLC 50 vii

4.3 SPECTROSCOPIC METHODS 52

4.3.1 Nuclear Magnetic Resonance (NMR) Spectroscopy 52

4.3.2 Mass Spectrometry (MS) 52

4.3.3 Other Techniques 53

4.4 BIOLOGICAL ACTIVITY 53

4.4.1 Estrogenic Activity 53

4.4.2 Serotonin (5-HT) Binding Activity 54

4.4.3 Other Biological Activities 54

5.0 RESULTS 55

5.1 PLANT PROCUREMENT 55

5.2 RAPD-PCR ANALYSIS 58

5.3 MICROSCOPY 63

5.3.1 Light Microscopy 63

5.3.2 Scanning Electron Microscopy (SEM) 64

5.4 CHROMATOGRAPHY 66

5.4.1 Solvent Extraction and Partitioning 66 of non-Polar Fractions

5.4.2 Solvent Extraction and Partitioning 67 of Polar Fractions

5.4.3 Amberlite XAD-2 69

5.4.4 MCI gel CHP20P 72

5.4.5 Sephadex LH-20 81

5.4.6 Silica Gel Separation 83

5.5 ISOLATION OF CONSTITUENTS 85 viii

5.5.1 Isolation of 1 (cimipronidine) 85

5.5.2 Isolation of 2 (fukinolic acid) 88

5.5.3 Isolation of 3 (actein(R/S)) 88

5.5.4 Isolation of 4 (isoferulic acid) 90

5.6 CHARACTERIZATION OF CONSTITUENTS 91

5.6.1 Characterization of 1 91

5.6.2 Characterization of 2 104

5.6.3 Characterization of 3 108

5.6.4 Characterization of 4 113

5.6.5 Additional Characterization Studies 117

5.7 Preparation of the Clinical Extracts 120

5.7.1 Collaborator Formulation of Phase I 130 Clinical Extract

5.7.2 Collaborator Formulation of Phase II 130 Clinical Extract

5.7.3 Phase I clinical capsule analysis 130

5.7.4 Phase II clinical capsule analysis 132

5.7.5 Overview of Clinical Material 132

6.0 DISCUSSION

6.1 Clinical Extract Formulation and Biological Activity 136

6.2 Biological Activity of Black Cohosh 139

6.3 Chemical and Botanical Nomenclature 140

6.4 Preparative and Analytical Techniques 142

6.5 NMR spectroscopy 149 ix

7.0 CONCLUSIONS 152

7.0.1 Review the Pharmacognosy of Black Cohosh 152

7.0.2 Preparation of a biologically, chemically and 152 botanically standardized extract for clinical trial.

7.0.3 Chemical characterization of the active butanolic 152 Black Cohosh fraction

7.0.4 Development of preparative and analytical methods 153

7.0.5 Dereplication of Botanical Center isolates 154

7.1 Summary of Conclusions 154

8.0 FUTURE DIRECTIONS 155

9.0 REFERENCES 158

10.0 APPENDICES 174

10.1 Additional Structural Data for Novel Center Isolates 174

10.2 Mass spectral data supporting the presence 184 of additional nitrogenous constituents in Black Cohosh

10.3 Additional Black Cohosh documentation 188

11.0 VITA 205

x

LIST OF ABBREVIATIONS

ACN acetonitrile (HPLC grade)

Ac acetyl

AP alkaline phosphatase

[α]D specific optical rotation

APT attached proton test

C5D5N deuterated pyridine

CGI Clinician’s Global Impression Scale

CHCl3 chloroform

CHP20P mci-gel CHP20P resin

13C NMR carbon-13 nuclear magnetic resonance

COSY COrrelation SpectroscopY cm-1 wave number

δ chemical shift

DEPT distortionless enhanced proton transfer

DSHEA Dietary Supplement Health and Education Act

d.d.i. distilled, deionized

(d) doublet

eV electron volt

ε molar absorptivity

EIMS electron impact mass spectrometry

ELSD evaporative light scattering detector

equiv. equivalent

xi

LIST OF ABBREVIATIONS(continued)

ER estrogen receptor

EtOH ethanol

EtOAc ethyl acetate

GPS global positioning system

HAM-A Hamilton Anxiety Scale

HMBC heteronuclear multiple-bond connectivity spectroscopy

HMQC heteronuclear multiple-bond quantum coherence spectroscopy

1H NMR proton nuclear magnetic resonance

HPLC high-performance liquid chromatography

5-HT serotonin (5-hydroxytryptamine)

Hz hertz

IPR isopropanol (2-propanol)

IR infrared absorption

IC50 inhibitory concentration (50%)

J coupling constant

KMI Kupperman Menopausal Index

λmax maximum wavelength

M molar concentration

MHz megahertz

MeOH methanol min minutes xii

LIST OF ABBREVIATIONS (continued)

mp melting point

m/z mass-to-charge ratio

MSS unspecified menopausal index using the Likert scale

νmax maximum frequency n-BuOH 1-butanol

NMR nuclear magnetic resonance

Open Open-Labeled study ovx Ovariectomized ppm parts per million

PDA photodiode array pet. ether petroleum ether

POMS Profile of Mood States Scale

(q) quartet

SDS Self-Assessment Depression scale

Si silica

(s) singlet

(t) triplet

TLC thin-layer chromatography tR retention time

UV ultraviolet absorption

VMI Vaginal Maturity Index

xiii

VAS Visual Analog Scale v volume w weight

XAD-2 amberlite XAD-2 ion exchange resin xiv

LIST OF TABLES

Table 1. Black Cohosh clinical studies 12

Table 2. Commercially available products that have been evaluated 15 in clinically studies

Table 3. Compounds isolated from C.racemosa roots 30

Table 4. Standard TLC solvent systems 43

Table 5. Analytical HPLC-ELSD conditions and method parameters 47

Table 6. Botanical Center database of Black Cohosh collections 56

Table 7. 1H and 13C NMR data of 1 103

Table 8. 1H and 13C NMR data of 2 107

Table 9. 1H and 13C NMR data of 3 112

Table 10. 1H and 13C NMR data of 4 116

Table 11. Biological activity of a MeOH Black Cohosh Extract. 121

Table 12. Compound activity data of triterpenes. 122

Table 13. Average constituent levels of 14 C. racemosa Collections 123

Table 14. Geographic averages of constituent levels of 124 14 C. racemosa Collections

Table 15. Percentage of Active standards in extracts 126

Table 16.Serotonin binding activity of crude extracts 128

xv

LIST OF FIGURES

Figure 1. Flowering raceme of C. racemosa. 1

Figure 2. Triterpenes from Black Cohosh Roots/Rhizomes. 33

Figure 3. Phenolic constituents from Black Cohosh Roots/Rhizomes. 35

Figure 4. Mixed HPLC-ELSD standard trace. 48

Figure 5. Calculations for preparative HPLC scale-up. 51

Figure 6. RAPD-PCR of N. American Cimicifuga species. 61

Figure 7. PCR profiles utilizing different primers to distinguish 62 C. racemosa from potential adulterants.

Figure 8. Freehand microscopical drawings. 63

Figure 9. Scanning electron microscope pictures. 65

Figure 10. Solvent extraction and partitioning of plant material 67 to yield non-polar fractions for isolation from the EtOAc-soluble fraction

Figure 11. Solvent extraction and partitioning of plant material 68 to yield polar fractions for isolation from the BuOH-soluble fraction

Figure 12. Summary of XAD-2 fractionation and bioassay results 70

Figure 13. HPLC-ELSD of XAD-2 Fractions 71

Figure 14. Pilot CHP20P separation summary HPLC-ELSD 74 results F 21-23 to 21-29.

Figure 15. CHP20P separation of XAD-2 fractions 5 through 7 75

Figure 16. HPLC-ELSD results of CHP20P fractions 76 xvi

LIST OF FIGURES (continued)

Figure 17. LC-MS screening of CHP20P fractions for compounds 80 with an odd number of nitrogen atoms

Figure 18. HPLC-ELSD of sephadex LH-20 separation of F-12. 82

Figure 19. Semi-preparative RP-HPLC isolation of 1. 86

Figure 20. Qualitative analytical HPLC-ELSD results of 87 semi-preparative HPLC-ELSD of separation of fraction G-15.

Figure 21. Isocratic analytical HPLC-ELSD of 2. 89

Figure 22. QTOF-2 HREIMS of 1 with fragmentation pattern. 95 Figure 23. 13C-NMR APT of 1 96

13 Figure 24. Gated C-NMR spectrum of (1) in D2O 97

Figure 25. HMBC of cimipronidine (1) in D2O 98

Figure 26. HSQC of 1 in D2O 99

1 1 Figure 27. H- H gCOSY spectra of (1) in D2O 100

Figure 28. The NOESY correlations of 1 101

1 Figure 29. H-NMR spectra of 1 in d6-DMSO and D2O 102

Figure 30. Fukinolic acid (2) 1H-NMR spectrum in MeOH-d4 105

Figure 31. 13C-NMR spectrum of fukinolic acid (2) in MeOH-d4 106

Figure 32. LC-MS/MS identification of 3 109

Figure 33. Proton NMR spectrum of actein (3) in pyridine-d5 110

13 Figure 34. C-NMR spectra of actein (3) in pyridine-d5 111 xvii

LIST OF FIGURES (continued)

Figure 35. Carbon NMR of (4) isoferulic acid in pyridine-d5 114

Figure 36. Proton NMR of isoferulic acid (4) in pyridine-d5 115

Figure 37. Positive ESI LC-MS of a fraction containing 118 cimipronidine (1) and its analogues

Figure 38. Positive-ion electrospray tandem mass spectral 119 data to identify N-methyl cimipronidine

Figure 39. Phase II clinical capsule analysis standard curves 125

Figure 40. HPLC-ELSD overlay of Black Cohosh extracts 127 using different solvents

Figure 41. Positive-ion electrospray LC-MS chromatographic 128 overlay of Black Cohosh extracts using different solvents

Figure 42. Electrospray LC-MS (negative-ion mode) Total Ion 134 chromatogram (TIC) of the Phase I clinical extract

Figure 43. Clinical capsule comparison 135

Figure 44. Comparison of clinical starting material triterpene 138 content to Asian species

Figure 45. HPLC-EL:SD trace showing the polarity of the 147 BuOH fraction

Figure 46. Potential complexation of cimipronidine 148 and fukinolic acid

Figure 47. Other guanidine isolates from higher 151

xviii

SUMMARY

This work was conducted within Project 1 of the UIC/NIH Center for

Botanical Dietary Supplements Research. The focus of project one is the

standardization of botanicals for womens’ health for FDA-style Phase I and II

clinical studies. This work details primarily the botanical and chemical aspects of

botanical dietary supplement standardization. The specific aims of this work are;

1) To effectively review the existing literature on the chemical, biological and botanical aspects that would be essential for the standardization of black cohosh

(Cimicifuga racemosa (L.) Nutt.). 2) To botanically standardize a black cohosh extract using the appropriate microscopical, taxonomic and genetic fingerprinting

techniques available to the UIC/NIH Center, 3) To isolate and characterize

known and novel constituents for the chemical standardization and biological

standardization of a black cohosh extract, and to develop preparative

methodologies for isolation and characterization where needed. 4) To chemically standardize a Black Cohosh extract using the appropriate techniques such as

HPLC-ELSD, and develop methodologies where appropriate. 5) Ultimately, to apply the aforementioned aims to best formulate a standardized black cohosh extract for clinical study.

Figure 1. Flowering raceme of Cimicifuga racemosa (L.) Nutt. 1.0 INTRODUCTION

A component of the Dietary Supplement Health and Education Act

(DSHEA) of 1994 led to the establishment of the Office of Dietary Supplements

(ODS) and the National Center for Complementary and Alternative Medicine

(NCCAM) within the National Institutes of Health.1 The purpose of ODS and

NCCAM, as mandated by Congress, is to provide accurate information on alternatives to conventional means of healthcare to the public. One of the methods for achieving this goal is the funding of extramural research. In 1999,

ODS and NCCAM jointly funded two centers, one at the University of Illinois at

Chicago (UIC) and one at the University of California-Los Angeles (UCLA), for

research of botanical dietary supplements. The UIC/NIH Center was organized to

focus its research on botanicals traditionally used for womens’ health concerns

(i.e., menopause, PMS). Black cohosh, Cimicifuga racemosa (L.) Nutt. (syn.

Actaea racemosa L.),a was chosen as one of the plants to be developed for the

clinical component of the Center based on it’s widespread use for relief from

symptoms related to menopause.2-12 The unique purpose of the study being that it would be one of the first extract dosage forms standardized botanically, biologically and chemically to undergo FDA-style phase I and II clinical trials.

Menopause, a normal biological event, is generally regarded as the time in a woman’s life when she stops menstruating, usually between the ages of 48 and

52. In the year 2000 approximately 31.2 million American women were over the age of 55, that number is estimated to be 45.9 million by the year 2020.13

Because of the average life expectancy over the past few decades, it is

a racemosa is not an officially accepted synonym by the International Botanical Congress

2 estimated that most women will live one-third to one-half of their lives after menopause.

While menopause is a natural condition, the decrease in hormonal concentrations that occur during menopause and hereafter, can increase the risk of osteoporosis (bone loss) and heart disease. Additionally, more common throughout menopause are atrophic and psychic symptoms, which occur in pre- and peri-menopause. The development of menopausal symptoms involves many complex interactions between exogenous and endogenous factors. Menopausal symptoms result as the ovaries produce less estrogen and progesterone, these decreases initially result in irregular periods, and other symptoms such as hot flushes, vaginal dryness and sleep disturbances. Via this process, the pituitary gland is prompted to increase follicle stimulating hormone (FSH) production to try to stimulate the ovaries to produce more estrogen.14

Basic endocrinology research has elucidated several pathways for managing the climacteric symptoms related to menopause. Premarin™

(Conjugated Pregnant Mare Estrogens) was introduced in the 1960’s with the idea of managing climacteric symptoms (hot flushes, night sweats, bone loss and vaginal atrophy) through steroidal hormone therapy, commonly referred to as estrogen replacement therapy (ERT). Hormone replacement therapy (HRT), is estrogen replacement, like Premarin™, administered in conjunction with progesterone or synthetic progestagens. Currently 38% of postmenopausal women in the U.S. use HRT during menopause.14, 15 ERT is prescribed for women who have had a hysterectomy, others are generally prescribed combined HRT to

3 prevent endometrial hyperplasia. Oral estrogen preparations include conjugated equine estrogens (CEE), synthetically derived piperazine estrone sulfate, estriol, micronized estradiol, and estradiol valerate. Alternatively, transdermal estradiol is available in the form of a patch or gel, a slow-release percutaneous implant, or an intranasal spray. Intravaginal estrogens include topical tablets, rings, creams and pessaries. Oral estrogen preparations result in up to ten-fold higher concentrations of circulating estrone sulfate than do transdermally administered estradiol of comparable or even higher doses. The primary indication for systemic HRT is, therefore, the relief of moderate-to-severe vasomotor symptoms. Vaginal estrogens are effective for urogenital symptoms.

The Women’s Health Initiative, a multi-armed study, one of the study arms, using more recent HRT formulations, such as Prempro™ (conjugated estrogen and progesterone), demonstrated an increased risk toward development of breast cancer, endometrial cancer, gallbladder disease, coronary heart disease, stroke, and embolism.16-25 A review of all observational studies found that, in women taking estrogen alone, the relative risk of developing breast cancer was 1.32 26, for women taking estrogen plus progestogen, the relative risk was 1.41 for developing breast cancer, in comparison with women who did not take hormones.27 Thus, there is an imperative to develop safer alternatives to

HRT to relieve climacteric symptoms.28-31

Development of medical systems incorporating plants can be traced back as far as recorded history.32 According to the World Health Organization (WHO), approximately 65% of the world’s population use plants as their primary modality

4 of healthcare.33-37 The four primary ways plants are used in health care are: 1) to

isolate bioactive compounds for direct use as drugs, 2) to isolate compounds for

use as lead compounds in semi-synthesis, 3) to use isolated agents as

pharmacological tools and 4) to use the a) whole plant and/or b) plant part(s) as

a herbal remedy, and/or c) an extract of a) and b). According to estimates, the

use of herbal remedies in the US increased as much as 380% between 1990 and

1997.38 Despite recent data to suggest a decrease in usage of complementary and alternative medicines, the relatively of herbal remedies in the USA is currently a $4 billion dollar-per-year industry.39, 40 National surveys have shown

that the average user of herbal products is more inclined to be a college-

educated female between the ages of 35 and 49.38

5

2.0 STATEMENT OF PROBLEM

With over 50 years of clinical research, supporting the use of black cohosh

(Cimicifuga racemosa (L.) Nutt. Syn. L.) for climacteric

symptoms related to menopause,9, 41-46 (i.e. hot flushes, depression) it’s

mechanism of action is unclear. Black cohosh has long been believed to be

estrogenic, with estrogenicity attributed to the presence of estrogenic

isoflavones, commonly referred to as phytoestrogens. A study done at the

UIC/NIH Center for Botanical Dietary Supplements Research confirmed no direct

estrogenic effect of black cohosh.47 Additional studies have shown no presence

of estrogenic isoflavones.48, 49 A goal of the UIC/NIH Center for Botanical Dietary

Supplements Research is to prepare a standardized dosage form of black

cohosh for clinical studies. This extract will be standardized to specific biological,

botanical and chemical characteristics. The specific challenges addressed in this study are:

1. Chemical characterization of bioactive constituents of black cohosh

2. Development of methods to identify and isolate bioactive constituents

3. Revision and review of reference data for future investigations of bioactive

constituents

6

3.0 LITERATURE REVIEW

3.1 TAXONOMY OF BLACK COHOSH

Cimicifuga L., from the Latin “” (bedbug) and “fugare” (to banish) has

been used as a medicine across several cultures and continents. Common

names include:

Americas: Black cohosh, Black Snake-root, Macrotys, Rattleweed,

Rattleroot, Snakeroot, Bugbane, Squaw root, Squaw weed

China: Sheng Ma

3.1.1 Ranunculaceae

Often referred to as the buttercup family, Ranunculaceae, comprises roughly 2000 species of plants across 50 genera, mainly spread in temperate countries. Herbs are perennial or annual, sometimes subshrubs, herbaceous or woody vines. Leaves are basal and cauline, alternate, rarely opposite or whorled, simple or variously compound, palmately nerved, rarely penninerved, with or without stipules. Inflorescence appears as a simple or compound monochasium, dichasium, as a simple or compound raceme, or with solitary flowers. Flowers are bisexual, sometimes unisexual, actinomorphic, rarely zygomorphic, and hypogynous. Sepals show 3–6 or more, they appear free, petaloid or sepaloid, imbricate or sometimes valvate in bud. Petals are present or absent, usually 2–8 or more, free, usually with nectaries. Stamens are numerous, rarely few, and free. Filaments appear linear or filiform. Anthers are latrorse, introrse, or extrorse, with some sterile stamens becoming staminodes. Carpels are numerous or few,

7 rarely singular. The carpels are free and rarely connate to varying degrees.

Ovaries can possess one or many ovules. Fruits are follicles or achenes, rarely capsules or berries. Seeds are small, with abundant endosperm and a minute embryo. A majority of the species are herbs and lianas having large flowers.50-52

3.1.2 Genus Cimicifuga

The genus Cimicifuga L. includes 18 species, one of which is native to

Europe, six from North America, and the remainder from northeast Asia. If the genus is expanded into Actaea L., as is proposed on the basis of recent morphological and DNA sequencing studies, it would include a total of 24 species.53 In the phylogenetic study in question, Actaea was reclassified to include Cimicifuga and Souliea. However, for the present account, the genus

Cimicifuga remains distinct on the basis of no such formal change by the World

Conservation Union (IUCN) International Botanical Congress.54, 55 In North

America, the genus is comprised of six species; three Western species (C. arizonica Wats., C. elata Nutt., C. laciniata Wats.) and three Eastern species (C. americana Michx., C. racemosa (L.) Nutt., C. rubifolia Kearney).

The genus is morphologically characterized by the following: The herbs, are perennial. The rhizomes are robust, and creeping, with fibrous roots. The stem is terete. The stem appears apically and frequently with several branches.

Leaves are 1–3 ternately sect or subpinnately compound with a long petiolate.

The Inflorescence appears densely racemose, sometimes spicate, simple or branched. The rachis is densely glandular, pubescent and hairy. The bracts are

8 small, subulate to narrowly triangular. Flowers are dense, small, actinomorphic,

hermaphroditic or rarely unisexual. The plants are dioecious. There are 4 or 5

sepals which appear petaloid, white, obovate-orbicular, or caducous. Petals are

elliptic to sub-orbicular, entire, slightly concave or forked-lobed with 2 empty

anthers, rarely with a nectary. Stamens are numerous and filaments narrowly

linear to filamentous, with yellow anthers. The anthers are broadly ellipsoid to

suborbicular. One to eight follicles appear either stalked or sessile, oblong-

ellipsoid to obovate-ellipsoid, adaxially convex veined, with a beaked apex.

Seeds are few, yellowish brown, ellipsoid to narrowly ellipsoid.50-52

3.1.3 Species

Cimicifuga racemosa is an erect, smooth-stemmed perennial 1-2.5 meters

in height. Large compound leaves are alternately arranged and triternate on short

clasping petioles. Basal leaf petioles are grooved in young specimens, this

shallow, narrow sulcus in C. racemosa disappears as the petiole enlarges and matures, where it remains present throughout the life of the related Eastern North

American species C. rubifolia and C. americana.56 Terminal leaflets are acute, glabrous with sharp serrated margins often trilobate, occasionally bilobed. Fruits are ovoid follicles occurring sessile on the pedicel. The flowering structure, the raceme, is a long wand-like structure with showy white flowers. Flowers possess numerous characteristic stamens with slender filaments with distinctive white anthers.57 The roots are branched and knotted structures. The roots have a dark

brown exterior, but are usually internally white and mealy and are rarely brown

9 and waxy. The upper surface of the plant has several buds and numerous large

stem bases that are terminated frequently by deep, cup-shaped, radiating scars,

each which show a radiate structure. These bases are less frequently terminated

by fibrous strands. The lower and lateral surfaces show numerous root scars and

a few short roots with horny fractures. The plant gives a slight odor and has a

distinctive, bitter and acrid taste.58

3.2 BLACK COHOSH IN TRADITIONAL MEDICINE

The use of black cohosh as a medicine is generally traced back to native

American groups. References point to its use by the Oklahoma,b Delaware,

Cherokee, Penobscots (Algonquins) and Micmac tribes.59, 60 The ethnomedical

uses of the roots by these indigenous peoples include, but are not limited to, use

as an emmenagogue to treat rheumatism, as a tonic, diuretic, or anodyne to treat

kidney trouble, and to treat fever.

Use was not limited to indigenous peoples. Black cohosh was a well

known home remedy and widely used in mainstream medical circles, it was

included in a secondary list of the first United States Pharmacopoeia (USP) in

1820 as Cimicfuga serpentaria (snakeroot). The uses that are generally cited are

as a relief from rheumatism, astringent, sudorific, anodyne, repellant,

emmenagogue and subtonic. With a shift in the materia medica of the USP to

‘mainstream’ medicine, the use of black cohosh and interest in its properties

shifted toward “eclectic” or herbal medicine. John King, author of The Eclectic

b This is taken from the text, the use of the herb this far to the west is out of the range of distribution of C. racemosa

10 Dispensatory of the USA, made significant note of black cohosh, or as he

referred to it, as Macrotys. King claimed that Macrotys was most beneficial as a remedy for “abnormal conditions of the principle organs of reproduction in the female.” The text further states that, the root is “very efficacious in the treatment of chronic ovaritis, endometriosis, and menstrual derangements, such as amenorrhea, dysmenorrhea, menorrhagia, frigidity, sterility, threatened abortion,

uterine sub-involution, and to relieve severe after pains.” King worked closely in

Cincinnati with the Lloyd brothers who later put forth a drug treatise on Macrotys.

In this treatise, the Lloyd brothers pointed to the use of Macrotys as a remedy for

rheumatism and for correcting the “wrongs” of menstruation.61

3.3 EFFECTS ON CLIMACTERIC SYMPTOMS RELATED TO MENOPAUSE

With a history spanning almost 50 years of clinical study, mainly in

Europe, black cohosh is one of the more popular alternatives to hormone

replacement therapy (HRT) or Estrogen Replacement Therapy (ERT).62 Most of

the clinical research has been carried out on the commercially available

Remifemin®.63 The formulation and dosage of Remifemin® used in human

studies has changed over time as shown in table 1. However, other commercial

formulations are available, and have been clinically studied as evident in table 2.

However, most commercial products have not been clinically trialed.46 The

results of clinical studies have been measured using a variety of parameters.

Self-assessments, physician assessments and physiological parameters are

usually used together when designing studies to measure

11

Table 1. Black cohosh clinical studies

Author Year Extract/ Study n Outcome Measure/Result Study Design Formulation length /Dosage

Hernandez- 2003 BNO 1055 12 months 136 Combination therapy with Open, Munoz et al.,64 tamoxifen (20 mg) reduced Randomized, severity and incidence of hot Patient self- flushes. assessment Baier- 1995 Cimisan® T 4-8 weeks 157 Open, Jagodinski65 Tropfen, Uncontrolled variable dose Wuttke et al.,66 2003 Klimadynon® 3 months 62 Equipotent to 0.6 CE for relief Randomized, /BNO1055 of climacteric complaints and double-blinded, for bone resorption. No effect placebo on endometrial thickness. controlled ,multi- center. MRS Schotten67 1958 Remifemin® 3-4 weeks 22 Alleviation of neurovegetative Case series 20 drops and psychic complaints associated with menopause and pre-menopause. Kesselkaul68 1957 Remifemin® 2 weeks 63 Alleviation of climacteric Case series 60 drops complaints in 95% of patients. Nesselhut69 1999 Remifemin® 3 Months 28 Good to very good alleviation of Open, Tablets, 10 menopausal symptoms in Postmarket equiv. to 80% of study participants Surveillance 136 mg dried herb/day Foldes70 1959 Remifemin®, Unknown 41 31 patients of the verum group Placebo 3 tablets/day responded to the treatment with Controlled, a decrease in menopausal Open, complaints. Crossover, Patient Self- Assessment Starfinger71 1960 Remifemin®, 1 year 105 Decreased climacteric Case series 3-20 complaints without incidence of drops/day side effects or resulting in unphysiological bleeding. Liske et al.,72 2000 Remifemin®, 6 months 57 Alleviation of symptoms in both Double blinded, equiv. to 39 groups. Results similar after 3 randomized, or 127 mg months Good-Clinical dried Practice herb/day compliant, KMI, SDS, CGI Schlidge73 1964 Remifemin®, Variable 135 Case series Fluid extract 60 drops/day

12 Table 1 (continued). Black cohosh clinical studies

Author Year Extract/ Study n Outcome Measure/Result Study Design Formulation length /Dosage

Daiber74 1983 Remifemin®, 12 weeks 36 Alleviation of climacteric Open, KMI, CGI Fluid extract complaints (hot flushes, 80 drops/day insomnia, sweating and restlessness). Stolze75 1982 Remifemin®, 6-8 weeks 629 Alleviation of neurovegetative Open, Physician Fluid extract and psychological menopausal and Patient Self- 80 drops/day symptoms in 80% of patients Assessment Vorberg76 1984 Remifemin®, 12 weeks 50 Significant or highly significant Randomized, Fluid extract alleviation of menopausal Open, KMI, CGI, 80 drops/day (neurovegetative and psychic) POMS complaints, study included subjects contraindicated to Hormone Therapy. Warnecke77 1985 Remifemin®, 12 weeks 20 Significant alleviation of Randomized, Fluid extract symptoms (psychic and Open, KMI, 80 drops/day neurovegetative) in the black HAM-A, SDS, cohosh, conjugated estrogen CGI, and diazepam groups. Vaginal Karyopyknosis cytology of treatment group index, Eosinophil was comparable to estrogenic index. stimulation.

Heizer78 1960 Remifemin®, 2-18 66 Alleviation of menopausal Case series tablets 3- months (neurovegetative and psychic) 6/day complaints in 47% of patients with intact uteri and 35% with hysterectomies

Jacobson, et 2001 Remifemin®, 60 days 42* No change in median number Double blinded, al.79 tablets equiv. or intensity of hot flushes randomized, to 40 mg placebo- dried controlled, herb/day patient self- assessment, VAS, MSS Duker et al.,43 1991 Remifemin®, 2 months 110 LH suppression In vitro study tablets equiv. using blood from to 40 mg menopausal dried women taking herb/day black cohosh Stoll80 1987 Remifemin®, 12 weeks 26 Significant alleviation of Double-Blinded, Tablets climacteric symptoms (vaginal Randomized, equiv. to 8 atrophy, neurovegetative and Placebo- mg psychic complaints) in Controlled, KMI, extract/day comparison with estrogen and HAM-A, VMI placebo groups. (vaginal epithelium)

13 Table 1 (continued). Black cohosh clinical studies

Author Year Extract/ Study n Outcome Measure/Result Study Design Formulation length /Dosage

Lehman- 1988 Remifemin®, 6 months 15 Significant alleviation of Randomized, Willenbrock et Tablets climacteric symptoms in black Open, KMI al.,42 equiv. to 8 cohosh and drug treatment mg groups. No significant change extract/day in gonadotropin (FSH, LH) levels. Petho81 1987 Remifemin®, 6 months 50 KMI decreased significantly Open, KMI, Tablets, from 17.6 to 9.2, correlates with Patient Self- Unspecified a significant reduction in Assessment dose neurovegetative symptoms. Severity of subjective self- assessments of subject’s physical and psychological symptoms decreased.

Gorlich82 1962 Remifemin®, Variable 41 Alleviation of climacteric and Case series tablets, (258) vascular symptoms in 85% of variable dose patients

Brucker83 1960 Remifemin®, Variable 87 Alleviation of menopausal Case series tablets, (517) complaints variable dose Mielnik84 1997 Uncharacteri 6 months 34 Alleviation of climacteric Open, KMI zed extract, (neurovegetative) symptoms in 4 mg daily 76% of patients after 1 month

Georgiev85 1997 Uncharacteri 3 Months 50 Alleviation of climacteric Open, KMI, zed extract, symptoms in 90% of patients. HAM-A, VMI Unspecified Increase in vaginal cell dose proliferation (VMI) in 40% of treated women. Liske et al.,72 2002 Unique C. 6 months 152 No direct systemic estrogenic Drug racemosa effect on serum levels of FSH, Equivalence preparation, LH, SHBG, prolactin, & 17-ß Trial, KMI, SDS, equiv. to 39 estradiol. No change in vaginal CGI or 127.3 cytology. Higher dose had a mg/day more significant reduction in KM index after 6 months. Significant reduction with both doses in neurovegetative and psychic complaints.

*-all with breast cancer history

14

Table 2. Commercially available products that have been evaluated in clinically studies.*

Brand Name/ Delivery Effective Indications Manufacturer form ingredients (Country)

Cefakliman® capsules Concentrated menopausal and premenstrual symptoms, mono/Cefak extract from dysmenorrhea (Germany) Cimicifuga Root Cefakliman® solution Ethanolic extract menopausal and premenstrual symptoms, mono/Cefak from Cimicifuga dysmenorrhea Root Cimipure-PE® capsules Dried climacteric symptoms related to menopause 2.5/ Pure World hydroalcoholic (USA) extract Cimisan®-T/ blister Concentrated premenstrual and dysmenorrheic as well as APS pack extract from neurovegetative symptoms from menopause (Germany) Cimicifuga Root Cimisan-T/APS drops Liquid extract premenstrual and dysmenorrheic as well as neurovegetative symptoms from menopause Femilla®/ tincture Hydroalcoholic neurovegetative symptoms with painful menstruation Steigerwald extract from (dysmenorrhea) as well as during menopause (Germany) Cimicifuga root Klimadynon®/ blister Concentrated menopause-caused neurovegetative symptoms Menofem/BNO pack extract from 1055/Bionorica Cimicifuga Root (Germany)

Klimadynon®/ solution Liquid extract menopause-caused neurovegetative symptoms Menofem/BNO 1055/ Bionorica Remifemin dragees Extract of menopausal symptoms; such as hot flushes, sweats, Plus®/ Hypericum depressive moods and psychovegetative problems Schaper & (aerial parts) and such as despondency, inner tension, irritability, lack Brümmer† Cimicfuga root of concentration, insomnia, fear and/or nervousness; (Germany) premenstrual vegetative symptoms Remifemin®/ tablets Concentrated menopausal symptoms; mild dysfunction after Schaper & extract from ovarectomy or hysterectomy; to aid treatment with Brümmer† Cimicifuga Root sexual steroids; premenstrual neurovegetative and emotional problems; juvenile menstrual irregularities Remifemin®/ solution‡ Percolate extract menopausal symptoms; mild dysfunction after Schaper & of Cimicifuga root ovarectomy or hysterectomy; to aid treatment with Brümmer sexual steroids; premenstrual neurovegetative and emotional problems; juvenile menstrual irregularities

*-independent confirmation as to the identity and/or quality of formulations is not publicly available. †-previously marketed in the US through GlaxoSmithKline, after 6/2005 will be distributed by Enzymatic Therapy. ‡-no longer marketed in this dosage form.

15 psychological, neurovegetative, somatic and physiological markers of

menopause or in the case of the treatment groups, relief from menopausal

symptoms. More significant when evaluating studies, is the study design. More

weight should be placed on studies that follow good clinical practice, are double-

blinded, placebo-controlled and use characterized plant material and/or

extracts.41, 86

The standard for clinical studies are randomized, double blinded, multi-

center, placebo-controlled designed studies. With that in mind, three studies on

black cohosh have been randomized, double blinded and placebo controlled.44, 66,

79 The Jacobson study, spanning only 60 days of treatment, suggests that study

length may have limited study findings.79 Additionally, the study participants all had a history of breast cancer. The outcome of the study was that the median number of hot flushes decreased 27% in both the placebo and black cohosh groups. No significant differences were observed between groups. Thus, black cohosh, on the basis of this study, was no more effective than placebo in the treatment of hot flushes. The source and formulation of the extract used in the

Jacobson study was not specified.

A more recent open-labeled study using breast cancer survivors, treated with either Tamoxifen or a combination of an unspecified BNO 1055 extract with

Tamoxifen, suggested a significant reduction in the number and severity of hot

flushes, in the combination treatment group.64 In another randomized, double-

blinded and placebo controlled clinical study, black cohosh was compared with

standard conjugated estrogen (CE) therapy (0.625 mg/daily) with a duration of 12

16 weeks.44 Patients’ physical and psychological symptoms were measured every four weeks. The end result of the study was that the black cohosh treated subjects demonstrated a significantly lower index score with both the Kupperman

Menopausal (KM) and the Hamilton Menopausal (HAM-A) scales compared with placebo. This indicated a decrease in severity and frequency of hot flushes following black cohosh treatment. In addition, an increase in the number of estrogenized cells in the vaginal epithelium were noted in this study.44

In 2003, a study compared two different preparations of BNO 1055 extract

(defined in table 2, page 15) to CE therapy (0.625 mg/daily).66 The study

outcomes were: a patient self-assessment (diary and menopause rating scale

[MRS]), crosslaps (to measure bone resorption), bone specific alkaline

phosphatase (marker of bone formation), and endometrial thickness (measured

by ultrasound). Both BNO 1055 extracts were equipotent to CE therapy and

significantly greater than placebo in reducing climacteric complaints. Additionally,

the study demonstrated that both BNO 1055 preparations had beneficial effects

on bone metabolism in serum, specifically an increase in bone-specific alkaline

phosphatase, and no reduction in bone resorption, thus an increase in bone

turnover formation. No change in endometrial thickness was observed in both

BNO 1055 treatment groups, but it was significantly increased with CE therapy.

However, an increase in superficial vaginal cells was observed in both the CE

and BNO 1055 treatment groups. The authors of this study hypothesized that the

activities of both BNO 1055 preparations were similar to the effects of Selective

17 Estrogen Receptor Modulator (SERM) i.e., raloxifene, therapy on bone and

neurovegetative climacteric symptoms, without any uterotrophic effects.66

A recent double-blinded, randomized good clinical practice compliant

study used two dosages (low 39 mg, high, 127 mg) of Remifemin® (unspecified

formulation).87 The effectiveness was measured using a KM index, a self-

assessment depression scale (SDS), the clinical global impression scale (CGI),

serum concentrations of luteinizing hormone (LH), follicle stimulating hormone

(FSH), sex hormone binding globulin (SHBG), prolactin, and 17-β-estradiol and vaginal cytology. Reductions in the KM and SDS indices were significant. Global efficacy (CGI) was scored at good to very good in 80% and 90% of the patients in the treatment groups.87 No effect on serum hormone concentrations or vaginal

cytology was shown, prompting the authors of the study to suggest that black

cohosh does not have a direct estrogenic effect on serum hormone concentration

or vaginal epithelium.72 Two recent open studies using unspecified types of

extracts showed reduced KM index scores.64, 66 One study reported a significant

reduction in one month.84 While another study, also using the HAM-A scale,

recorded a 90% improvement in climacteric symptoms in menopausal women

after three months administration of an unspecified black cohosh extract.85

3.3.1 Conclusions of Clinical Studies of Black cohosh on Menopausal

Symptoms

Recently, an analysis interpreting the safety data from published clinical

trials, case studies, post-marketing surveillance studies, spontaneous report

18 programs and phase I studies were reviewed.86 The data obtained from over 20

studies, including over 2,000 patients, suggest that adverse event occurrence

with black cohosh is rare. The events are mild and reversible, the most common

events reported being gastrointestinal upsets and rashes.86 Recently, a summary

of case reports regarding potential liver toxicity of black cohosh, has received

much attention.88 In response to this report, NCCAM held a Workshop on the

Safety of black cohosh in Clinical Studies on November 24, 2004

(http://nccam.nih.gov/news/pastmeetings/blackcohosh_mtngsumm.pdf). Present at this workshop were a team of experts on black cohosh, spanning industry and academia, as well as basic and clinical science. The findings of this workshop were that “the group cautioned that there were problems establishing a cause and effect relationship on the basis of information presented (from the case studies). The hepatotoxicity might be coincidental or correlated with this adverse event but not necessarily be the cause”, and “at this time, there is no known mechanism with biological plausibility that explains any hepatotoxic activity of black cohosh”. It should be noted that no identification and/or characterization of black cohosh in the herbal preparations in question was made in any of the case reports1. Additionally, all black cohosh case report subjects were concurrently

using varied regimens of prescription pharmaceuticals.88

Placebo effects for any treatment of hot flushes have been well

documented, and self-reported measures of this endpoint are problematic. It

appears that the placebo effect peaks at 12 weeks.46 Thus, any study must

weight placebo effects appropriately. When clinical study design limitations are

19 considered along with Hawthorne effects,89 small sample sizes,42, 44, 67, 69, 70, 74, 77, 79, 84 and heterogeneous samples, it is not surprising that the literature in aggregate yields equivocal results concerning efficacy.46 However, in light of these studies,

the fact remains that the majority of clinical data, spanning over 2,000

randomized subjects has been positive in terms of efficacy and has shown no

incidence of liver toxicity, permanent or otherwise1. Based on the data provided, at the minimum, black cohosh appears to be a relatively safe alternative therapy to ERT or HRT, with a significant scientific basis to support its efficacy as an alternative therapy.

3.4 PHARMACOLOGY AND BIOCHEMISTRY OF Cimicifuga racemosa

EXTRACTS

Despite the extensive aforementioned clinical research, the mechanism of action of Black cohosh remains unclear. A majority of the older literature on black cohosh suggests a direct estrogenic effect. More recent studies have targeted the limbic system (hypothalamus) or more specifically the neurotransmitters involved in regulation of this system, as being responsible for the activity of black cohosh. Serotonin selective reuptake inhibitors (SSRI) have been used successfully to treat hot flushes in women with breast cancer, and there appears to be a link between estrogen and the regulation of serotonin receptors in the brain and regulation of tryptophan hydroxylase.90-93 Data fall into the following

categories: competitive estrogen receptor binding, receptor expression, plasma

hormone levels, hormonal secretion, osteopenia inhibition, uterine weight/estrous

20 induction, MCF-7 cell proliferation inhibition, and CNS effects/neurotransmitter

binding.

3.4.1 Competitive Estrogen Receptor Binding

The first report of estrogen receptor binding pointed toward clarifying the

mechanism of action of black cohosh.94 Additional studies were carried out to

further the substantiation of endocrine activity.43, 95 One significant factor frequently overlooked regarding black cohosh in these studies is the relative lipophilic nature of the extract used in making this determination. Extracts and fractions displaying estrogen receptor binding activity are of a significantly different chemical nature than the typical hydroalcoholic (ethanolic, methanolic or isopropanolic) extracts available for human use. For the purposes of this review lipophilic will be defined as having an affinity for, or, tending to combine with, or capable of dissolving completely or partially in lipids, solvents such as CHCl3,

EtOAc, will be classified as lipophilic. These lipophilic extracts have not been

administered clinically to date. A lipophilic (CHCl3) extract of the plant showed

relatively weak (35 µg/ml) estrogen receptor binding in rat uteri.94 One study also

confirmed the estrogen receptor binding activity of an unspecified lipophilic sub-

fraction (originating from CHCl3) on ovariectomized (ovx) rat uterine cells, with no

binding activity seen with a hydroalcoholic extract.43

Recent reports have contradicted the estrogen binding affinity of black

cohosh extracts.47, 96, 97 Using an unspecified root extract in an in vitro competitive

cytosolic estrogen receptor (from ovx rat livers) binding assay with

21 diethylstilbesterol (DES), an inhibitor of estrogen binding, showed a significant

inhibition of estradiol binding in the presence of DES.96 No binding was

demonstrated with the extract. A hydroalcoholic extract (50% aqueous ethanol)

was assayed for ER-binding to intact human breast cancer cell lines, MCF-7 and

T-47-D. No binding affinity was shown for the black cohosh extract. However,

binding activity was evident for other hydroalcoholic plant extracts.97 Using a methanol extract of black cohosh at a high concentration (200 µg/mL) on

recombinant diluted ER-α and ER-β, no binding activity was evident.47 A recent study using BNO 1055 (an unspecified hydroalcoholic liquid extract) showed contrasting results.98 The extract displayed dose-dependent competition with radio-labeled estradiol in both a porcine and human endometrial cytosolic estrogen receptor ligand binding assay (ER-LBA) system. By comparison, the extract did not displace human recombinant ER-α and ER-β. These findings prompted the authors to suggest that their product contains estrogenic compounds that have binding affinity for a putative ER-γ receptor. Another potential explanation for the activity is the presence of estrogenic fatty acids in black cohosh.99-101 Fatty acids, like linoleic acid have been shown to bind to

estrogen receptors, these fatty acids would most likely be found in more lipophilic black cohosh extracts.102-106

3.4.2 Receptor Expression

A lipophilic C. racemosa extract, was studied using luciferase expression

in a MCF-7 α- and β-estrogen receptor expressing subclone.107 The extract at a

22 concentration of 35 µg/mL, was shown to activate the transcription of estrogen-

regulated genes; the hydrophilic (hydroalcoholic) extract showed no activity. 107 A recent study measuring a commercial hydroalcoholic extract (standardized to

2.5% triterpene glycosides) at a low concentration (4.75 µg/L), increased estrogen receptor levels, an effect also produced by estradiol in human MCF-7 cells.108 An unspecified black cohosh extract tested in a transient gene

expression assay using HeLa cells co-transfected with an estrogen dependent

reporter plasmid in the presence of human ER-α or ER-β cDNA failed to show transactivation of the gene.109

3.4.3 Plasma Hormone Concentrations

The effect of black cohosh on serum concentrations of FSH and LH has

been studied extensively. Crude methanol and ethanol extracts, suppressed

plasma LH, with no effect on FSH in ovx rats.94, 95 Further fractionation of the

crude fraction resulted in the activity residing in an unspecified lipophilic fraction,

with the aqueous soluble fractions devoid of this activity.94 A later study in rats using lipophilic and hydrophilic extracts at high doses (140 and 216 mg/rat, i.p.) resulted only in LH suppression with a single injection of the lipophilic extract.43

The authors of another study reported LH suppression in ovx rats with an unspecified dose and extract type of C. racemosa.110 A recent study compared the effect of C. racemosa (BNO 1055) with that of estradiol on LH concentrations.107 Reduced concentrations were reported for the black cohosh

treated animals at 60 mg/day administered subcutaneously (s.c.) for seven days.

23 However, another study reported no estrogen agonistic effects on FSH, LH or

prolactin concentrations in ovx rats (DMBA model) with daily administration of a

40% isopropanolic extract (Remifemin®) for seven weeks.111

3.4.4 Hormonal Secretion

The effect of black cohosh on prolactin secretion in pituitary cell cultures was assayed using an unspecified ethanolic extract of C. racemosa.112 Basal and

TRH-stimulated prolactin levels were reduced significantly at doses of 10 and

100 µg/mL of the ethanolic extract. This effect was reversed by the addition of

haloperidol (a D-2 antagonist) to the cell cultures, suggesting dopaminergic

regulation of hormone secretion by C. racemosa.

3.4.5 Osteopenia Inhibition

The BNO 1055 black cohosh extract (60 mg/rat, s.c.) has been shown to

increase the expression of collagen I and osteocalcin in rats equivalent to ovx

rats treated with estradiol (8 µg).107 An additional study using the BNO 1055 liquid

extract demonstrated an osteoprotective effect, a reduced loss of bone mineral

density in rat tibia after three months of administration.113 A study using an

unspecified isopropanol extract of C. racemosa showed reduced urinary

parameters of bone loss. The authors of this study suggested this action was

similar to that of the raloxifene.114 A follow-up study using BNO 1055 (liquid

extract) in comparison with conjugated estrogen therapy, showed beneficial

effects of the extract on bone metabolism in humans, specifically an increase in

24 bone-specific alkaline phosphatase in serum.66 While no direct correlation

between species has been established it is of note that studies of the Asian

Cimcifuga species have demonstrated similar activity and may be of importance

for further investigation of this biological activity.115, 116

3.4.6 Uterine Weight/Estrous Induction

Uterine and ovarian weight increase, cell cornification and an increased

duration of estrous are generally considered evidence of endometrial estrogenic

activity. However, recently it has been debated that uterine weight is a poor

marker for endometrial effects.117 Three studies demonstrating that black cohosh

extracts (unspecified hydroalcoholic) increased the uterine weight of ovx rats.62,

110, 118 Two of these studies used an undetermined root extract.110, 118 One study using immature mice reported similar findings.62 By contrast, two studies on ovx

rats,107, 119 as well as four studies of immature mice, reported the converse.107, 109,

111, 120 One of these studies found that despite no increase in uterine or ovarian

weight, the duration of estrous was significantly increased by black cohosh.120 A study demonstrated no attenuation in uterine weight at variable doses (4, 40 and

400 mg/kg day) of a 40% isopropanol extract in ovx rats.121

3.4.7 MCF-7 Cell Proliferation Inhibition

The MCF-7 assay is a straightforward test for detecting weakly estrogenic

compounds. In this assay, oestrogen-dependent cells are grown in the presence

of test compounds. As with the estrogen receptor ligand binding assays, the

25 nature of the extract or fraction is a decisive factor in the expression of estrogen receptors (Improving the reproducibility of the MCF-7 cell proliferation assay for the detection of xenoestrogens).122( Lipopophilic extracts and fractions may contain constituents that have been reported to interefere with the estrogenic ligand binding assays, which may be responsible for receptor expression activity as well. 104, 108, 123, 124 An unspecified black cohosh extract failed to induce growth of

MCF-7 cells significantly when compared with untreated control cells.109 A study using isopropanolic and ethanolic extracts also failed to induce the growth of

MCF-7 cells.125

3.4.8 CNS Effects and Neurotransmitter Binding

A study using an unspecified extract of black cohosh (25-100 mg/kg, orally), measured its effects on mice body temperature and ketamine-induced sleep time. The study used bromocriptine (D-2 agonist) as a positive control, pretreated with sulpiride (D-2 blocker) and this suggested a receptor mediated dopaminergic effect.112 Another study was carried out to characterize neurotransmitter levels in the striatum and hippocampus after pretreatment in mice with the unspecified extract for 21 days.126 Serotonin and dopamine concentrations in the striatum were substantially lower in comparison with the control group. These studies have helped lead to the hypothesis that it is a dopaminergic action, rather than estrogenic-like activity that is responsible for the success of black cohosh in reducing climacteric symptoms.127 A study in the

UIC/NIH center, pointed to the effects of black cohosh being mediated by

26 serotonin (5-HT) receptors.121 Three different extracts (100% methanol, 40%

isopropanol, 75% ethanol) bound to the 5-HT7 receptor subtype, the methanol

extract demonstrated the greatest affinity with an IC50 ≤ 3.12 µg/mL. The 40%

isopropanol extract inhibited [3H]-lysergic acid diethylamide (LSD) binding to the

3 5-HT7 receptor with greater potency than [ H]-8-hydroxy-2(di-N-propylamino) tetralin (positive control) to the rat 5-HT1A receptor. Analysis of ligand binding

data suggests that the methanol extract functioned as a mixed competitive ligand

of the 5HT7 receptor. Further testing of the methanol extract in 293T-5-HT7 transfected HEK cells revealed elevated cAMP concentrations. These concentrations were reversed in the presence of the 5-HT antagonist methiothepin, indicating a receptor mediated process and possible agonist activity local to the receptor.121

3.4.9 Miscellaneous

A Black cohosh methanol extract protected s30 breast cancer cells

against menadione-induced DNA damage at variable concentrations, and

scavenged DPPH free radicals at a concentration of 99 µg/mL.128 With the recent

reports of black cohosh hepatotoxicity, black cohosh extract (100% MeOH) found

in the presence of glutathione (GSH) were screened using ultrafiltration liquid

chromatography-mass spectrometry-mass spectrometry (LC-MS-MS) for the

presence of glutathione (GSH) conjugates.129 The potential for toxicity stemmed

from the finding that catechols from black cohosh were activated to quinoid

metabolites. However, it should be noted that there appears to be no absorption

27 of these catechols across the intestinal epithelium by Caco-2 cells, only

triterpenoids demonstrated absorption by Caco-2 cells.130 A completed Phase I

study to measure the pharmacokinetics of black cohosh (75% EtOH extract) did

not detect any catechol metabolites in the blood or urine of ten women following

the oral administration of black cohosh extract (75% EtOH) at three dosages: 40,

80, and 120 milligrams.130

3.4.10 Conclusion on Extract Biochemistry and Pharmacology

While much research has been reported on the activity of black cohosh

extracts, there seems to be little consensus on exactly how the extracts, and as

important, the type of extract that best modulates the biochemical parameters

and targets of the climacteric symptoms related to menopause. It is of note that a majority of the research that supports direct estrogenic activity of black cohosh

(as indicated by ER binding assays, ER receptor expression and plasma hormone concentration) have been performed using lipophilic extracts which may contain other constituents, with estrogenic activity, not unique to black cohosh.

Additionally, these lipophilic extracts are not generally used in commercial products or clinical studies, thus limiting the relevance of these studies. With evidence to support minimal ER binding or receptor expression at high doses of non-lipophilic/hydroalcoholic extracts, it is difficult to maintain the position that these extracts mediate climacteric symptoms through a direct estrogenic effect.

In addition, the MCF-7 cell proliferation studies using hydroalcoholic extracts demonstrated no xenoestrogenic effect. Compounding the lack of estrogenic

28 data with the CNS activity that black cohosh extracts have shown, it seems more likely that black cohosh mediates menopausal symptoms by way of the CNS and a secondary messenger than by action at estrogen receptors.121, 131

3.5 PHYTOCHEMISTRY OF BLACK COHOSH

The natural product literature on Black Cohosh is vast and widely distributed. The literature holds an abundance of chemical information as well as the aforementioned pharmacological information pertaining to compounds and extracts. In order to quickly and thoroughly compile information on the analysis of natural products, the use of databases is necessary. The databases used for this work are Natural Products Alert (NAPRALERT),132 Chemical Abstracts (CA) and

Chemical Abstracts Registry, SCIFINDER, BEILSTEIN and MEDLINE among others.133 Isolated compounds from C. racemosa roots that have been reported in the literature are listed in table 3. The structures of some of these compounds are shown in figures 2 and 3.

29

Table 3. Compounds reported in the literature from C. racemosa roots based on NAPRALERT (percent yields in parentheses where available)

Isolate (yield) Reference Actaeaeposide-3-O-β-D-Xylopyranoside 134 Actein (0.0005-0.0156) 135, 136 Actein, 23-epi: 26-deoxy 137 Actein, 26(S): 137 Actein, 26-deoxy (0.0017) 137 Actein,2'-O-acetyl (0.0022) 138 Actein, deacetyl: 139 Acteol, 27-deoxy: acetyl: 140 Caffeic acid (0.01) 141 Caffeic acid methyl ester (0.00015) 142 Cimicifugic acid A 143 Cimicifugic acid A, dehydro: 144 Cimicifugic acid B 143 Cimcifugic acid B, dehydro 144 Cimicifugic acid C 145 Cimicifugic acid D 145 Cimicifugic acid E 143 Cimicifugic acid F 143 Cimicifugoside (0.0008) 139 Cimicifugoside H-1 (0.0034) 48 Cimicifugoside H-2 (0.01) 48 Cimicifugoside M (0.0005) 139 Cimicifugoside, 26-deoxy: (0.02) 138 Cimigenol, 12-β-21-dihydroxy: 3-O-α-L-arabinopyranoside 146 (0.00128) Cimigenol, 12-β-hydroxy: 3-O-α-L-arabinopyranoside 136 Cimigenol, 12-β-hydroxy: 3-O-β-D-xylopyranoside 136

Cimigenol,24(S): 3-O-α-L-arabinoside (0.0001) 48 Cimigenol,24(S): 3-O- β-D-xyloside (0.0009) 48 Cimigenol,25-anhydro: 3-O-α-L-arabinoside (0.0013) 138 Cimigenol,25-anhydro: 3-O- β-D-xyloside (0.0039) 138 Cimigenol,25-O-acetyl: 3-O-α-L-arabinoside (0.00024) 138 Cimigenol,25-O-acetyl: 3-O-β-D-xyloside (0.0079) 138

30 Table 3 (continued). Compounds reported in the literature from C. racemosa roots based on NAPRALERT (percent yields in parentheses where available) Isolate (yield) Reference Cimigenol, 25-O-acetyl-12- β-hydroxy: 3-O-α-L- 146 Arabinopyranoside (0.01) Cimigenol, 25-O-methyl: 3-O-α-L-arabinopyranoside (0.00196) 146

Cimigenol, 25-O-methyl: 3-O-β-D-xylopyranoside (0.01346) 147 Cimigenol-3-O-α -L-arabinopyranoside (0.00075) 146 Cimigenol-3-O-α-L-arabinoside (0.0019) 138 Cimigenol-3-O-β-D-xylopyranoside 134 Cimigenol-3-O-β-D-xyloside (0.0079) 138 Cimiracemate A (0.001) 142 Cimiracemate B (0.00006) 142 Cimiracemate C (0.00002) 142 Cimiracemate D (0.00002) 142 Cimiracemoside (0.0032) 148 Cimiracemoside A (0.05) 136 Cimiracemoside B 136 Cimiracemoside C 136 Cimiracemoside D 136 Cimiracemoside E 136 Cimiracemoside F (0.00076) 136 Cimiracemoside G (0.00019) 136 Cimiracemoside H (0.0009) 136 Cimiracemoside I (0.0008) 138 Cimiracemoside J (0.0019) 138 Cimiracemoside K (0.0025) 138 Cimiracemoside L (0.0018) 138 Cimiracemoside M (0.0016) 138 Cimiracemoside N (0.0079) 138 Cimiracemoside O (0.0013) 138 Cimiracemoside P (0.0013) 138 Cyclolanost-7-Ene, 9-19: 12-β-acetoxy-16-β-23;22- β -25- 147 diepoxy-3- β -23-24-trihydroxy: 3-O-β-D-xylopyranoside (0.0058) Cyclolanostan-3- β -ol, 9-19: 12- β -acetyl-oxy-16-β-23: 22-25- 146 diepoxy-23(R)-24(R)-dihydroxy: α-L-arabinopyranoside 22(R): (0.0003)

Cytisine,N-Methyl: 149

31 Table 3 (continued). Compounds reported in the literature from C. racemosa roots based on NAPRALERT (percent yields in parentheses where available) Isolate (yield) Reference Daucosterol-6'-linoleate (0.05) 138 Ferulic acid (0.01) 150 Formononetin* 94 Fukinolic acid 143 Glycerol-1-palmitate (0.013) 138 Isoferulic acid (0.07) Kaempferol* 151 Shengmanol, 23-acetoxy: 3-O-β-D-xyloside (0.22) 48 Shengmanol, 23-O-acetyl: 3-O-α-L-arabinopyranoside (0.0021) 152 Shengmanol, 23-O-acetyl: 3-O-β-D-xylopyranoside (0.01181) 147

Shengmanol, 23-O-acetyl: 3-O-β-D-xyloside (0.0019) 138 Shengmanol, 24-O-acetyl: (0.0012) 138 Shengmanol-3-O-α-L-arabinopyranoside (0.0012) 152

*-verification of these compounds from C. racemosa has not been confirmed

32 21 21 O OAc 24 22 O 18 OAc 18 24 20 27 20 27 23 25 23 25 17 17 19 O O 19 O O H OH H OH 26 26 H H 15 15 28 28 Xyl O Xyl O 30 29 30 29

actein (26S) actein (26R)

21 21 OAc O O 18 24 OAc 18 24 20 27 20 27 23 25 23 25 17 19 17 O O 19 O O H 26 H 26 15 15 28 28 Xyl O Xyl O 30 29 30 29

26-deoxyactein 23-epi-26-deoxyactein

21 21 O H 18 OAc 18 20 OAc 20 26 OH 17 17 19 O 19 O OH H H 15 O 27 15 28 OH 28 Xyl O Xyl O 30 29 30 29

cimiracemoside A 23-OAc-shengmanol-O-ß-D-xyloside

21 21 H 18 H O OAc 18 24 27 20 O 27 17 23 25 23 25 19 O R2 17 H 24 19 O O 15 O H 26 26

15 28 OH R O 28 1 Xyl O 30 29 30 29

26-deoxycimicifugoside R1= Xyl, R2= H, cimigenol-3-O-ß-D-xyloside (24S)

R1= Ara, R2=H, cimigenol-3-O-α-L-arabinoside (24S)

R1= Xyl, R2= Ac, 25-OAc-cimigenol-3-O-ß-D-xyloside (24S)

R1= Ara, R2= Ac, 25-OAc-cimigenol-3-O-ß-D-xyloside (24S)

Figure 2. Triterpenes isolated from black cohosh roots

33 O AcO O

O O O O

B-D-Xyl-O A-L-Ara-O

cimiracemoside I cimiracemoside N

O O AcO AcO 23R 23R O O O O O OH

Xyl-O 4'-O-acetyl-B-D-Xyl

cimiracemoside O cimiracemoside P

OAc O AcO 23R 24S 23R 24S O O O

OH OH RO RO

R= α-L-Ara, cimiracemoside J R=4’-O-acetyl-α-L-Ara, cimiracemoside L

R=ß-D-Xyl, cimiracemoside K R= 4’-O-acetyl-ß-D-Xyl, cimiracemoside M

21 21

18 18 HO 20 HO 20 OH O 17 O 17 O 19 O 19 O H H 15 OH 15 28 28 Xyl O Xyl O 30 29 30 29

cimicifugoside H-1 cimicifugoside H-2

Figure 2 (continued). Triterpenes isolated from black cohosh roots

34 O R1O CO2H R1 O CO2H R2O R2O HO CO2H R3 R1= H, R2= H, caffeic acid R = H, R = CH , isoferulic acid 1 2 3 OH R1= CH3, R2= H, ferulic acid

R1= OH, R2= H,R3=OH, fukinolic acid

R1= OH, R2= CH3, R3=OH, cimiciugic acid A

R1= OCH3, R2= H, R3=OH, cimicifugic acid B

R1= H, R2= H, R3=OH, cimicifugic acid C

R1= OH, R2= H, R3=H, cimicifugic acid D

R1= OCH3, R2= H, R3=H, cimicifugic acid E

R1= OH, R2= CH3, R3=H, cimicifugic acid F

O R3

O

O R1O OH

OR2 OH

R1= CH3, R2= H, R3=H, cimiracemate A

R1= H, R2= CH3, R3=H, cimiracemate B

R1= CH3, R2= H, R3=OCH3, cimiracemate C

R1= H, R2= CH3, R3=OCH3, cimiracemate D

Figure 3. Phenolic constituents that have been isolated from black cohosh roots

35 3.5.1 Triterpene Glycosides

Numerous chemical investigations of the roots of Cimicifuga have yielded

over 50 unique 9,19- cyclolanostane type triterpenes and glycosides.134, 135, 138, 140,

142, 146, 153-173 Actein was the first, discovered by Gennazi in 1952, as a result of his

experiments to explain the activity of Black Cohosh. Since that time over 40

triterpenes have been elucidated from C. racemosa.134, 136-139, 147, 148, 174, 175 The triterpene glycosides comprise approximately 0.5-2.0% of the dry weight of Black

Cohosh roots. The basic chemical composition of most of the triterpene glycosides is a 9,19-cycloartane triterpene skeleton, with different substitution patterns occurring. The position and variety of these chemical substitutions are further diversified by the stereochemical possibilities. Unique compounds like actein, which undergoes spontaneous mutarotation in solution, represent the cyclical or spiro- ketal variations of these constituents, which are of great interest to natural products hemists. Because of the distinctiveness of the triterpenes, and their relative abundance in commercially available extracts, they are generally regarded as the best choice as markers for chemical standardization.48,

144, 174

3.5.2 Phenolic Acid Derivatives

The cinnamic acid esters of fukiic and piscidic acid, fukinolic acid, cimicifugic acid, cimiracemates A-D, and cimicifugic acids A-F have been isolated from more-polar extracts and fractions of Black Cohosh roots. 142, 143, 150

Additional phenolic isolates include ferulic acid, isoferulic acid, caffeic acid and caffeic acid methyl ester. Esculetin, 4-O-acetyl-caffeic acid and sinapic acid have

36 been isolated from Asian Cimicifuga species, and are believed to be present in

C. racemosa on the basis of preliminary studies.176 Dehydrocimicifugic acid A,

and dehydrocimicifugic acid B were tentatively identified in C. racemosa by using

tandem LC-MS/MS.144

3.5.3 Flavonoids

Some debate surrounds the presence of the flavonoids biochanin A,

formononetin and kaempferol in the roots of Black Cohosh. An initial study in

1985, claimed the presence of formononetin in a lipophilic extract of the roots.94

An additional study in 1996 claimed to have found biochanin A in Black

Cohosh.177 The author of a study on isolation of kaempferol, later clarified that it

was only found in the aerial parts.151 Since those studies were published,

numerous other studies using more robust detection methods, have not detected

the purported flavonoids in the roots.48, 49 The presence of biochanin A and

formononetin would be considered significant because of their weak estrogenic

activity.

3.5.4 Miscellaneous

Fatty acids, tannins, resins, starches and sugars have also been reported

to occur Black Cohosh.132, 178 The sole report of the quinolizidine alkaloids cytisine

and N-methylcytisine from Black Cohosh is questionable, with no other reports of

these constituents found in Black Cohosh since that time.149 Concentrations of

the these alkaloids of Caulophyllum thalictroides (blue cohosh), a perennial herb

37 contained in dietary supplements marketed in the United States, have been well

established by analytical methods.179, 180 The teratogenic compounds anagyrine,

N-methylcytisine, baptifoline and taspine have been reported from Blue

Cohosh.179, 181 These constituents are known to interfere with the ability of a newly

fertilized ovum to implant in the uterus, damage the uterus and thyroid, and

cause severe birth defects in cattle and laboratory rats.181-189 Additionally, the compounds caulophyllosaponin and caulosaponin, appear to constrict coronary vessels, limiting blood flow to the heart and reducing its ability to pump.184, 186, 189

Population overlap with Black and Blue Cohosh is most likely responsible for this misidentification.182, 190

38

4.0 EXPERIMENTAL

4.1 PLANT MATERIAL

4.1.1 Procurement of Plant Material

The required National Park Service permits for collections in the Blue

Ridge Parkway and Great Smoky Mountains National Park are available in the appendix (section 10.3). Roots were separated from above ground parts on collection. Plant material was taken from the collection sites to the UIC

Pharmacognosy Field Station (Downers Grove, IL) to be washed, dried and milled to a coarse powder. Plant material was stored at the UIC Pharmacognosy

Field Station.

4.1.2 RAPD-PCR plant identification

Dried plant material (roots and aerial parts) was ground using a mortar and pestle under liquid nitrogen. Plant DNA extraction was carried out using the

QIAgen (Valencia, CA) DNeasy plant mini kit. The extraction procedure was based on the protocol provided by the supplier with modification to enhance overall yield of root material. Suspension and lysis buffers were added to the ground frozen plant material (10 mg), and the lysate was then applied to the

QIAshredder spin column to remove precipitates and cell debris by centrifugation. The flow-through fraction was then applied to the DNeasy mini spin column, centrifuged, washed, and eluted with the elution buffer (2 aliquots of

50 µL). Samples were all stored at -20ºC. The polymerase chain reaction (PCR)

39 was initiated by combining the following in each tube to make a total volume of

50 µL: 5 µL DNA template to PCR tubes (concentration and volume adjusted for

25 ng DNA in final volume of 50 µL), 37 µL DEPC water or nuclease-free water,

5 µL 10X Clontech (Mountain View, CA) Advantage 2 PCR Buffer, 1 µL 10 mM dNTP from invitrogen (Carlsbad, CA), 1 µL of working primer (1:10 stock primer solution, OPA-19: CAAACGTCGG, OPC-15: GACGGATCAG, OPA-05:

AGGGGTCTTG, Operon Ltd., [Huntsville, AL]), 1.0 µL Clonetech Advantage 2

Platinum Taq, final Volume is 50 µL. Then 50 µL Mineral Oil was added to each tube, vortexed and spundown. The positive control contained all of the above with 1.0 µL of Saccaromyces cerevisiae or mammalian DNA used as the template. The negative control contained all of the above without template DNA.

When preparing more than two samples, a master mix of the above ingredients was prepared without template DNA. Template was then added individually to each tube. Tubes were placed in a thermocycler (Perkin-Elmer (Wellesley, MA)

GenAmp PCR 2400) and run according to the following procedure: Repeat for 34 cycles: Denature: 94˚C for 1 min., Anneal: 36 ˚C for 1 min., Extension: 72˚C for 2 min. Final Extension: 72 ˚C for 10 min. All DNA was stored at -20˚C. Samples were run on a 2% agarose gel in 1X TBA containing Vistra Green NA Stain

(Amersham-Pharmacia Biotech, Inc. (Piscataway, NJ)). Loading dye was added to each sample in a ratio of 1:5 and 5.0 µL of each sample or control was added to a well. These samples were run with two wells that each contained 2.0 µL of

DNA ladder at 100 mV for 45 min to 1 hour. The resultant gel was digitally photographed under UV light.

40 4.1.3 Microscopic analysis

4.1.3.1 Light microscopy

The powdered plant material (BC 001) was placed on a microscope slide and treated with either water and washed with iodine to indicate the presence of starch, 10% phloroglucinol/HCl to indicate lignin, and/or chloral hydrate to verify calcium oxalate crystals.191 A cross sectional slice of roots was mounted on a slide with water. Cellular structures were documented by freehand drawings.

4.1.3.2 Scanning electron microscopic analysis

Powdered plant material (BC 001 and 010) and cross-sectional slices of dried plant material were placed on an SEM-aluminum stub with an adherent pad. Scanning electron microscope images were obtained on a JEOL-JSM 35C in secondary electron imaging (SEI) scanning mode with both platinum-palladium

(Pt-Pd) sputter coated using the secondary electron detector and uncoated stubs for observation in low vacuum mode. The waveform monitor was adjusted for the best exposure. The exposure settings were: RAPID 1, Photo horizontal @ 6, vertical @ 7, collector @ 5, load current 60-100 µA, magnification 1,000x-

10,000x.

41

4.2 CHROMATOGRAPHY

4.2.1 Solvent Partition and fractionation of Black Cohosh

HPLC grade methanol and acetonitrile were obtained from Fisher

Scientific (Fair Lawn, NJ) and used for all TLC and HPLC experiments. USP

grade (200 proof) ethanol was used for extracts. The bulk solvents chloroform, 1-

butanol, EtOAc, acetone and petroleum ether were distilled before use in

fractionation, column chromatography or TLC. Distilled, deionized, ultrafiltered milliQ™ water (Waters, MA) was obtained from the Bioassay Research Facility,

UIC College of Pharmacy (Chicago, IL).

4.2.2 Column Chromatography

For the isolation of compounds from the polar n-BuOH soluble fractions and the non-polar EtOAc fractions, either Amberlite XAD-2 (Sigma, St. Louis,

MO), MCI gel CHP20P (Mitsubishi, Tokyo, Japan), silica gel (Merck 60Å, 70-230 mesh), Sephadex LH-20 (Amersham Biosciences, Piscataway, NJ) and reverse phase HPLC were used to separate fractions. Once assay results were obtained for ‘active’ fractions, they were pooled or individually separated using one of the aforementioned techniques. The subfractions were subsequently submitted for bioassay. From this point due to the large generation of subfractions produced from these chromatographic techniques, the fractions were purified by semi- preparative RP-HPLC. All subfractions were analyzed by analytical RP-HPLC with inline PDA and ELSD detection.

42

4.2.3 Thin-layer chromatography (TLC)

Fractions and pure isolates were spotted on 0.25 mm thick pre-coated F254 normal phase glass-backed silica gel plates (20 x 20 cm; Merck, Darmstadt,

Germany). Plates were cut to 10 cm in length, and various widths. The solvent systems in table 4 were used. TLC chromatograms were scanned at 600 dpi (.tif file). Most TLC chromatograms were sprayed with either sulfuric acid (10% in

EtOH), ninhydrin, or anisaldehyde and subsequently heated by a heat gun or a

Fisher oven. Heating continued until spots ranging throughout the spectrum of available colors appeared on the white background of the plate. Additionally UV

(λ-254 and 365 nm) light was used for general chemical detection.

Table 4. Standard TLC Solvent Systems used for fraction control, isolation and structure elucidation.

SSE= EtOAc based

SSE6 EtOAc-MeOH 90:10 SSE7 EtOAc-MeOH-H2O 87:13 SSE8 EtOAc-MeOH-H2O 77:15:8 SSE9 EtOAc-MeOH-H2O 67:25:8

SSC= CHCl3 based

SSC6 CHCl3-MeOH-H2O 85:15:0.5 SSC7 CHCl3-MeOH-H2O 80:19:1 SSC8 CHCl3-MeOH-H2O 75:24:1 SSC9 CHCl3-MeOH-H2O 60:39:1

43 4.2.4 High-Performance Liquid Chromatography (HPLC)

HPLC was carried out using a Waters Delta 600 convertible unit with an inline 996 photodiode array detector (PDA), Waters 717 autosampler,

Millennium32™ chromatography manager, YMC-ODS AQ columns were used for both analytical (4.6 x 250mm, 5 micron) and semi-preparative (20 x 250 mm, 10 micron) purposes, as indicated. A Waters Delta-Pak RP-18 guard column was used for the analytical HPLC. An inline Sedex 75 Evaporative Light Scattering

Detector (ELSD) (Sedere, France) was used at variable nitrogen pressure and variable nebulizing temperature, with the output signal (mV) connected to the

Millennium32™ software through a SAT/IN analogue box. MeOH, ACN, both

HPLC grade were purchased from Fisher Scientific. TFA was purchased from

Sigma. Water was drawn from a Millipore reverse osmosis NANOpure™ unit.

HPLC conditions for analysis are summarized in table 5.

44 Table 5. Analytical HPLC-ELSD conditions and method parameters

Conditions for triterpene quantitative analysis (method A):

ELSD Nebulization Temperature : 43 °C Gain: 11 N2 Pressure: 3.4 Bar

Column: YMC ODS 250 x 4.6 mm, 5 micron Flow 1.6 ml/min

A- ACN, B-H2O, C-0.05% TFA/H2O

Gradient Elution (all gradient curves linear)

Time %A %B %C 0 20 80 8 20 80 15 32 68 55 64 36 65 85 15

Conditions for phenolic acid/polar constituent analysis (method B):

ELSD Nebulization Temperature : 61 °C Gain: 11 N2 Pressure: 2.9 Bar

Column: YMC ODS 250 x 4.6 mm, 5 micron Flow 1.0 ml/min

A- 0.05% TFA/5% MeOH/ H2O, B- 0.05% TFA/MeOH

Gradient Elution (all gradient curves linear) Time %A %B 0 97 3 50 25 75 61 25 75 62 10 90 67 10 90

45 4.2.4.1 Analytical HPLC analysis

Milled authenticated plant material (BC009) Black Cohosh roots collection

material was extracted with MeOH, 40, 60, 75% EtOH and 40% Isopropanol to

determine the optimal conditions for the clinical extract. Extracts were prepared

by exhaustively extracting ca. 1 g of plant material in 10 ml of the previously

mentioned solvents. Extracts were concentrated in vacuo until dry. A mass of

extract, usually 100 mg, was weighed in a 10 mL class A volumetric flask, 9 mL

of HPLC-grade MeOH was added. The sample was sonicated for 5 min. Each

sample was appropriately diluted then syringe filtered with 0.22 µm sterile filters.

Additionally, clinical extracts were provided in bulk by our collaborators from

BC009 for the Phase I clinical extract. Quantitative analysis of clinical samples

was performed in duplicate. Standards for quantitative analysis of the clinical

samples were made from serial dilutions of 1.0 mg/mL stock solution of 23-epi-

26-deoxyactein. A working stock was prepared by making a 1:5 dilution of the

stock solution in MeOH. The dilutions for the standard curve were prepared by

diluting the 400, 300, 200, 100, 50, and 25 µL of working stock solution in HPLC

MeOH to make the final volume 0.5 mL. The standards were syringe filtered

through 0.22 µm sterile filters. Standards were injected in duplicate. The dried

extracts were submitted for bioassay in the 5HT7 binding assay performed by

Project 2 of the Center. ELSD output is measured in mV, a power function based on droplets analyzed through the detector. Thus, the formula to calculate the percentage of 23-epi-26 deoxyactein in the samples is:

Log(c) = −m × Log(AUC) + b

46 Where c is the mass of sample per 10 µL injection, m is the slope of the standard curve, AUC is the area under the curve, and b is the y-intercept of the standard curve. From the standard curve the r2 value must be greater than 0.995,

or linearity between peaks cannot be assumed. Once this criterion was

established the other marker triterpene glycosides present in the extract, as

specified by the UIC Center, specifically 26-deoxyactein, actein-(26R) and actein-

(26S) were calculated as 23-epi-26-deoxyactein using a 1:1 ELSD response.192,

193 Retention times of the UIC Center marker compounds were determined by

running a mixed standard including 26-deoxyactein, actein-(26R) and actein-

(26S) were calculated as 23-epi-26-deoxyactein.

For assessing diurnal and geographical variation of Black Cohosh roots,

nineteen reference standards were used. The reference standard

cimiracemoside A was purchased from Chromadex Inc., (Laguna Hills, CA). The

reference caffeic acid was obtained from Sigma (St. Louis, MO). The reference

standards kaempferol and formononetin were obtained from Indofine Chemical

(Somerville NJ). The reference standards actein-(26S), actein-(26R), ferulic acid,

isoferulic acid, cimicifugoside H-1, cimicifugoside H-2, 26-deoxycimicifugoside,

23-epi-26-deoxyactein, 23-OAc-shengmanol-3-O-β-D-xyloside, 26-deoxyactein,

25-OAc-cimigenol-3-O-α-L-arabinoside, 25-OAc-cimigenol-3-O-β-D-xyloside,

cimigenol-3-O-α-L-arabinoside, cimigenol-3-O-β-D-xyloside were isolated and

purified from Cimicifuga racemosa roots. An HPLC-ELSD chromatographic trace

of the standards is detailed in figure 4. The identities of the isolates were

characterized by means of spectral (NMR and MS) analyses and/or X-ray

47 200. 200. 400. 600. 400. 600. 00 00 mV 0. 0. 0. 0. 00 00 00 00 00 00 00 00

0. 0. 0. 0. 0. 0. (24 x xyloside(24 cimigenol-3-O- ß-D-xylos 10) 26-deoxycimic cimiracemoside A,7)formononetin,8) ferulic acid,3)isoferulic4) compounds, elutedusingmethodA.The Figure 4 00 00 00 00 00 00 y loside R ,25 5. 5. 5. 5. 5. 5. 00 00 00 00 00 00

R 1 ), 15)25-OAc-cimigenol i . HPLC-ELSDchromatogramus de, 13)23- 10. 10. 10. 10. 10. 10. S 00 00 00 00 00 00

), 18)ci 2

α 3 15. 15. 15. 15. 15. 15. -L-arabinos 00 00 00 00 00 00 i fugoside, 20. 20. 20. 20. 20. 20. migenol-3- epi 00 00 00 00 00 00 -26-deoxyactein (24 25. 25. 25. 25. 25. 25. ide(24

44 11) actein(26 00 00 00 00 00 00 O 30. 30. 30. 30. 30. 30. 5 R - 00 00 00 00 00 00 6 α ), 17)25-OAc-cimigenol-3-O-ß-D- 7 -L-arabinoside, 19)cimigenol-3-O-ß-D- -3-O-ß-D-xylos

cimicifugoside H-2, 8 M M M M M M 48 35. 35. 35. 35. 35. 35. i i i i i i nut nut nut nut nut nut 9 cimicifugoside H-1,9)actein(26 00 00 00 00 00 00 es es es es es es 10

compounds are:1)caffeicacid,2) 11

S 1212 40. 40. 40. 40. 40. 40. ing themixedstandardof19 ), 12)23-OAc-shengmanol-3-O- 1313 00 00 00 00 00 00 S ,25 45. 45. 45. 45. 45. 45. 14 ide(24 00 00 00 00 00 00 S ), 14)26-deoxy 50. 50. 50. 50. 50. 50. 00 00 00 00 00 00 R 5) kaempferol,6) ), 16)25-OAc- 55. 55. 55. 55. 55. 55. 1515 00 00 00 00 00 00 1616 60. 60. 17 60. 60.

60. 17 60. 17 00 00 00 00 00 00 a 65. 65. 65. 65. 65. 65. ctein R 00 00 00 00 00 00 1818

), 19 70. 70. 70. 70. 70. 70. 00 00 00 00 00 00

crystallography. The purity was characterized by HPLC. The structures of these

reference standards are shown in figures 2 and 3 (pages 33-35). All reference

standards were dissolved in 10 mL of methanol at ranges of concentrations from

0.10–0.50 mg/mL. Serial dilutions were prepared as needed for the standard

curve. Standard stock solutions were stored at -20 ˚C and brought to room

temperature before use. Fourteen wild-crafted root collections (BC001, 005, 007,

010, 012, 014, 031, 034, 036, 038, 072, 089, 090, 092) were extracted exhaustively with MeOH, concentrated in vacuo to yield dried, powdered extracts. Each sample was prepared by dissolving approximately 100.0 mg ± 5.0 mg in 10 mL of MeOH, and syringe filtered with a 0.22 µm filter. Again, the

formula to calculate the percentage of the nineteen standards in the samples is:

Log(c) = −m × Log(AUC) + b

Where c is the mass per 10 uL injection, m is the slope of the standard curve, AUC is the area under the curve, and b is the y-intercept of the standard curve. From the standard curve the r2 value must be greater than 0.995, or linearity between peaks cannot be assumed.

49

4.2.4.2 Semi-Preparative HPLC

Semi preparative HPLC to purify compounds was performed using a

YMC-ODS AQ (20 x 250 mm, 10 µm) column. Once a retention time and the

corresponding elution conditions were established for a peak of interest in the

aforementioned analytical gradient elution method, an isocratic analytical method

was developed to optimize the separation and resolution of the peak/constituent

of interest. Once the isocratic gradient conditions were established, the equations

in figure 5 were used for scaling up the preparative column and determining the

appropriate flow rate and concentration of analyte.194 The amount of sample injected on to the column was dependent on the solubility of the analyte in the injection solvent and the relative polarity of the injection solvent comparable to the mobile phase. If the injection solvent is substantially more or less polar in comparison, the column will be volume overloaded and the sample resolution will broaden at injection. In an isocratic system this broadening problem, is not improved as it can be in gradient systems. Thus, for optimal resolution the injection solvent should be slightly less polar than the chromatographic mobile phase in a reverse phase HPLC system.195

50 L1

r1 ƒ1 Column 1-Analytical

L2

r2 ƒ2

Column 2-Preparative

2 ƒ1 r1 x1 x2 1 = 2 2 = 2 • ƒ2 r2 π •r1 π •r2 CL

flow= ƒ, column radius=r, column length =L, concentration or amount= x, ratio of column lengths= L2/L1= CL

Figure 5. The calculations necessary for preparative HPLC scale-up method development from analytical HPLC. The optimal flow rate is determined from an analytical HPLC method, this rate is used to determine the sample concentration and flow rate for the optimization of preparative HPLC.

51

4.3 SPECTROSCOPIC METHODS

4.3.1 Nuclear magnetic resonance spectroscopy

NMR spectra were recorded on Bruker Avance™(Billerica, MA) 300, 360,

400 or 500 MHz instruments where indicated. Dr. David C. Lankin in the

Department of Medicinal Chemistry and Pharmacognosy maintained the 300 and

400 MHz instruments. The 360 and 500 MHz instruments were maintained by Dr.

Robert Kleps of the Research Resources Center (RRC, UIC). Spectra were

referenced internally to TMS (Pyridine-d5, MeOH-d4, DMSO-d6). Offline

processing was performed with either NUTS™ (NMR utility transform software;

Acorn NMR Inc.) or MestR-C™ (www.mestrc.com). Line resolution of

experimental data was enhanced by Lorentz-Gauss (LG) transformation where

indicated. Online data processed with Bruker Software. Simulation experiments

to determine relative stereochemistry were carried out with Perch (Kuiopo,

Finland) and ACD™ software (www.acdlabs.com). NOESY relaxation delay time

was D1=1.5 seconds, mixing time D8=0.60 seconds.

4.3.2 Mass spectrometry

Drs. R.B. van Breemen and D. Nikolic (Core C of the UIC/NIH Center) in the UIC Botanical Center, Project 3, herein provided all mass spectra, including exact mass measurements and tandem mass spectra. Nominal mass, exact mass and LC-MS data were obtained using a high-resoultion Waters Micromass

(Manchester, UK) Q-Tof2 resolution hybrid orthogonal angle time-of-flight tandem

52 LC mass spectrometer equipped with electrospray ionization (ESI) and featuring

a quadrupole mass filter and collision cell. Argon was used as the collision gas.

The LC system was a Waters Alliance™ HPLC. Mass spectra were analyzed

using Masslynx™ software version 3.4.

4.3.3 Other Techniques

Optical rotations were measured with a Perkin-Elmer™ 241 polarimeter

(Perkin-Elmer, Inc. MA.). UV spectra were obtained with a Beckman™ DU-7

Spectrophotometer. Samples were loaded into a quartz cell (path length 1.0 cm).

FTIR spectra were obtained with a Jasco™ 4390 (Tokyo, Japan) equipped with a

Golden Gate™-ATR.

4.4 BIOLOGICAL ACTIVITY

Drs. Judy L. Bolton, J.E. Burdette, J. Liu, R. Ruhlen and C. Celimine in the

UIC Botanical Center, Project 2, herein provided all biological activity data.

4.4.1 Estrogenic Activity

Percent estrogenic values at given concentrations, for both α- and β-

estrogen receptors (ER) were determined by Project 2 within the center, for

extracts, chromatographic fractions and purified compounds.47 Two distinct estrogen receptors in our bodies: ER-α and ER-β. While they both bind estrogen as well as other agonists and antagonists, the two receptors have distinctly different localizations and concentrations within our body. Structural differences

53 also exist between the two allowing for a wide range of diverse and complex processes to take place. The cell binding assay to assess the binding affinities of botanical extracts followed a standard pattern. The receptors are infused with

17β-estradiol, and a binding curve is obtained based on varying concentrations of

17β-estradiol present. Then, the extract in question is added in varying concentrations, acting as a competitive inhibitor, and its ability to bind is plotted against 17β-estradiol.

4.4.2 Serotonin (5-HT) Binding Activity

Either IC50 values or percent inhibition of receptor binding in transfected human CHO- cells for fractions and compounds were determined by personnel in

Project 2 of the Center.121 Serotonin activity was first assessed by the inhibition of radioligand binding to cell membrane preparations containing recombinant human serotonin receptor (5-HT) subtypes. The botanical extracts, fractions or purified compounds in question are assayed for competitive binding against

[3H]lysergic acid diethylamide (LSD) in human 5-HT7 receptor or [3H]8-hydroxy-

2-(di-N-propylamino)tetralin to the rat 5-HT1A receptor. Ligand binding data was then analyzed to determine if the extract, fraction or purified compound behaved as a competitive ligand of the serotonin receptor (5-HT) subtypes.

4.4.3 Other Biological Activities

AP induction, PS2 induction on Ishikawa cells and antioxidant activity

(DPPH) were provided by personnel in Project 2.47, 196

54 5.0 RESULTS

5.1 PLANT PROCUREMENT

Twenty-seven wild collections of C. racemosa roots and aerial parts were

made from N. Carolina to Pennsylvania. In addition, 20 collections of C.

americana and C. rubifolia were gathered in order to validate the chemical and

genetic authenticity of the C. racemosa collections. Collection records included

date of collection, genera, species, part, mass of collection (grams), collection

location and GPS coordinates, where indicated, are presented in table 6. Due to

overlap in distribution of C. racemosa and C. americana, morphological field

identification was essential for distinguishing collections. The major

morphological characteristic in the flower spike to distinguish between C.

racemosa and C. americana is that C. americana possesses three glabrous pistils vs. only one in C. racemosa; and the seeds are covered with broad, lacerate scales vs. no scales in C. racemosa.

Initial collections made in June of 1999 with assistance of the North

American botanical expert for the genus, G.W. Ramsey, Ph.D. Voucher specimens are housed at the Searle Herbarium, Field Museum of Natural History

(Chicago, IL) and the Ramsey-Freer Herbarium, Lynchburg College (Lynchburg,

VA).

55

Table 6. Botanical Center Database Summary of Black Cohosh and related species collections (PX=aerial parts, RR=roots).

Accession Date Genera Species Part Mass in Location number grams (GPS coordinates) BC001 6/28/1999 Cimicifuga racemosa RR 4827 Rockbridge County, Va. (37 48.27 N & 79 18.67 W)

BC002 6/28/1999 Cimicifuga americana RR 175 Rockbridge County, Va. BC003 6/29/1999 Cimicifuga rubifolia RR 177 Scott County, Va. BC004 6/29/1999 Cimicifuga rubifolia RR 280 Hancock County, Tn. BC005 6/29/1999 Cimicifuga racemosa RR 2106 Sevier County, Tn. (35 38.48 N & 83 28.47 W) BC006 6/30/1999 Cimicifuga americana RR 95 Sevier County, Tn. BC007 6/30/1999 Cimicifuga racemosa RR 800 Sevier County, Tn. BC008 6/30/1999 Cimicifuga americana RR 165 Swain County, NC. BC009 5/30/1999 Cimicifuga racemosa RR 10173 Somerset Co., PA BC010 8/26/1999 Cimicifuga racemosa RR 4359 Sevier County, Tn. BC011 8/26/1999 Cimicifuga americana RR 87 Sevier County, Tn. BC012 8/26/1999 Cimicifuga racemosa RR 829 Sevier County, Tn. BC013 8/26/1999 Cimicifuga americana RR 214 Swain County, NC. (35 35.30 N & 83 21.52 W)

BC014 8/26/1999 Cimicifuga racemosa RR 721 Swain County, NC. BC015 8/27/1999 Cimicifuga rubifolia RR 75 Scott County, VA, BC016 8/27/1999 Cimicifuga rubifolia RR 95 Scott County, VA, BC017 8/26/1999 Cimicifuga racemosa PX 498 Sevier County, Tn. BC018 8/26/1999 Cimicifuga racemosa PX 336 Sevier County, Tn. BC019 8/26/1999 Cimicifuga americana PX 167 Sevier County, Tn. BC020 8/26/1999 Cimicifuga americana PX 52 Swain County, NC. BC021 8/27/1999 Cimicifuga rubifolia PX 71 Scott County, VA, BC022 8/27/1999 Cimicifuga rubifolia PX 86 Scott County, VA, BC023 6/29/1999 Cimicifuga rubifolia PX 97 Scott County, Va. BC024 6/29/1999 Cimicifuga rubifolia PX 171 Hancock County, Tn.

56 Table 6 (continued). Botanical Center Database Summary of Black Cohosh and related species collections

Accession Date Genera Species Part Mass in Location number grams

BC025 6/29/1999 Cimicifuga racemosa PX 259 Rockbridge County, Va.

BC026 6/29/1999 Cimicifuga racemosa PX 351 Sevier County, Tn.

BC027 6/29/1999 Cimicifuga racemosa PX 111 Sevier County, Tn.

BC028 6/28/1999 Cimicifuga americana PX 88 Rockbridge County, Va.

BC029 6/30/1999 Cimicifuga americana PX 160 Sevier County, Tn.

BC030 6/30/1999 Cimicifuga americana PX 150 Swain County, NC.

BC031 6/22/1999 Cimicifuga racemosa RR 1471 Butler Co., PA

BC032 6/22/1999 Cimicifuga racemosa PX 650 Butler Co., PA

BC034 10/16/1999 Cimicifuga racemosa RR 295 Westmorland Co., Cook Townshi p, PA.

BC035 10/16/1999 Cimicifuga americana RR 844 Westmorland Co., Cook Townshi p, PA.

BC036 10/16/1999 Cimicifuga racemosa RR 710 Elk township, PA.

BC037 10/16/1999 Cimicifuga racemosa RR 582 Somerset, Co.,Summit Townshi p PA.

BC038 10/18/1999 Cimicifuga racemosa RR 2098 Butler Co., Little Buffalo Townshi p, PA.

BC043 5/30/1999 Cimicifuga racemosa PX 791 Somerset, Co.,PA

BC072 5/7/2000 Cimicifuga racemosa RR 5711 Somerset Co., PA

BC089 11/7/2000 Cimicifuga racemosa RR 452 Bedford Co. ,PA

BC090 11/7/2000 Cimicifuga racemosa RR 235 Bedford Co. ,PA

BC091 11/7/2000 Cimicifuga americana RR 32 Forbes State Forest, PA. BC092 11/7/2000 Cimicifuga racemosa RR 320 Rockbridge County, Va.

BC093 11/7/2000 Cimicifuga americana RR 37 Rockbridge County, Va.

BC094 11/7/2000 Cimicifuga racemosa RR 23510 Forbes State Forest, PA. BC095 11/7/2000 Cimicifuga racemosa RR 123 Sevier Co. TN, Smokey

BC102 4/16/2001 Cimicifuga racemosa RR 170000 Somerset Co, PA

57 In addition, C. americana flowers from August until October in the southern

states, roughly one month after C. racemosa (June-September).56, 57 With climate changes flowering characteristics are not always readily available, thus non- flowering characteristics must be used, the most distinguishing of which is the scar present on the stem at the point of branching on C. americana.56, 57 The roots

of C. americana also appear to be yellow and relatively smaller when compared

with the larger, whiter roots of C. racemosa. Roots were separated from aerial

parts on collection. Asian Cimicifuga species, C. acerina (BC164) (Sieb. Et

Zucc.) Tanaka, C. simplex (BC165) (DC.) Wormsk. ex Tursk., C. heraclefolia

(BC167) Kom., and C. dahurica (BC166) (Turcz) Maxim, were provided through

Dr. Norman R. Farnsworth, who secured the material through Toyama Medical

and Pharmaceutical University (Toyama, Japan) where voucher specimens are

maintained.

5.2 RAPD-PCR ANALYSIS

Initial studies to verify the species C. racemosa in comparison with the

other Eastern North American species (C. rubifolia and C. americana) have been

previously reported by the author and collaborators.197 This work is shown in

figures 6 A & B. Additional RAPD-PCR work was used to differentiate the Asian

Cimicifuga species (usually C. simplex), believed to be substituted for

C.racemosa in commercial products for economic reasons, as well as

C.americana, commonly named, yellow or white cohosh, which grows alongside

C. racemosa in wild populations. 192,56, 57 This work was undertaken, based on

58 information from members of the American Herbal Pharmacopoeia, believed

commercial Asian Cimicifuga material was being mislabeled and sold to

manufacturers as C. racemosa.198, 199 With the use of different primers, we were

able to distinguish the C. racemosa clinical extract starting-material provided by

our commercial collaborator, Pure World Botanicals, from the Asian Cimicifuga

species and C. americana. The PCR profiles are pictured in figures 7 A, B & C.

We conclude from figure 7 that the commercial root sample (BC163) was not

adulterated with any of the Asian species C.simplex, C. dahurica, and C. acerina.

The commercial sample, BC163, is consistent with C. racemosa. Possibly, the

commercial sample (BC163) may represent a subspecies or another subtype of

C. racemosa. This may explain the slight PCR band differences between the

authenticated material collected and used in this study by the UIC/NIH Center, in

contrast to the commercial material provided by our collaborators. Although there

is no report in the literature, these data match the observations made.

It was difficult to reproduce results using RAPD-PCR on root/rhizome

material. Extraction was performed using Qiagen’s plant DNA extraction kit,

which only provided DNA from Pure World’s C. racemosa (BC163) C. racemosa

(BC001), and C. americana leaf (BC030). A small quantity of DNA was available

from C. acerina (BC164), C. simplex (BC 165) and C.dahurica (BC166). The

DNA quality from these samples proved insufficient to produce reliable PCR

profiles. As a net result, it was extremely difficult to visualize high-resolution

profiles of poor quality DNA on an electrophoresis gel. The RAPD profiles of the commercial sample (BC 163) are consistent with those of C.racemosa (BC001).

59 DNA extraction is the limiting step. Unfortunately, only root material of the commercial sample (BC163) was available. The problems we observed in obtaining quality DNA from stored, hard pieces of root material match the experiences and reports of other groups.200

These data indicated that none of the Asian species of Cimicifuga that were tested, could be found in the C. racemosa material, which was the purpose of the investigation from the outset. Only primers OPA-05, OPC-15 and OPA-19 were used and therefore a more thorough analysis is needed using several more primers to ensure that these primers are not amplifying regions of the plant that are identical to all species.

60 Figure 6. RAPD-PCR of N. A American Cimicifuga species

6A. RAPD-PCR profiles of Cimicifuga species amplified by OPA-10 primer. Lanes 1- 6, C. racemosa; Lanes 7-14, C. americana; lanes 15-20, C. rubifolia. M-100 bpdna ladder

6B. RAPD-PCR profiles of Cimicifuga species amplified by OPA-15 primer. Lanes 1- 3, C. racemosa; Lanes 4-6, C. americana; lane 7 commercial C. racemosa & lane 8 C. rubifolia, M-100 bp DNA ladder, x- unknown

M 1 2 3 x M 4 5 6 M 7 8 B

61 100bp ladder - + + 12345 6 A Figure 7. PCR profiles utilizing different primers to distinguish C. racemosa from potential adulterants. The following samples were assayed: commercially supplied roots (BC163), authenticated C. racemosa roots (BC001), authenticated C. simplex roots (BC 165), authenticated C. dahurica roots (BC 166), authenticated C. acerina roots (BC 164), authenticated C. americana leaf (BC030).

7A. RAPD-PCR profiles of Cimicifuga species amplified by OPA-05 primer. M + - + 121 2233 M Lane 1- C. acerina, 2-C.heraclefolia, 3- C. dahurica, 4- C. simplex, 5- C. B racemosa, 6- Pure world C. racemosa material, +-positive control dna (S. cerevisae and ), - -negative control

7B. RAPD-PCR profiles of Cimicifuga species amplified by OPC-19 primer. Lane 1- C. americana, 2- C.racemosa, 3- Commercial C.racemosa material+- positive control DNA (S. cerevisae and ), - -negative control

7C. RAPD-PCR profiles of Cimicifuga species amplified by OPC-15 primer. M 12 333 4 5 - + M Lane 1-C. dahurica, 2-C. simplex, 3- C. americana, 4-C. racemosa, 5- C Commercial C. racemosa material +- positive control DNA (S. cerevisae), - - negative control

62

5.3 MICROSCOPY

5.3.1 Light microscopy

The following characteristics were observed in light microscopy:

parenchyma, trachea in secondary xylem, broad-wedge shaped medullary rays,

wood fibers, tabular cells, simple and compound, starch grains.58 These are

shown in figure 8 below. No distinct microscopic features of either coarsely-

powdered, finely-milled, or cross sectional slices of eastern North American

Cimicifuga species could be determined to distinguish one from another.

phloem

cortex a

epidermis a

medullary rays xylem

endodermis

b

b

Figure 8. Freehand microscopic drawings a) Cross sectional drawing of root/rhizome (BC001); b) root/rhizome (BC001) vessels and fibers.

63 5.3.2 Scanning-electron microscopy (SEM)

These aforementioned cell structural characteristics were also identified by scanning electron microscope (SEM). The advantage of SEM was in the magnification strength, up to 10000 times, to observe the cortex and starch granules in the coarse-ground samples (BC010). From the finely-milled powder phloem fibers were observed. SEM pictures representing these structures are presented in figure 9. Scanning electron microscopy is a central tool for identification of ground plant material, where no voucher specimen is available. It was important in this study to identify some of the morphological characteristics of Cimicifuga spp. at the cellular level.

64 A B

SG

C D

E F PF

G H RE

Figure 9. Scanning electron microscope pictures in decreasing magnification (5A-5C and 5F-5H are coarsely ground plant material (BC010), 5D & 5E are finely ground material (BC001)). Observed characteristics include phloem fibers (PF), starch granules (SG), root/rhizome exodermis (RE)

65 5.4 CHROMATOGRAPHY

5.4.1 Solvent extraction and partitioning to yield fractions for triterpene/standard isolation

The initial fractionation of C. racemosa was necessary for the development of less polar fractions and the subsequent isolation of triterpene glycosides to be used as marker compounds for the clinical extract. The dried, milled roots of C. racemosa (8 kg, BC001) were exhaustively extracted with

MeOH. The MeOH extract was concentrated in vacuo to yield 1250 g of a syrup residue. A sample (625 g) of the residue was suspended in water-MeOH (9:1,

1500 mL) and fractionated by successive partitions with EtOAc (2000 mL x 3) followed by n-BuOH (2000 mL x 5, sodium sulfate was added to the separatory funnel to enhance separation) to produce EtOAc-soluble (262 g) and n-BuOH- soluble fractions (100 g). This fractionation is detailed in figure 10 (page 67).

While the directive of this fractionation was to yield a large, relatively non-polar,

EtOAc-soluble fraction, to yield as many triterpene constituents as possible to properly identify and characterize a clinical black cohosh extract all resultant fractions (EtOAc, BuOH and H2O-soluble) were bioassayed. The 5-HT7 bioassay results indicated that the BuOH-soluble fraction produced from this separation demonstrated the greatest activity of all of the extracts and fractions generated by the Center at that time.201

66 C. racemosa roots (BC001) ca. 8.0 kg, MeOH ext. (1250 g) IC50 14.0 µg/mL, conc. in vacuo, re-suspended H2O/MeOH (9:1)

EtOAc

EtOAc (262 g) Mother Liquor IC50 21.0 µg/mL

BuOH Aqueous IC50 4.78 µg/mL 14% inhibition

Figure 10. A) Solvent extraction and partitioning of plant material to yield polar fractions for isolation from the butanol soluble fraction. 5HT7 bioassay data reported with the respective fractions. B) Fractionation of black cohosh roots for isolation of non-polar constituents from the EtOAc-soluble fraction. 5-HT7

5.4.2 Solvent extraction and partitioning of plant material to yield polar fractions

The previously published activity of the BuOH-soluble extract, led to the selection of the BuOH-soluble extract as the best candidate for further separation and isolation efforts.121 In order to increase the yield of the BuOH-soluble fraction

CHCl3:IPR (3:2) was used to initially partition the MeOH soluble extract.

The dried, milled roots of C. racemosa (3.75 kg, BC010) were further milled to a fine powder by a tissue homogenizer (ultra-turrax) and extracted with

MeOH (10 x 8,000 mL). The crude organic extract was concentrated in vacuo (<

40˚ C), to yield ca. 1.1 kg of a syrupy residue, the residue was reconstituted in d.d.i. H2O (1:9, 4 x 2000 mL) and subsequently partitioned with CHCl3:IPR (3:2, 4 x 2000 mL) and BuOH (4 x 2000 mL), to give >500 g and 150 g of CHCl3-soluble

67 and BuOH-soluble extracts, respectively. The fractionation scheme with bioassay

results is detailed in figure 11. The bioassay data that was generated for these

C.racemosa fractions showed decreased activity of the resultant BuOH-soluble fraction.

C. racemosa roots (BC010) ca. 3.75 kg, MeOH ext. (900 g) 39% inhibition, conc. in vacuo, re-suspended H2O/MeOH (9:1)

CHCl3/IPR

CHCl3/IPR Mother Liquor 44% inhibition

BuOH (150 g) from 40+ L Aqueous 35% inhibition 0% inhibition

Figure 11. A) Solvent extraction and partitioning of plant material to yield polar fractions for isolation from the butanol soluble fraction. 5HT7 bioassay data reported with the respective fractions bioassay data reported with the respective fractions.

68 5.4.3 Amberlite XAD-2, Second Level of Separation

For the first chromatographic separation of the n-BuOH soluble fraction,

150 g of the butanol-soluble extract from BC010 was loaded to 1.735 kg of

Amberlite XAD-2 (Sigma) Column. Column specifications are as follows: i.d. 50 mm, length: 1200mm, total volume: 2400 mL, density of XAD-2: 0.736 g/cm3 .

The column was washed with acetone until no residue was present upon evaporation, then it was subsequently washed with MeOH and water at a flow rate of 5 ml/min regulated by the MPLC Isco (Lincoln, NE) unit. The syrupy butanol-soluble extract (138 g) was suspended and sonicated in 600 mL of d.d.i. water and added to the top of the column. The columns was resealed at both ends, inverted and eluted with water, water with increasing percentages of

MeOH, 100% MeOH, MeOH with increasing acetone, and 100% acetone. This yielded 10 unique fractions, F01-F10 as determined by TLC and HPLC-ELSD.

The results are summarized in figures 12 and 13 (pages 70, 71). Bioassay data for these fractions indicated that the best candidates for further fractionation were

F05-F07.

69 step a 01-75-15 (150 gms .)

01-90-15 % 5HT 7 binding @ 100 µg/ml F-01 0 01-90-16 F-02 0

01-90-17 F-03 15.7

01-90-18 F-04 32.4

01-90-20 F-05 83.2

01-90-21 F-06 79.3

01-90-22 F-07 90.4

01-90-23 F-08 72.3

01-90-24 F-09 49.8

01-90-25 F-10 64.4

Figure 12. Summary of XAD-2 fractionation and bioassay results. Step a is the elution of the column with water, water with increasing percentages of MeOH, 100% MeOH, MeOH with increasing acetone, and 100% acetone. 01-75-15 is the butanol-soluble fraction from Figure 9. B on the TLC refers to the same entity. IFA and FKA represent isoferulic acid and fukinolic acid standards, respectively.

70 BuOH Fraction

F-01

F-02

F-03

F-04

F-05

F-06

mV F-07

F-08

F-09

F-10

Cimicifugic acid A

Fukinolic acid

Isoferulic acid

0.00 5.00 10.00 15.00 20.00 25.00 30.00 35.00 40.00 45.00 50.00 55.00 60.00 65.00 Minutes Figure 13. HPLC-ELSD chromatograms of the resultant XAD-2 fractions (F-01 through F-10), the fractions are overlayed with cimicifugic acid, fukinolic acid and isoferulic acid standards to show the relative polarity of the fractions, the capacity of XAD-2 to separate the polar butanol-soluble fraction, and served as a level of ad-hoc dereplication. Fraction concentrations ca. 10 mg/mL ran by method B. Injection volume was 10 µL.

71 5.4.4 MCI gel CHP20P

For the third level of separation of the n-BuOH soluble fraction (from

BC010), MCI gel® CHP20P was selected. The initial study to test the feasibility of this resin was on 4.3 grams of F-08 dissolved in 40 mL of 20% aq. MeOH into

100 g of a CHP20P MPLC column at a flow rate of 1 ml/min. The summary of the pilot column is depicted in figure 14. Column specifications for the pilot column are as follows: i.d. 20 mm, length: 500 mm, total volume: r2h= 157079.5 mm3 or

157.1 cm3, density of CHP 20P: 0.636 g/cm3. The sample was eluted with 30%

MeOH, at a 10% gradient until it reached 100% MeOH, then eluted with 100%

EtOH and 100% acetone, subsequently.

For the scaled-up column, the resin was initially washed with acetone followed by EtOH, MeOH, and d.d.i. water. The wash flow-through was evaporated in vacuo until no CHP20P residue eluted from the column. After the column wash was completed. Fractions F-05 to F-07 (8.6 g) were resuspended in

5% MeOH and loaded onto 400 gm of CHP20P. The extract was eluted with water, water with increasing percentages of MeOH, 100% MeOH, MeOH with increasing acetone, and 100% acetone. This yielded 35 unique fractions (G01-

G35) as determined by TLC fraction control and HPLC-ELSD. These results are summarized in figures 15 and 16. The HPLC-ELSD results of the resultant G- fractions, show that the use of CHP20P, the separation followed a ‘predictable’ pattern. The earlier fractions possessed primarily more polar, unknown constituents, gradually the pattern shifted to predominately the known phenolic acid esters (i.e. fukinolic acid). Because of the large number of fractions, it would

72 have been a poor use of resources to obtain bioassay data on all fractions.

Chemical screening of resultant fractions was used to better characterize the fractions. The use of LC/MS provided a targeted screening strategy that indicated the possible presence of compounds possessing an odd number of nitrogens in the resultant fractions, these fractions were then selected to pursue for further isolation work. Additional LC-MS chromatograms of fractions possessing compounds with an odd number of nitrogens are depicted in figure 17 and the appendix (section 10.2).

73 step a 01 - 75 - 15 (150 gms .)

01 - 90 - 23 F - 08 (4.3 g)

21-23 02- 21 - 23 F - 11 (1.25 g)

02 - 21 - 24 21-24 F - 12 (1.08 g)

02 - 21 - 25 21-25 F - 13 (0.63 g)

21-26 02 - 21 - 26 F - 14 (0.04 g) 21-27 02 - 21 - 27 F - 15 (0.26 g) 21-28 02 - 21 - 28 F - 16 (0.24g) 21-29 02 - 21 - 29 F - 17 ( 0.05 g)

02 - 21 - 30/31 0.00 10.00 20.00 30.00 40.00 50.00 60.00 Minutes F - 18 (0.02 g)

90-2321-23 21-24 21-26 21-25 21-27 21-28 21-29 21-30 21-31 90-23

Figure 14. Pilot column CHP20P separation summary of F-08 (72.3 % inhibition of 5HT-7 at 100ug/ml). The HPLC and TLC results of the resultant pilot column CHP20P fractions show the proficiency of the resin to provide a means for separating a complex polar fraction. These results led to the selection of CHP20P as the stationary phase for further large scale separation.

Step a-2.0 kg XAD-2 eluted with water, aq. MeOH, MeOH, Acetone Step b-100 gm CHP20P eluted w/ water, aq. MeOH, MeOH, EtOH, acetone HPLC-Method B TLC-Anisaldehyde, SSC 7 74

3 distinct fractions (F-05 through F-07), over 10 distinct spots (Anisaldehyde SSC8) 1 2 3 4 5 6 7 8 9 10

ca. 35 distinct fractions (g-1 through g-36), over 100 distinct spots (UV-254, SSC8)

Figure 15. Large Scale Separation CHP20P separation of 01-90-20 ,-21 and -22 (F-05 through –07, 8.6 g) by 400 gm CHP20P eluted w/ water, aq. MeOH, MeOH, EtOH, Acetone. The ‘active’ XAD-2 fractions were further separated by CHP20P. The complexity of the resultant fractions, on the 3rd level of separation can be seen by the large number of distinct spots on the UV(254) developed TLC plate.

75 2

3

4

5 mV 7

9

10

11

0.00 5.00 10.00 15.00 20.00 25.00 30.00 35.00 40.00 45.00 50.00 55.00 60.00 65.00 Minutes Figure 16. HPLC-ELSD results of CHP20P separation. Fractions G-2, 3, 4, 5, 7, 9, 10, 11 by method B. The chromatograms show the complexity of the third level fractions, and the potential for isolation of new constituents from C. racemosa. Fraction concentration ca. 5 mg/mL. The separation followed a ‘predictable’ pattern. The earlier fractions possessed primarily more polar, unknown constituents, gradually the pattern shifted to predominately the known phenolic acid esters (i.e. fukinolic acid).

76 12

13

14

15

mV 16

17

18

19

0.00 5.00 10.00 15.00 20.00 25.00 30.00 35.00 40.00 45.00 50.00 55.00 60.00 65.00 Minutes Figure 16 (continued). HPLC-ELSD results of CHP20P separation. Fractions G-12 through - 19 by method B. The chromatograms show the complexity of the third level fractions, and the potential for isolation of new constituents from C.racemosa. Fraction concentration ca. 5 mg/mL. The separation followed a ‘predictable’ pattern. The earlier fractions possessed primarily more polar, unknown constituents, gradually the pattern shifted to predominately the known phenolic acid esters (i.e. fukinolic acid).

77 21

22

23

24

mV 25

26

27

28

0.00 5.00 10.00 15.00 20.00 25.00 30.00 35.00 40.00 45.00 50.00 55.00 60.00 65.00 Minutes Figure 16 (continued). HPLC-ELSD results of CHP20P separation. Fractions G-21 through -28 by method B. The chromatograms show the complexity of the third level fractions, and the potential for isolation of new constituents from C. racemosa. Fraction concentration ca. 5 mg/mL. The separation followed a ‘predictable’ pattern. The earlier fractions possessed primarily more polar, unknown constituents, gradually the pattern shifted to predominately the known phenolic acid esters (i.e. fukinolic acid).

78

29

30

31

32

mV 33

34

35

36

0.00 5.00 10.00 15.00 20.00 25.00 30.00 35.00 40.00 45.00 50.00 55.00 60.00 65.00 Minutes Figure 16(continued). HPLC-ELSD results of CHP20P separation. Fractions G 29-36 by method B. The chromatograms show the complexity of the third level fractions, and the potential for isolation of new constituents from C. racemosa. Fraction concentration ca. 5 mg/mL. The separation followed a ‘predictable’ pattern. The earlier fractions possessed primarily more polar, unknown constituents, gradually the pattern shifted to predominately the known phenolic acid esters (i.e. fukinolic acid).

79 Sample g7 on amide column CRJULY150405A TOF MS ES+ 9.87 166 100 166.1 1.45e3

% 16.12 130.1 2.90 229.1 0 CRJULY150405A TOF MS ES+ 10.35 186 100 186.1 2.40e3

%

12.01 186.1 0 CRJULY150405A TOF MS ES+ 13.52 172 100 172.1 8.70e3 Relative abundance

%

0 CRJULY150405A TOF MS ES+ 13.52 TIC 100 172.1 1.04e4

3.23 15.07 2.90 337.1 154.1 229.1 % 9.21 10.35 12.85 3.87 4.25 314.1 14.30 186.1 11.29 136.1 163.0167.15.19 6.22 7.82 330.2 16.99 344.2 19.30 254.1 210.1 246.2 20.67 132.1 316.2 316.2 0 Time 4.00 6.00 8.00 10.00 12.00 14.00 16.00 18.00 20.00 22.00 Minutes

Figure 17. LC-MS screening for N-bearing compounds possessing an odd number (n=1,3,5,…) of nitrogens. Random chemical screening by LC/MS indicated the possible presence of compounds bearing an odd-number of nitrogen atoms in fraction G-7, indicated by the even m/z in positive mode. With a large number (over 30) of 3rd level fractions bioassay-guided fractionation would be a poor use of resources, thus this technique proved useful to identify fractions for isolation of potentially new constituents, as a method of dereplication.

80 5.4.5 Sephadex LH-20

Gel permeation chromatography offered by Sephadex LH-20 was selected for the third level of separation in parallel with MCI gel CHP20P. Gel permeation chromatography is a particular type of liquid/liquid chromatography for the separation of substances according to their differing molecular sizes. The column was washed with MeOH and H2O. Sample was dissolved in aq. MeOH and added to the gravimetric column through a 0.45 micron paper filter. Column specifications for the sephadex LH-20 column are as follows: i.d. 20 mm, length:

1000 mm. F-12 (0.5 g) was loaded onto 50 gm of Sephadex LH-20 and eluted with 100% MeOH. Five combined fractions were observed; SF-1 (0.05 g), SF-2

(0.15 g), SF-3 (0.11 g), SF-4 (0.07 g), and SF-5(0.02 g). Results and HPLC-

ELSD of resultant fractions are summarized in figure 18.

81 F-12 (0.5 g)

SF-1

SF-2

SF-3 SF1 SF2 SF3 SF4 SF5 SF-4 SF-5

Cimicifugic acid A

Fukinolic acid Isoferulic acid

mV SF-1

SF-2

SF-3

SF-4

SF-5

0.0 5.0 10.0 15.0 20.0 25.0 30.0 35.0 40.0 45.0 50.0 55.0 60.0 65.0 Minute Figure 18. Fractionation and TLC of Separation of F-12 (02-21-24) by 50 g of Sephadex LH-20 eluted with MeOH HPLC-ELSD of Sephadex LH- 20 separation of F-12 (02-21-04) by method B compared with standards of cimicifugic acid, fukinolic acid and isoferulic acid. Fraction concentration ca. 5 mg/mL.

82

5.4.6 Silica Gel Separation

For the initial separation of the EtOAc soluble fractions to generate

triterpene standards (figure 10, page 67) silica gel column chromatography (SiO2,

2.0 kg) was used. Column specifications are as follows: .i.d. 60 mm, length: 1000

mm, total volume: 2800 mL, density of silica gel: 0.708 g/cm3. A portion of the

EtOAc-soluble fraction (262 g) was eluted with CHCl3, CHCl3-MeOH (10%),

CHCl3-MeOH (20%), CHCl3-MeOH (30%), CHCl3-MeOH (40%), CHCl3-MeOH

(50%), CHCl3-MeOH (75%), and MeOH (100%) to yield eight combined fractions:

S-1 (oil), S-2 (3.0 g), S-3 (1.2 g), S-4 (2.3 g), S-5 (23 g), S-6, S-7 (60 g), S-8 (120 g) as depicted in figure 22 as previously cited.137 Column cuts were taken every

500 mL until the resultant eluent was colorless. The approximate volume eluted was 35 L. These eight fractions were used to produce thirty (30) compounds by project 1 of the Center.137, 138, 142 Further separation of fraction S-4 (2.3 g) afforded; glyceryl-1-palmitate (5 mg, 0.0013% of dried plant material); daucosterol-6'- linoleate (400 mg, 0.05%). In addition to the novel isolates 4',23-O-

diacetylshengmanol-3-O-α-L-arabinopyranoside, cimiracemoside M (6.1 mg,

0.0016%); 4'-O-acetyl-26-deoxyactein, cimiracemoside O (5.0 mg, 0.0013%);

4',23-O-diacetylshengmanol-3-O-β-D-xylopyranoside, cimiracemoside L (7.0

mg, 0.0018%): 2'-O-acetylactein (7.0 mg, 0.0022%), cimiracemate A (8 mg),

cimiracemate B (5 mg), cimiracemate C (2.1 mg) and cimiracemate D (1.8

mg). Fraction S-5 (23 g) was used to produce 23-O-acetylshengmanol-3-O-α-L- arabinoside (8.1 mg, 0.0021%), 23-O-acetylshengmanol-3-O-β-D-xyloside (7.3

83 mg, 0.0019%), cimigenol-3-O-β-D-xyloside (24 mg, 0.0063%), cimigenol-3-O- -L- arabinoside (7.1 mg, 0.0019%), cimigenol-3-O-β-D-xyloside (6 mg, 0.0016%), 26-

deoxycimicifugoside (3.0 mg 0.00079%), 25-anhydrocimigenol-3-O-β-D-xyloside

(30 mg, 0.0039%), 25-anhydrocimigenol-3-O-α-L-arabinoside (5.1 mg, 0.0013%),

25-O-acetylcimigenol-3-O-β-D-xyloside (40 mg, 0.01%), 25-O-acetylcimigenol-3-

O-α-L-arabinoside (30 mg, 0.0079%), cimicifugoside H-2 (8 mg, 0.0021%), cimicifugoside H-1 (13 mg, 0.0034%), 26-deoxyactein (210 mg) 40 mg of actein

(R,S) and the novel isolates 12-O-acetyl-25-anhydrocimigenol-3-O-α-L- arabinopyranoside, Cimiracemoside J (7.3 mg, 0.0019%), 12-O-acetyl-25-

anhydrocimigenol-3-O-β-D-xylopyranoside, Cimiracemoside K (9.4 mg,

0.0025%); 16-β:23;24:25-diepoxy-12-O-β-acetyl-3-hydroxy-9,19-cyclolanost-

23,26-olide-O-β-D-xylopyranoside, Cimiracemoside P (5.1 mg, 0.0013%); 23-

epi-26-deoxyactein (5.1 mg), 7-dehydro-23-epi-12,26-dideoxyacteol-3-O-β-D-

xylopyranoside, Cimiracemoside I (3.2 mg, 0.0008%), and 23-epi-acetylacteol-

3-O-α-L-arabinopyranoside, cimiracemoside N (30 mg, 0.0079%). The structural

data for the fourteen new constituents (in bold type above) from these fractions,

are available in the appendix (section 10.1).

84 5.5 ISOLATION OF CONSTITUENTS

5.5.1 Isolation of 1 (cimipronidine)

Compound 1 was isolated by semi-preparative RP-HPLC from fraction G-

15 resulting from the CHP20P fractionation (figure 13, page 71) of the combined

XAD-2 fractions F-5 through F-7 (figure 15, page 75) Thus, compound 1 was isolated from the fourth level of separation of the butanol-soluble fraction.

Isolation of 1 occurred following elution with 10% aqueous methanol. The analytical HPLC-ELSD separation of G15 (figure 16, page 77) indicated 5 major peaks. Eight peaks were collected as indicated in figures 19 and 20 (page x).

The PDA was used as a detector, the scan setting was from 190-410 nm. Six injections of 50 mg of fraction G-15 in 300 µL of MeOH were made. Column flow rate was 10 ml/min, HPLC stationary phase was 10% MeOH. Compound 1 was eluted isocratically. The six injections yielded a total of 25 mg of compound 1.

85

Wash out 1234 567 8 (mostly fukinolic acids) 350.00

300.00 nm

250.00

200.00

10.00 20.00 30.00 40.00 50.00 60.00 70.00 Minutes Figure 19. Semi-preparative RP-HPLC isolation of 1 (cimipronidine-P2) from fraction G-15 , PDA detector scan from (300 mg-6 runs), flow rate- 10.0 ml/min, isocratic 10% MeOH. Dead time of 2.0 minutes was used to reduce dead volume.

86 1

2

3 4 mV 5

6

7

8

0.00 5.00 10.00 15.00 20.00 25.00 30.00 35.00 40.00 45.00 50.00 55.00 60.00 65.00 Minutes Figure 20. Qualitative analytical HPLC-ELSD results of semi-preparative HPLC- ELSD of separation of fraction G-15, peaks 1 through 8. Chromatograms reveal additional peaks, than that of the initial HPLC-ELSD separation. Additional peaks were realized following preparative separation of fraction G-15 than were apparent from the initial HPLC-ELSD analysis (figure 16, page 77).

87 5.5.2 Isolation of 2 (fukinolic acid)

Compound 2 was isolated by semi-preparative RP-HPLC of fraction G-15 from the CHP20P fractionation (figure16, page 79) of the combined XAD-2 fractions F-5 through F-7 (figure 13, page 77). Thus, compound 2 was also isolated from the fourth level of separation of the butanol-soluble fraction from figure 11 (page 68). Isolation of 2 occurred when G-15 was eluted with 10% aqueous methanol. Fukinolic acid eluted approximately 25 min after cimipronidine from the same g-15 subfraction. Many polar compounds and plant extracts are routinely separated by RP-HPLC with small volumes of acid or base

(0.1-1.0%) added to improve retention time and peak shape of pH-sensitive analytes such as polyphenolics and alkaloids, respectively. in this instance, added TFA reduced the retention time of the highly polar material. Potentially complicating the effort further, complexation between nitrogenous constituents and highly conjugated phenolic acids have been previously reported.202, 203 The analytical HPLC-ELSD separation of G-15 (figure 14) indicated 5 major peaks.

Eight peaks were collected as indicated in figures 19 and 20 (pages 86, 87). A total of 65 mg of 2 were collected from the semi-preparative HPLC. Additional fukinolic acid was isolated in the production of fraction G-19 (Figure 16) from

CHP20P (31 mg).

5.5.3 Isolation of 3 (actein-(R/S))

Compound 3 was isolated by normal phase silica gel chromatography.

Compound 3 was eluted with CHCl3:MeOH (6:4), fraction S-6 from figure 22

88 (page 87). A total of 1.47 g of compound 3 was isolated from silica gel column chromatography, the 2nd level of fractionation of the EtOAc soluble fraction (figure

10, page 67). Actein was isolated as a mix of the 26-R and 26-S anomers

(stereoisomers). The yield of R and S was determined to be 20 and 80% respectively by HPLC-ELSD as shown in figure 21. This mixture was subjected to clean-up by semi-preparative RP-HPLC using a isocratic ACN:0.05% TFA in H2O

(65:35) mobile-phase, with a flow rate of 16.0 mL/min. A chiral column was not used. A flow splitter to utilize the ELSD for detection was graded at 10%. The

ELSD parameters were run according to method A. The additional preparative

HPLC step offered no further purification of the anomers due to mutarotation of the actein E ring in solution.

26-S

26-R

Figure 21. Isocratic analytical HPLC-ELSD of actein (26-R) and actein (26-S). Eluted with ACN:0.05% TFA in H2O (60:40) mobile-phase at 1.0 mL/min. flow rate. ELSD parameters according to method A.

89

5.5.4 Isolation of 4 (isoferulic acid)

Compound 4 was isolated by normal phase silica gel chromatography.

Compound 4 was eluted with CHCl3:MeOH (5:5), fraction S-7. A total of 1.34 g of compound 4 was isolated by silica gel column chromatography, the 2nd level of fractionation of the EtOAc soluble fraction shown in figure 10 (page 67).

90 5.6 CHARACTERIZATION OF ISOLATED COMPOUNDS

5.6.1 Characterization of 1 (cimipronidine)

The high-resolution exact mass measurement of compound 1, gave a protonated molecule of m/z 172.1014 indicating a molecular formula of

13 C7H14N3O2. Seven C-NMR resonances established the presence of a single carboxylic acid functionality (δC 179.826) and a single imine functionality (δC

154.573), the latter of which was confirmed by the MS/MS fragment ion of m/z

+ 13 130.0887 [MH-CN2H2] (figure 22). Both of the low field C NMR signals were shown to be quaternary carbon resonances (figure 23). One methine resonance and four methylene resonances were present in the APT 13C-NMR spectrum.

These assignments were confirmed by the gated 13C NMR experiment. (figure

24). Since there are no additional sites of unsaturation, the third double-bond equivalency requires a ring. HMBC data obtained for 1 failed to reveal all of the correlations necessary for full assignment of the structure. Contrary to expectations, no HMBC correlations could be observed between C-5 and the protons at H-2, H-3, and H-4. In addition, the expected correlations between C-2 and the protons H-3 and H-4b were undetectable as well (figure 25). Missing

HMBC correlations have previously been noted in the course of elucidation of guanidine structures.204 The absence of these correlations combined with broad, undefined peaks in the proton spectrum, the exceptions being the signals for H-

8a and H-8b, presented a difficult problem in terms of structure elucidation.

A probable explanation for the absence of cross peak correlations from the HMBC spectrum of 1 is the combination of the zwitterionic nature of the

91 2 compound 1 in conjunction with the fact that (i) typical JH,C couplings are generally small in magnitude and (ii) for the expected 3-bond correlations (C-5,

H-2) and (C-5, H-3) the dihedral angle between these protons and the C-5 carbon approach 90o and, therefore, the expected correlation are too weak or completely absent from the spectrum. Further complicating the situation is the chemical exchange phenomenon associated with the guanidine unit as well as the rapid fluctuations associated with the 5-membered ring, which are known to occur. Appropriate HMBC correlation cross peaks between the methylene protons at C-8 and carbons C-5 (two-bond) and C-4 (three-bond) were observed, while the expected HMBC correlations cross peaks used for defining the 5- membered ring were absent. These observations are consistent with the proposed structure. At this point the ACD database was used to generate potential structures that best fit the available data.205 Gradient HSQC data further confirmed the carbon and proton shift assignments of C/H-3 as well as C/H-4.

The C-4 protons appear as two broadened multiplets in the 1D-1H experiments

(D2O) at ~1.97 and ~2.20 ppm and are obscured by the two-proton multiplet around 2.15 ppm of the C-3 protons (figure 26). The 1H-1H-COSY data showed the following correlations, indicating abundant long-range coupling in the molecule and, thus, confirming a cyclic structure: H-2 (a,b) with both H-3 (a,b) and H-4 (a,b); H-3 (a,b) with H-2 (a,b), H-4 (a,b), and H-5; H-4 (a,b) with H-2

(a,b), H-3 (a,b), and H-5; H-5 with H-3 (a,b), H-4 (a,b), H-8a, and H-8b; and H-8a and H-8b with H-5 (figure 27).

92 The gradient NOESY results (D2O) provided further evidence consistent with structure 1. In addition to the presence of strong cross-peaks between the

C-2, C-3, C-4, and C-8 geminal protons, correlations consistent with vicinal relationships were observed between H-5 and both C-8 protons, as well as between H-5 and the C-4 protons. This is consistent with structural features inferred from the observed gHMBC data. In addition, there were strong cross- peaks between both of the C-2 protons and the 2H multiplet of the H-3 protons

(H ~2.15). Weaker cross-peaks were observed between one of the C-8 protons

(the higher field doublet-of-doublets at H 2.410) and both C-4 protons. H-5 exhibited a weak cross-peak in the gNOESY spectrum to the high-field C-2 proton multiplet (H 3.594). Its geminal partner, the low-field multiplet at H 3.467, showed a weak cross-peak to the low-field C-8 proton (doublet-of-doublets at H

2.692). A summary of the observed NOESY data obtained in D2O, except for the geminal NOE correlations, is shown in figure 28 and is consistent with proposed

1 structure 1. The H-NMR data for (1) in D2O permitted observation of all the protons except for overlapping C-3 protons and the low-field multiplet of one of the C-4 protons, which could be differentiated by the gHSQC, gNOESY, and gCOSY results. This served to confirm the relative stereochemistry of 1. The

1 absence of the guanidine protons in the H-NMR spectrum of 1 in D2O is clearly attributed to exchange of the guanidine protons with deuterium of the solvent.

Following the tentative assignment of structure 1, additional 1H-NMR experiments were performed on a dilute solution of 1 in DMSO- d6, in order to further confirm the presence of the guanidine moiety shown in figure 29. The spectrum showed

93 two (2H) broadened singlets centered at δH 9.291 and 7.828, respectively attributed to the guanidine zwitterions. A 1H- NMR spectrum of dilute 1 in DMSO- d6 with added trifluoroacetic acid (TFA), showed a broad singlet with all guanidine protons appearing at δH 13.578. Relevant NMR data for 1 are summarized in table 7. Off-white powder (H2O); [α]D: 36.21 (c 0.1), H2O; UV

-1 (H2O)λmax (log ε):285 (1.04), 333 (1.62) nm; IR (ITR-neat) νmax (cm ) 3365,

1631.

Compound 1 was given the trivial name cimipronidine({1-

[amino(imino)methyl]pyrrolidin-2-yl}acetic acid), unique to plants. The novel β- amino acid type structure, exhibiting zwitterionic behavior is not typical of the previously reported guanidine structures obtained from plants.206-208 Most of these guanidines have been shown to possess longer aliphatic side chains.206-208

94 100 172.1014 [M+H]+

CO H 2 CO2H N N N H H NH H2N 2 e

% 130.0933 [M-CN2H2] bundanc A e v i 70.0584 [Pyrolidine] lat e R

154.1106 [M-H2O] 94.0658 112.0831 113.0794 137.0804

0 m/z 70 90 110 m/z 130 150 170

Figure 22. High resolution positive-ion electrospray tandem mass spectrum of the protonated molecule of m/z 172.1014 (cimipronidine (1)) obtained utilizing a quadrupole time-of-flight mass spectrometer.

95 179. 154. 57. 41. 31. 22. 47 .

123 213 289 682 361 78 68 0 5 C - 5

ppm C O C N 2 H 3 H 3 C - 8 C - 2 C C - - 4 3

13 Figure 23. C-NMR APT of cimipronidine (1) in D2O (500 MHz). The low field 13C NMR signals were shown to be quaternary carbon resonances. One methine resonance and four methylene resonances were confirmed with a gated-carbon NMR experiment.

96 y it C-3 360 MHz). The ( O ity for each carbon. C-4 2 D ) in 1 he multiplic 97 C-8 rmation that was used to confirm the multiplic

C-2 C-NMR spectrum of ( 13

. Gated C-NMR experiment revealed t 13

C-5 gated of the carbon chemical shifts from the APT experiment. Figure 24 Important structural info

H H - - 3 4 a a

H H - - H H H H H 8 8 a b - - - - - 2 2 4 3 5

a b b b

0

C-3 C-4 C-8 C-2 50 C-5

100

150

ppm (t1

5.00 4.50 4. 00 3.50 3.00 2.50 2.00 ppm ppm (t2)

Figure 25. HMBC of cimipronidine (1) in D2O (360 MHz) no HMBC correlations could be observed between C-5 and the protons at H-2, H-3, and H-4. In addition, the expected correlations between C-2 and the protons H-3 and H-4b were also undetectable.

98

20.0 3

25.0

4 30.0

35.0

8 40.0

2 45.0

50.0

5 55.0

4.00 3.50 3.00 2.50 2.00

Figure 26. HSQC in D2O (400 Mhz). Gradient HSQC data confirmed the carbon shifts. Additionally, the HSQC experiment was used to confirm the chemical shifts of both H-4 protons and both H-3 protons, which appeared as a two broad singlets in the 1D-1H experiments with the peak at 1.75 ppm to be one proton by integration and the peak at 1.97 ppm to be 3 protons by integration .

99

1.50

2.00

2.50

3.00

3.50

4.00

4.00 3.50 3.00 2.50 2.00 1.50

1 1 Figure 27. H- H gCOSY spectra of (1) in D2O (400 MHz). The gCOSY shows the contiguity and connectivity of the ring. The H- H-COSY data showed the following correlations, indicating abundant long-range coupling in the molecule and, thus, confirming a cyclic structure: H-2 (a,b) with both H-3 (a,b) and H-4 (a,b); H-3 (a,b) with H-2 (a,b), H-4 (a,b), and H-5; H-4 (a,b) with H-2 (a,b), H-3 (a,b), and H-5; H-5 with H-3 (a,b), H-4 (a,b), H-8a, and H-8b; and H-8a and H- 8b with H-5.

100 H N H H Hb O O H 8 5 N N H 1 H 4 H H 2 Ha b 3 H

H a H NOE

Figure 28. The NOESY correlations of 1 (tmix = 1.5 seconds, D2O, 400 MHz) except for the observed geminal NOE contacts, which are left out to improve the clarity of presentation.

101 34 9 2 5 COOH 1 N 8

6 7 1 HN NH2

A

A

B

B

10 9 8 7 6 5 4 3 2 1 ppm

4a+4b 3a 8a 8b 5 3b 2a 2b

4.0 3.5 3.0 2.5 2.0 ppm

1 Figure 29. H-NMR spectra of 1 in d6-DMSO (A, B) and D2O (C) demonstrating the broad chemical shifts, the pH variability of NMR solvents and the presence of guanidine protons and the proton multiplicities of all resonances (D2O), respectively. The data also illustrate why D2O is the preferred NMR solvent for dereplication of 1 and its congeners.

102 1 13 Table 7. H and C NMR data of cimipronidine (1) (400/500 and 125/100 MHz, respectively, D2O and DMSO-d6)

a a 1 1 δC mult δH δH mult. [H] J [Hz] H- H COSY Position (D2O) [C] (D2O) (DMSO-d6) (D2O) (D2O) (D2O) 3.594 (2a) 3.36 (2a) ddd/mc 2.5 (3), 7.6 (3), 10.4 (2b) 2 47.31 CH 3a, 3b, 4a, 4b 2 3.467 (2b) 3.25 (2b) ddd/mc ~1 (3), 9.3 (3), 10.4 (2a) 2.17 (3a) 1.91 (3a) 3b 31.13 CH m 2a, 2b, 4a, 4b, 5 2 2.13 (3b) 1.87 (3b)

103 4b 2.20 (4a) 1.92 (4a) 22.61 CH m 2a, 2b, 3a, 3b, 5 2 1.97 (4b) 1.68 (4b) ~1 (4),~5.4 (8a), ~7 (4), 8.4 5 56.99 CH 4.331 4.163 dddd/mc (8b) 6 154.57 C – – – – 2.692 (8a) 2.272 (8a) dd 5.4 (5a), 15.1 (8b) 8 41.02 CH 5 2 2.410 (8b) 2.127 (8b) dd 8.4 (5b),15.1 (8a) 9 179.83 C – – – –

a H,C correlations established by HSQC experiment b The determination of δH from the 1D spectrum was hampered by the broadening of the resonances and by the overlap in the AB-type signal of protons H-3 and 4. However, the HSQC data gave correlations that permitted the determination of the chemical shifts of these protons. c Apparent ddd and dddd multiplicities, respectively, under first-order assumptions. However, additional long-range coupling and higher order effects give rise to a more complex signal multiplicity.

5.6.2 Characterization of 2 (fukinolic acid)

The proton-NMR resonance signals at δH 7.72 and 6.42 (each 1H, d, J

=16.1 Hz) indicated the presence of a trans-ethylene group conjugated with the

aromatic ring. Two groups of typical ABX spin system signals for 1,2,4-tri-

substituted aromatic ring protons were observed the first at δH 7.10 (1H,d J=2.3

Hz), 7.01 (1H, dd, J=2.3, 8.3 Hz), and 6.80 (1H,d J=8.3 Hz). The second at δH

6.74 (1H,d J=8.0 Hz), 6.64 (1H,d J=2.0 Hz), and 6.59 (1H,dd J=2.2, 8.2 Hz). One methylene signal at δH 3.08 (d, J=13.5 Hz) and the other δH 2.95 (d, J=13.5 Hz) were consistent with the expected proton signals for fukinolic acid (figure 30).

The 13C-NMR spectra showed 20 carbon atoms, signals ascribable to the trans-

ethylene group at δC 115.32 (C-2’’’) and 146.85 (C-3’’’), as well as two signal groups for 1,2,4 tri-substituted aromatic rings at δC 127.79 (C-1’’’), 115.32 (C-2’’’),

146.85 ( C-3’’’), 149.83 (C-4’’’), 116.55 (C-5’’’),123.25 (C-6’’’); with the other ring

signals δC 128.04 (C-1’ ), 118.83 (C-2’), 145.71 (C-3’), 145.30 (C-4’), 115.94 (C-

5’) and 123.05 (C-6’) depicted in figure 31. Additionally, the spectra also showed

one methylene carbon δC 42.22 (C-4), two carboxylic groups δC 174.78 (C-4),

170.90 (C-1) and one esterified group δC 168.29 (C-1’’). The NMR spectral data

compiled in table 8 are were consistent with the literature values.143, 159, 209

104 2''' 3'' HO 3''' 1''' 2''

1'' 6''' HO 4''' OO 5''' 2 1 HO CO2H 3 CO2H 5 2' 4 3' OH 1' 4' 6' OH 5'

H-4a/4b

7 6 5 4 3

H-5’’’ H-5’ H-6’’ H-2’’ H-2’’’ H-2’

H-6’’’

ppm 7.00 6.50

Figure 30. Compound 2 (fukinolic acid) 500 MHz 1H-NMR spectrum in MeOH- d4. Chemical shifts indicate the presence of two (2) tri-substituted aromatic rings.

105

150 100 50 ppm

5’ 2’’’

6’’’ 3’’ 6’ 5’’’ 1’ 3’ 2’ 2’’ 3’’’ 1’’’ 4’’’ 4’

4’’’

150. 140. 130. 120. ppm

Figure 31. Compound 2 (fukinolic acid) 500 MHz 13C-NMR spectrum in MeOH-d4. The data confirmed the presence of two tri-substituted aromatic rings as indicated in the 1H-NMR experiment.

106

Table 8. Spectroscopic 13C and 1H-NMR data of fukinolic acid (500 MHz, MeOD-d4)

Position signal δ H δ C m J (Hz)

1 C (COOH) - 170.90 - -

2 CH 5.65 78.28 s -

3 C - 80.44 d 15.89

4 CH2 2.95a, 42.22 d 13.5 3.08b d 13.5

5 C (COOH) - 174.78 - -

1’ C - 128.04 - -

2’ CH 6.74 118.83 d 2.2

3’ C - 145.71 - -

4’ C - 145.30 - - 5’ CH 6.64 115.94 d 8.2

6’ CH 6.59 123.05 dd 8.2, 2.2

1’’ C - 168.29 - -

2’’ CH 6.42 114.26 d 16.0

3’’ CH 7.72 148.22 d 16.0

1’’’ C - 127.79 - -

2’’’ CH 7.10 115.32 d 2.3

3’’’ C - 146.85 - -

4’’’ C - 149.83 - -

5’’’ CH 6.80 116.55 d 8.3

6’’’ CH 7.01 123.25 dd 8.3, 2.3

107 5.6.3 Characterization of 3 (actein (26-R and S))

The high-resolution exact mass measurement of compound 3, gave a m/z

703.3704 indicating a molecular formula of C37H58O11Na (figure 32). From the

1H-NMR spectrum, the cyclopropane methylene signals (H-19) of 3 were

observed at δH 0.21 and 0.55 (each 1H, d, J = 4.1 Hz). The spectrum also

showed seven methyl groups at δH 0.77 (H-28), 0.97 (H-21, J =6.45 Hz), 0.98 (H-

30), 1.31 (H-29), 1.35 (H-18), 1.71 (H-27), and 2.11 (-OAc), the anomeric proton

at 4.85 (H-1’, J = 7.6 Hz) and the distinctive hemi-acetal signal δH 5.74 (H-26,

split due to stereoisomeric-anomeric mixture, one R and one S), indicative of

spontaneous interconversion in solution, also known as mutarotation as is seen

in figure 33. The 13C and DEPT NMR spectra of 3 showed signals ascribable to

four oxygen-bearing methine carbons at 88.1 (C-3), 77.1 (C-12), 73.01 (C-16),

and 63.47 (C-24), two oxygen-bearing quaternary carbons at 105.85 (C-23) and

67.16 (C-25). With the field strength of the 500 MHZ instrument two hemi-acetal peaks were distinguishable at δC 98.45 (C-26 a) and (C-26 b) for the anomers actein (26-S) and (26-R), respecitvely as seen in figure 34. The spectra also

posessed five oxygenated carbons assignable to the xylopyranose moiety 107.5

(C-1’), 75.6 (C-2’), 78.7 (C-3’), 71.3 (C-4’), 67.1 (C-5’) and an acetyl group

(170.6, 20.7 for -OAc). The NMR spectral data compiled in table 9 matches the

literature reference values.137

108 Relative Abundance

time-of-flight massspectrometer. protonated moleculeof Figure 32. Highresolutionpositive-ionelec m/ z 685.3549(actein(

109 m/z

tros pray tandemmassspectrumofthe 3 )) obtainedutilizingaquadrupole

H - 2 6

a

&

b B A O H H H - - - 2 2 H H A - 2 7 1 H c - - 1 3 8 - 2 8 0 A 9 H-1’-anomeric

B

Figure 33. Proton NMR spectrum of actein (3) in pyridine-d5 (500 MHz). The distinctive hemi-acetal signal H-26 is seen at the top. The spectrum also showed seven methyl groups (A), the anomeric proton at 4.85 (H-1’, J = 7.6 Hz) (B).

110

ppm 2 2 6 6 S R

ppm

13 Figure 34. C-NMR spectra of actein (3) in pyridine-d5 (500 MHz) instrument two hemi-acetal peaks were distinguishable at δC 98.45 (C-26 a) and (C-26 b) for the anomers actein (26-S) and (26-R).

111 1 13 Table 9. Anomeric actein (26-R and 26-S) H and C NMR data in Pyridine-d5 (500 MHz). The data represents the 26-R anomer as the more abundant constituent. However the signal at 26 indicates a doublet representing mutarotation position signal δ H δ C m J (Hz)

1 CH2 1.10, 1.54 31.92 m,m -

2 CH2 1.88, 2.19 30.21 m,m -

3 CH 3.44 88.10 dd 4.4, 11.2

4 C - 41.23 - - 5 CH 1.22 47.01 m -

6 CH2 0.77, 1.45 20.40 m,m -

7 CH2 0.92, 1.21 25.71 m,m -

8 CH 1.61 45.66 m -

9 C - 19.55 - -

10 C - 26.80 - -

11 CH2 1.20, 2.69 36.71 m, dd 9.1, 16.0

12 CH 5.09 77.10 dd 3.9, 8.6

13 C - 47.88 - -

14 C - 48.81 - -

15 CH2 1.50, 1.68 43.62 m,m -

16 CH 4.59 73.01 dd 7.4,14.7

17 CH 1.76 56.44 m -

18 CH3 1.35 13.52 s -

19 CH2 0.21, 0.55 29.50 d,d (4.3), (4.0) 20 CH 1.84 26.12 m -

21 CH3 0.97 21.65 d 6.45

22 CH2 1.65, 2.22 37.67 m -

23 C - 105.85 - -

24 CH 3.94 63.47 s -

25 C - 67.16 - -

26 CH 5.74 98.45 d 4.6

27 CH3 1.71 15.32 s -

28 CH3 0.77 19.51 s -

29 CH3 1.31 25.71 s -

30 CH3 0.97 15.32 s -

1’ CH 4.85 107.56 d 7.5

2’ CH 4.02 73.63 t

3’ CH 4.15 78.67 t 8.7

4’ CH 4.21 71.28 dd 5.25, 9.24

5’ CH2 3.73, 4.38 67.15 t, dd (10.2), (5.1, 11.2)

-OAc Ac 7.01 170.54, 21.02 dd 8.3, 2.3

112 5.6.4 Characterization of 4 (isoferulic acid)

13 The C NMR spectral data at δC 148.41(C-3) and 118.41 (C-2) was indicative of a trans-ethylene group. One signal group was ascribable as a 1,2,4 tri-substituted aromatic ring the conjugated quaternary signals at δC 150.98 (C-7),

148.84 (C-6), 129.11 (C-4), and the conjugated methine resonances at δC 121.49

(C-9), 115.67 (C-8), 112.42 (C-5). Additionally, the δC169.84 (C-1) was

ascribable to a carboxyl group, with the methoxy- group at δC 56.05 as depicted

in Figure 42. The proton resonances for the trans-ethylene group were observed

at δH 8.34 (H-3, J=15.9) and 7.12 (H-2, J=15.89). The conjugated ring proton

shifts were 7.89 (H-5, J=8.6), 7.42 (H-9, J=8.3), 7.18 (H-8, J=8.3) with the

methoxy- shift at 3.59. NMR data are compiled in table 10. 1H-and 13C-NMR data

are consistent with data in the literature.143

113

9 3 1 HO 4 CO2H 8 2

5 MeO 7 6 C- 8 C- C- 5 9 C- 3 C- O-M 2 e C- C- C- 4 7 6

ppm

Figure 35. Carbon NMR of (4) isoferulic acid in pyridine-d5 (500 MHz). The data revealed ten (10) carbons and one methoxy group.

114

O- Me H- 9 H- H 5 - 8 H- H- 3 2

ppm

Figure 36. Proton NMR of isoferulic acid (4) in pyridine-d5 (500 MHz). The ABX spin system signals for 1, 2, 4-trisubstituted aromatic ring protons were observed at δH 7.89, 7.42 and 7.18.

115 Table 10. Isoferulic acid 1H and 13C-NMR data in Pyridine-d5 (500 MHz)

Position signal δ H δ C m J (Hz)

1 C - 169.84 - -

2 CH 7.12 118.41 d 15.89

3 CH 8.34 148.84 d 15.89

4 C - 129.11 - -

5 CH 7.89 112.42 d 8.6

6 C - 148.84 - -

7 C - 150.98 - -

8 CH 7.18 115.67 d 8.3 9 CH 7.42 121.49 d 8.3

-OCH3 CH3 3.97 56.05 s -

116 5.6.5 Additional Characterization Studies

Review of the RP-HPLC on resultant fraction G-11 from the CHP20P

fractionation (figure 16, page 76) of the combined XAD-2 fractions F-5 through F-

7 (page indicated the possible presence of additional nitrogen bearing

compounds (figure 17, page 80). Additional high resolution LC-MS/MS analysis

by project 4 of the center indicated the presence of an additional guanidine

analog with the protonated molecule of m/z 186.1818 (figure 37), consistent with

the molecular composition of C8H16N3O2, the product ion tandem mass spectrum

of this analog contained a fragment ion of m/z 130 (figure 38) corresponding to

the loss of N-methyl carbondiimine from the guanidine group, strongly indicating that an additional methyl is present. Supporting LC-MS data of other potential nitrogenous constituents are shown in the appendicies (section 10.2). The compound was identified based on quasimolecular ions and MS-MS fragmentation pattern, at least four methylated and one dehydrated cimipronidine exist at the fourth level of separation of the butanol-soluble fraction from figure

10. The mass of fraction G-11 was 135 mg.

117 Sample g11 on amide column crjuly150403 TOF MS ES+ 9.71 100 268 268.2 6.29e3

%

6.11 137.1 0 crjuly150403 TOF MS ES+ 16.87 100 166 166.1 1.56e4

%

0 crjuly150403 TOF MS ES+ 14.16 100 172 172.1 1.73e4

% 16.65 172.2

0 crjuly150403 TOF MS ES+ 10.94 100 186 186.2 9.15e3 12.58 % 186.2 10.20 13.58 200.2 144.1 0 crjuly150403 TOF MS ES+ 14.16 16.85 TIC 100 166.1 3.06 172.1 2.79e4 5.90 163.1 3.89 12.93 15.67 137.1 9.71 10.94 15.19 163.1 5.35 12.16 144.1 154.1 % 268.2 186.2 180.1 334.2 6.83 8.56 297.2 298.2 19.09 21.68 27.30 28.17 169.2 196.1 290.2 342.3 316.2 0 Time 4.00 6.00 8.00 10.00 12.00 14.00 16.00 18.00 20.00 22.00 24.00 26.00 28.00

Figure 37. Positive ESI LC-MS of a 5-HT7 active fraction containing cimipronidine (1) and its analogues. Following from their quasimolecular ions and MS-MS fragmentation pattern, at least four methylated and one dehydrated cimipronidine (1) analogue are contained in the active fraction. Compounds with no obvious structural correlation are marked with * and molecular weights provided based on MH+ quasi molecular ion calculations.

118

OH CO2H Me N N N

N NH HN NH2 CH 2 70.1059 2 10 0 Me

84.1201

154.1461 186.1818 % 126.1369 e 112.1220 130.1339 71.1139 86.1004 113.1053 abundanc e

v i 95.1023 t 83.07 145.1429 168.1620 a l

e R 0

m/z

CO2H CO H 2 N N N 70.1059 100 H H

HN NH2 130.1246 Me

% e

130.0966 abundanc

e 186.1818 v i t 95.0904 a l 113.1053 168.1620 e 71.0864 R 86.1080 137.1125 155.1259 169.1535 0 m/z 70 90 110 130 150 170 190

Figure 38. Positive ion electrospray tandem mass spectral (MS-MS) data to identify N-methyl cimipronidine. The variability with respect to the structural data of a mesomeric guanidine constituent. While further study is needed to confirm the N-methyl cimipronidine structure, the proposed fragmentation above, based on the structural fragmentation data of (1), is supportive of the N-methyl structure.

119 5.7 Preparation of the Clinical Extracts

Methanol extracts of Black Cohosh (BC001) were initially evaluated for

estrogenic activity using a battery of biological assays by Project 2 of the Center

as previously reported.47 These data are available in table 11. Without a direct estrogenic binding effect to explain the clinical activity of Black Cohosh, a methanol extract of Black Cohosh was sent out to a contract lab (PanLabs) to be assayed for serotonin (5-HT) binding activity, where the extract demonstrated

121 significant activity in receptor subtypes 5-HT1A and 5-HT7. The compounds

isolated from Black Cohosh by Project 1 of the center,137, 138, 142 were then tested

for their serotonin binding activity. The activities of some triterpenes isolated by

Project 1 are detailed in table 12. The triterpenes did not demonstrate strong

activity (ca. 50 µg/mL) especially in comparison to some of the phenolic

constituents (i.e. cimicifugic acids A (IC50 0.52 µg/mL) and B (0.91 µg/mL)).

However, it was determined by center personnel that the compounds 26-

deoxyactein, 23-epi-26-deoxyactein, actein (26-R) and actein (26-S) would be

the best candidates for marker compounds based on their abundance in the

black cohosh in combination with their biological activity. These markers were

used to confirm the identity of Cimicifuga racemosa in the extract as well as

correlate the activity of the extract with the relative abundance of these

constituents. in A previous study to evaluate diurnal compound variability was

used to determine the relative abundance of these active/marker compounds in a

methanol extract of Black Cohosh (tables 13 and 14).210 Standard curves for the

clinical capsule analysis and additional variation data are available in figure 39.

120 Additionally no formononetin or kaempferol were detected in the extracts as previously reported.48, 210 Ethanolic (40%, 60% and 75 %) and a 40% isopropanolic extract were analyzed by HPLC for the % of ‘active’ marker compounds. These data, (table 15, figures 40 and 41) and the bioassay data for these extracts, in table 16, led to the selection of a 75% ethanolic extract for the clinical study formulation.

Table 11. Biological activity of a MeOH Black Cohosh extract (from BC001), ER binding, AP induction, PR and pS2 mRNA expression and cytotoxicity. (Adapted from Liu, et al., 2001). Black Cohosh demonstrated no direct estrogenic effect.

ER-α-binding IC50 >> 50 µg/mL

ER-β-binding IC50 >> 50 µg/mL

AP induction, Ishikawa cells IC50 >> 50 µg/mL PR expression, Ishikawa cells NA (Intensity ratio)

Toxicity, Ishikawa cells ED50 >> 20 µg/mL pS2 expression, S-30 cells NA (Intensity ratio)

Toxicity, S-30 cells ED50 >> 20 µg/mL

121 Table 12. Serotonin (5HT) binding activity data of compounds isolated from within the UIC/NIH Center. The italicized compounds were selected by the

Center as standards to correlate the chemical fingerprint with biological activity of the extract. Actein (26R/S), 23-epi-26-deoxyactein, and 26-deoxyactein were selected based on their activity and relative abundance to the other constituents which displayed similar or greater activity. The biological activity refers to the percent at which the extract inhibits [3H]-lysergic acid diethylamide (LSD) binding to the human 5HT-7 receptor. The polyphenolics cimicifugic acids A and B demonstrated the greatest activity in this assay. Despite the activity of these constituents, they are less abundant in a 75% ethanolic extract than the triterpenes. These compounds are abundant in the BuOH-soluble fraction.

Compound 5HT-7 % inhibition IC50 @ 100 µg/ml Cimicifugic acid A 0.53 µg/ml or 1.2 µM

Cimicifugic acid B 0.91 µg/ml or 1.2 µM

Fukinolic Acid 53

Cimifugate A 45

Actein (R,S)* 50

23-epi-26-deoxyactein* 31

26-deoxyactein* 55

25-anhydrocimigenol-3-O-ß-D- 55 xyloside

25-O-acetylcimigenol- 3-O-ß-D- 46 xyloside

2-O-Acetylactein 47

Cimiracemoside I 69

Cimiracemoside M 48

Cimiracemoside O 50

122 Table 13. Average standard constituent concentrations in µg/g of 14 Cimicifuga racemosa root/rhizome methanol extracts determined by HPLC- ELSD (injected at 10 mg/ml). This study was used to determine the relative concentration of the major constituents in Black Cohosh. These collections were made in different locations and at different periods throughout the growth season, to analyze the degree of variation within the botanical. From the data the trends observed were the variability at which the concentration of the major constituents appeared influenced by geographical and diurnal factors, evident from the standard deviations. In addition, to the relative order of average abundance of the standards: actein (26S) > 23-OAc-shengmanol- xyloside > cimiracemoside A >23-epi-26-deoxyactein> isoferulic acid > 26- deoxyactein > 26-deoxycimicifugoside > actein (26-R) > caffeic acid > cimicifugoside H-1 > ferulic acid.

Standard Average Concentration

Caffeic acid 133 ± 63 Ferulic acid 66 ± 98

Isoferulic acid 452 ± 277

Cimiracemoside A 927 ± 939

Cimicifugoside H-1 102 ± 165

Actein (26-R) 264 ± 174

26-deoxycimicifugoside 370 ± 368

Actein (26-S) 2153 ± 1088

23-epi-26-deoxyactein 878 ± 338

26-deoxyactein 379 ± 168

23-OAc-shengmanol-xyloside 1573 ± 558

123 Table 14. Average constituent levels in µg/g of 14 Cimicifuga racemosa root methanol extracts determined by HPLC-ELSD (injected at 10 mg/ml). A) 7 southern states collections (NC, VA and TN) B) 7 Northern Appalachian Collections (PA). These collections were made within their respective, southern and northern regions at different periods of the growth season. The analysis of the standard deviations demonstrates similar variability to the analysis in table 13, this trend indicated that variability in the growing season may have more influence on constituent concentration than geographic origin.

Average Concentration A Standard Caffeic acid 105 ± 79 Ferulic acid 46 ± 122 Isoferulic acid 528 ± 383 Cimiracemoside A 1355 ± 1208 Cimicifugoside H-1 184 ± 202 Actein (26R) 305 ± 226 26-deoxycimicifugoside 384 ±353 Actein (26S) 2393 ± 1174 23-epi-26-deoxyactein 778 ± 339 26-deoxyactein 418 ± 186 23-OAc shengmanol-xyloside 1682 ± 608

B Standard Average Concentration

Caffeic acid 161 ± 22 Ferulic acid 87 ± 70 Isoferulic acid 388 ± 144 Cimiracemoside A 499 ± 150 Cimicifugoside H-1 21 ± 55 Actein (26R) 223 ± 103 26-deoxycimicifugoside 357 ± 412 Actein (26S) 1913 ± 1026 23-epi-26-deoxyactein 978 ± 330 26-deoxyactein 339 ± 151 23-OAc shengmanol-xyl 1465 ± 526

124 Standard curve of 23-epi-26 deoxy 03-12-03

7 6.9 6.8

a 6.7 e r 6.6 Linear (Standard curve 23- epi - A

g 6.5 26)

Lo 6.4 6.3 6.2 y = 1.6978x + 1.3416 6.1 R2 = 0.9982 2.8 2.9 3 3.1 3.2 3.3 Log Concentration

Figure 39. Phase II clinical capsule analysis standard curve. Chemical standardization of the extract was dependent on traditional analytical experimental design to the application of new methodology. The four (4)-point standard curve was generated to determine the concentration of 23-epi-26-deoxyactein in the clinical capsules by calibration of the ELSD response. The four points represent the logarithm of four different duplicate injections by concentration in the the HPLC- ELSD system. The analysis was only considered valid if the r2 value was greater than 0.995. The concentration of the other standards in the capsule (actein (26R), actein (26S) and 26-deoxyactein) were determined by calculating their respective ELSD response as 23-epi-26-deoxyactein. The calibration of the signal was inferred to be 1:1 for this analysis.

125

Table 15. Percentage of center standards in extracts determined by

HPLC-ELSD analysis. All were produced from BC 001. Samples ran in triplicate. Standard deviations for this analysis were within ± 5% of the average constituent concentrations. From the data, to 40% and 75% EtOH extracts appeared to be the most logical choice for selection to formulate the Center clinical extract.

Compound IPR 60% EtOH 40% EtOH 75% EtOH Commercial Extract Actein (26R) 0.21 0.23 0.31 0.26 0.05

Actein (26S) 2.40 2.55 3.34 3.26 0.75

23-epi-26- 0.80 0.90 1.08 1.01 0.19 deoxyactein 26-deoxyactein 0.62 0.37 0.92 0.48 0.20

Total 4.03 4.05 5.65 5.01 1.19 Percentage

*-supplied by PureWorld Botanicals™, PE 2.5% extract

126 40% EtOH

60% EtOH

75% EtOH mV

40% IPR

MeOH

0.0 5.0 10.0 15.0 20.0 25.0 30.0 35.0 40.0 45.0 50.0 55.0 60.0 65.0 Minutes

Figure 40. HPLC-ELSD chromatographic overlay of Black Cohosh (BC001) extracts produced with different solvents. The overlay revealed the variability in extraction efficiency of the “marker” constituents of Black Cohosh in addition to the data provided in table 15. The solvent used in the preparation of the clinical extract was 75% EtOH, which also demonstrated slightly greater biological activity than the 40% isopropanol and 60% ethanol extracts. Biological activity refers to the percent at which the extract inhibits [3H]-lysergic acid diethylamide (LSD) binding to the human 5HT-7 receptor.

127 60% EtOH

40% IPR

40% EtOH

MeOH Relative Abundance

75% EtOH

BuOH

Minutes

Figure 41. Positive-ion electrospray LC-MS chromatographic overlay of Black Cohosh (BC001) extracts produced with different solvents. Observation of m/z 685 a molecular-ion of deoxyacteins, acteins and related, unidentified cimigenols demonstrated the variability in extraction efficiency of the “marker” constituents of Black Cohosh. The solvent used in the preparation of the clinical extract was 75% EtOH, which also demonstrated slightly greater biological activity than the 40% isopropanol and 60% ethanol extracts. Biological activity refers to the percent at which the extract inhibits [3H]-lysergic acid diethylamide (LSD) binding to the human 5HT-7 receptor

128

Table 16. Serotonin binding activity of crude Black Cohosh extracts (BC001). The biological activity refers to the percent at which the extract inhibits [3H]-lysergic acid diethylamide (LSD) binding to the human 5HT-7 receptor. The methanol extract displaced radioligands from the 5-HT1A (IC50 = 2.5 ± 0.6 g/mL) and 5-HT7 (IC50 = 2.2 ± 0.2 g/mL) receptors equally well. While the crude methanol extract exhibited activity greater than or equivalent to the other extracts, it was not a candidate for the clinical extract due to the potential for toxicity with residual solvent. This biological data in conjunction with the chemical fingerprinting studies was used to select the 75% ethanolic extract as the specification for clinical study.

Extract 5HT7 % inhibition IC50 µg/mL @ 100 µg/mL

Crude MeOH 90

40% Isopropanol 89 14

60% EtOH 17

75% EtOH 12

129 5.7.1 Collaborator Formulation for the Phase I Clinical Extract

The specifications were given to our corporate collaborators at Pure World and Pharmavite, where they formulated the extracts and capsules for clinical trial and analysis. Due to the unexpected binding/interference of maltodextrin in the serotonin bioassay, rice flour was used as a flow-through, filler material. The plant material (BC001) was extracted with 75% EtOH. The resultant extract was spray dried. The Information provided from Pure World and Pharmavite is as follows; additional documentation is available in the appendix:

Phase I extract Production Lot #01I-2810 CR-01-49-10

Pure world notebook # KH-13-164, 02584-208

UIC received 8 kg from Pure World on 02/04/02 and sent 4.7 kg to Pharmavite via FedEx on 02/11/02 for encapsulation.

5.7.2 Collaborator Formulation for the Phase II Clinical Extract

The plant material (9-1744) was extracted with 75% EtOH. The resultant extract was spray dried. Plant material from Pure World Lot # 9-1744. Again, rice flour was used as a flow-through/filler.

5.7.3 Phase I Clinical Capsule Analysis

The mass of capsule was 0.511 gm ± 0.012. The mass of contents was

0.429 gm ± 0.010. The calculated mass of standards per capsule was found to be; 23-epi-26-deoxyactein: 1.41 mg ± 0.33; actein-(26S): 0.73 mg ± 0.14; actein-

(26R): 0.12 mg ± 0.02. The percentage of standards per capsule extract was

130 (2.26 mg ± 0.49 triterpene glycosides per 32 mg extract): 7.04%. The combined averages are given with standard deviations of 5 capsules run in triplicate. Rice flour was filtered out with a 0.22 µm syringe filter. LC-MS studies as depicted in figure 42 have shown stability of the most abundant mass ions of the extract used for the clinical capsule from time zero through one year.

Phase 1 Extract Activity (estimated error limit ±10%)

Estrogenic Activity

α-receptor binding >50% @ 200 µg/mlβ-receptor binding >50% @

200 µg/ml

PS-2 induction- no up-regulation @ 20 µg/ml

Ishikawa PR induction-no up-regulation @ 20 µg/ml

Antiestrogenicity:

Ishikawa cell assay >50% @ 20 µg/mL

DNA Damage:

Comet assay* 7.7% @ 20 µg/ml

Antioxidant Activity:

DPPH (IC50-76 µg/ml)HL-60* 88% @ 200 µg/ml

Serotonergic Activity:

5-HT7 receptor binding IC50 13 µg/ml

Cytotoxicity: @ 20 µg/mlIshikawa Cells- no reduction

s30 Cells- no reduction

LC-MS Assays:

131 GSH adduct formation* (m/z-460.5)

*Negative in PUF estrogen assay

5.7.4 Phase II Clinical Capsule Analysis

HPLC-ELSD analysis of the phase II clinical capsules found the average

percentage of dry weight of capsule content and standard deviation of ‘active’

standards per two capsules (128 mg) to be.: 23-epi-26-deoxyactein 2.84% ±

0.08, 26-deoxyactein 0.72% ± 0.02, actein-(26S) 2.77% ± 0.07, Actein-(26R)

0.94% ± 0.02. This gave a total of 5.67% active standards per two clinical

capsules. The combined averages are listed with standard deviations of five (5)

capsules run in duplicate. Rice flour was filtered out with a 0.22 µm syringe filter.

Phase II extract activity (estimated error limit ±10%)

Serotonergic Activity:

5-HT7-receptor binding IC50 18.1 ± 6.45 µg/mL

Phase II capsule activity:

5-HT7-receptor binding @ 10 µg/mL 61.153% ± 9.25

5.7.5 Overview of Clinical Material

The Phase II clinical extract was botanically authenticated (by PCR, vouchers, microscopy), chemically standardized to 5.6 % active triterpene glycosides and biologically standardized to competitively inhibit the 5-HT7 receptor subtype with an IC50 of 18 µg/mL. This 75% EtOH extract is being

132 evaluated in accelerated stability studies for a minimum of three years. The

chemical standardization and specification the clinical extracts display (figure 43)

appeared consistent. In comparison with a popular commercial product, the

commercial extract only demonstrated roughly 1/20th the concentration of ‘active’ constituents as the Black Cohosh extracts created for use within the Center for clinical study. Heavy metals, pesticide, bacterial counts and mutagenic assay data for the clinical extract were performed in independent contract labs

(appendix section 10.3). Heavy metals, pesticide and bacterial values of the extract were all within acceptable limits. 211, 212

133 Relative Abundance

extract intermsoftheabundantmolecularions. bottom afteroneyear.Theoverlayed was analyz chromatogram (TIC)ofthePhaseI Figure 42. ed fortriterpeneglycosidesattimezeroand Electrospray

Minutes LC-MS (negative-ionmode)totalIon 134 clinical extract. TIC’s validateth The TIConthetop e stabilityofthe t he TIConthe

Phase I extract

Phase I clinical cap

mV

Remifemin™ (200 mg/ml)

Phase II clinical cap

0.00 5.00 10.00 15.00 20.00 25.00 30.00 35.00 40.00 45.00 50.00 55.00 60.00 65.00 Minutes Figure 43. Clinical capsule HPLC-ELSD triterpene comparison. Phase I extract (01- 73-03), Phase I clinical capsule (Cap 3 102401), Remifemin(101141)*, Phase II clinical capsule (group 5 032303). Concentration 10 mg/mL.

135 6.0 DISCUSSION

6.1 Clinical Extract Formulation and Biological Activity

The major undertaking of the Center and the dissertation was to formulate

a botanically, biologically and chemically standardized extract for administration

in FDA-style clinical trials. Therefore, the three potential areas for difficulties are:

i) Botanical identification

ii) Chemical Standardization

iii) Biological significance of the formulation

Initially plant material collected by Center personnel to ensure the

research end of development would use the appropriate plant material. However,

with large scale production taking place outside of Project 1 and the University,

the authenticity of material was not guaranteed. Generally speaking, voucher

specimens, which could be used to positively identify a botanical used for

production of an extract are not available from raw material and finished product

manufacturers alike. Therefore, PCR methods as previously discussed were

used to evaluate starting material. These methods were shown to have some

difficulty with dried root material. For our purposes, however, they were sufficient to authenticate the starting plant material as shown in the results. On the other hand, with Black Cohosh increasing in popularity, the amount of biomass to fill the demand will be an area of concern. With most black cohosh commercial products using wild-crafted material for production, the issue of economically adulterated products is becoming an issue. Products using related Cimicifuga

136 species are considerably less expensive than C. racemosa and would be the logical candidates to be used as adulterants. PCR methods are generally not employed by commercial manufacturers to authenticate plant material.

Additionally, classical pharmacognosy techniques such as microscopy will not offer any selectivity between root material cellular characteristics among different species, and TLC which might not be sensitive or selective enough to establish differences among species. Preliminary HPLC-ELSD data on related Asian species (figure 44) shows distinctive differences between methanolic extracts of the commercial material and these Asian species shown. Only C.racemosa, C. foetida and C.heraclefolia appear to possess the triterpene 23-epi-26- deoxyactein. This methodology can be developed further for potential use to distinguish adulterated C. racemosa extracts by assaying additional samples and identifying other significant marker compounds that may be unique to Cimicifuga species. Development a library of chromatographic and spectral data, using a variety of standard compounds and extracts from the Cimicifuga species used in commerce, using the methodology herein will most likely provide unique fingerprinting data to properly identify and authenticate the species of the extract.

Chemical standardization of the extract was performed by HPLC-ELSD and is discussed in section 6.3. It is of significance to note that, based on our work in this area members of Project 1 in the center were selected as experts to contribute to the USP expert committee panel on the standardization of Black

Cohosh (PF 28(5)) and the American Herbal Pharmacopoeia monograph on

Black Cohosh.

137

1

2

3

mV

4

5

6

0.00 5.00 10.00 15.00 20.00 25.00 30.00 35.00 40.00 45.00 50.00 55.00 60.00 65.00 Minutes Figure 44. Comparison of Pure World (1) starting material to Asian species; Cimicifuga foetida (2), C. acerina (3) , C. heraclefolia (4), C. dahurica (5), C.simplex (6) Arrow indicates the presence of 23-epi-26-deoxyactein. The presence or absence of a peak in this instance is used as a biomarker to identify chemical differences within species. In this instance the three species without any detectable 23-epi-26-deoxyactein would be difficult to verify as C. racemosa, due to their lack of a substantial constituent of the botanical in question.

138 6.2 Biological Activity of Black Cohosh

Next to soy, black cohosh is the most widely studied botanical for menopausal symptoms. At the inception of the Center in 2000, the mechanism of action for Black Cohosh remained somewhat of an enigma. Long believed to have a direct estrogenic effect, studies produced by the Center as well as other research groups, has results to the contrary. The timing of this research fortunately coincided with the largest Women’s Health Study using estrogens to date, the Women’s Health Initiative (WHI). The risk factors discovered in the

WHI, increased the concern of healthy women towards using ERT and HRT for relief of climacteric symptoms. In addition, the use of selective-serotonin re- uptake inhibitors (SSRI), to relieve climacteric symptoms was awarded that indication by the FDA. A short period after the Center produced results indicating serotonin binding activity of Black Cohosh extracts. These extracts displayed competitive binding activity to the 5HT-7 and 5HT-1A cell lines in comparison to

[3H]-lysergic acid diethylamide (LSD) and [3H]-8-hydroxy-2-(di-N- propylamino)tetralin, respectively. Analysis of the ligand binding data indicated that components of a black cohosh methanol extract functioned as a mixed competitive ligand of the 5-HT7 receptor. Additionally, a black cohosh methanol extract elevated cAMP levels in 293T-5-HT7-transfected HEK cells, suggesting the extract acted as a partial agonist at the receptor. The elevation in cAMP mediated by the black cohosh extract could be reversed in the presence of the antagonist methiothepin, indicating a receptor-mediated process.121

139 The Center data implies that Black Cohosh is at least a partial serotonin agonist.

However, the compound(s) responsible for this activity in humans and if black

cohosh actually reaches serotonin receptors in the brain and has effects on hot

flashes in women is still unknown. While the BuOH-soluble fraction, in addition to

some of its constituents (i.e. cimicifugic acids A and B) have shown significant

activity within the Center, it is doubtful that a) these polyphenolic constituents would reach the CNS based on metabolic considerations, and b) the administration of a BuOH-soluble extract, would be unlikely, due to potential toxicity from residual solvent.

With the discovery of the guanidine constituents in Black Cohosh, the potential for complexes with the polyphenolics exists. This discovery may potentially provide an abundant number of possible novel structures for bioassay.

These guanidines, exhibit mesomeric chemical behavior, depending on the pH of their chemical environment, this may offer additional stability to the polyphenolics in the body, when complexed, possibly presenting the feasibility to reach the

CNS.

6.3 Chemical and Botanical Nomenclature

The proposed nomenclatural change of 27-deoxyactein (β-D- xylopyranoside, (3β,12β,16 β,23S,24R,25R)-12-(acetyloxy)-16,23:23,26: 24,25- triepoxy-9,19-cyclolanostan-3-yl) to 23-epi-26-deoxyactein was published by the

UIC/NIH Center. The basis for the change was that the designation for actein (β-

D-xylopyranoside,(3β,12β,16β,23R,24R,25S,26S)-12-(acetyloxy)16,23:23,26:

140 24,25-triepoxy-26-hydroxy-9,19-cyclolanostan-3-yl) has the hydroxyl group in

located at C-26. The corresponding reduced derivative, the former, 27-

deoxyactein is missing the hydroxyl group at that same position. Hence, 27-

deoxyactein is an incorrect name. In addition, the name 26-deoxyactein to the

former 27-deoxyactein would imply that the compound possesses the same

stereochemistry as actein, which would also be incorrect. The former 27-

deoxyactein is an epimer at C-23, thus the name 23-epi-26-deoxyactein was

applied to the former 27-deoxyactein in the effort to lend consistency to the

chemical literature.137 While this distinction was resolved as a result of a structure

elucidation effort, confusion still surrounds the stereochemistry, and consequently the nomenclature of other black cohosh triterpenes. The best example, actein (26-R/S), first isolated in the 1950’s, and the most abundant consitiuent of black cohosh, is near identical to 26-deoxyactein, yet no report in the literature exists of an epimer, stereochemically related to 23-epi-26- deoxyactein.

With the correction in the chemical nomenclature of 27-deoxyactein to 23- epi-26-deoxyactein by the Center, the issue of botanical nomenclature also needs to be addressed. Based on the evidence of one paper cross referencing the genetic similarities between Cimicifuga and Actaea, some within the scientific community now refer to Black Cohosh as Actaea racemosa,53 which is a

synonym that has not been formally adopted by the International Code of

Botanical Nomenclature (IUCN). Avoidable nomenclatural changes based on

genetic data should be avoided in order to maintain nomenclatural stability for

141 global communication about plant genetic resources. If the provisions of the

International Code of Botanical Nomenclature are followed, then very large

numbers of new combinations (names) will be needed at the species and

infraspecific level, which would lead to much confusion. Therefore the use of

Actaea racemosa for Cimicifuga racemosa is inconsistent with the intent of

Article 14 of the International Code of Botanical Nomenclature, which aims to

provide the means, by which the interests of nomenclatural stability may be best

served and in this instance preserved.54 Unless the conversion of all Cimicifuga species to Actaea is officially recognized and adopted by the authoritative bodies of botanical nomenclature it is best to use the generic name Cimicifuga to ensure the consistency of the literature and communication.

6.4 Preparative and Analytical Techniques

Based on the early study within the UIC Center, the n-BuOH fraction demonstrated the greatest activity To better characterize the n-BuOH soluble fraction, the isolated constituents, which include cimicifugic acids A (tR 42.63

min), B (43.26) and F (45.91), fukinolic acid (39.31), ferulic acid (34.58), isoferulic

acid (35.58), and cimipronidine (21.12), were analyzed individually by HPLC-

ELSD to determine their relative reverse-phase HPLC retention times. Individual

UV spectra were recorded during HPLC using a photodiode array (PDA) detector

to confirm the identity of the known constituents ferulic acid, isoferulic acid,

fukinolic acid, cimicifugic acids A, B, and F in the fraction, all with known

chromaphores. The HPLC-ELSD chromatogram in figure 45 also demonstrates

the very high polarity of the active fraction.

142 The butanol-soluble fraction contains phenolic acids and more polar constituents than the previously reported phytochemical studies of Cimicifuga.134,

135, 137, 138, 140, 142, 143, 146-148, 152-158, 160-171, 173, 213-228 The polarity of this fraction is demonstrated in Figure 50. The standard phytochemical isolation strategies usually incorporate silica as a ’normal phase’ stationary support. The use of silica with polar constituents is problematic, the more polar the compounds are, the more they are adsorbed, maybe even absorbed. Thus, the use of the standard phytochemical isolation strategies to obtain constituents from the more polar fractions and extracts is undesirable. Alternative chromatographic methods were considered to effectively isolate compounds from the more polar fractions/extracts. These included weak ion-exchange (XAD-2), size-exclusion

(LH-20), and other non-silica, reverse-phase methodologies (MCI® gel CHP20P).

These methodologies, while not entirely novel are underutilized by the natural products research community, with most of the research focused on relatively non-polar plant extracts and fractions. However, these methods as supported by this work may be the most efficient and advantageous way to isolate polar constituents from higher plants. The most significant advantage is the capability of these stationary phases to produce diverse fractions with polar starting material in a semi-predictable manner as shown in figures 15 and 16.

Amberlite® XAD-2 was selected as a bulk resin based on its reported ability to separate phenolics, and relative low cost compared to C18, MCI® Gel

CHP20P, Sephadex® LH-20 and zirconium (Zr) resins that were considered.

XAD-2 is a styrene based sized exclusion resin that must be washed thoroughly

143 prior to use with all eluting solvents. With the resultant fractions from XAD-2,

which was monitored by TLC (with UV and spray reagents (anisaldehyde and

sulfuric acid (10 % in EtOH) for detection) and HPLC-ELSD, we were able to

observe 35-45 distinctive spots and peaks, respectively, after the 2nd level of

fractionation, enhancing the initial solvent fractionation by roughly 10-fold. MCI

gel® CHP20P and Sephadex® LH-20 were used for the 3rd level of fractionation.

From the TLC and HPLC-ELSD fraction control results, 100 distinct spots and

peaks, representing numerous fractions and compounds, were detected. With

semi-preparative HPLC purification of the resultant fractions, as shown with

fraction G15, one 3rd level fraction, additional peaks were detected from the semi-preparative separation in comparison to what was visible in the analytical

HPLC due to analytical column overload. If 3rd level separation efforts were

initiated on ALL second level XAD-2 fractions (figure 13), HPLC purification of

ALL of theses resultant 3rd level fractions (figure 15), would yield 4th level separation that could potentially yield thousands of constituents. With the complexity of the 3rd and 4th level fractions the development of ‘universal’

detection methods was essential for the production of a relatively humble number

of fractions and compounds. In order to make comprehensive characterization of

a Cimicifuga racemosa extract a reality, the limiting factor will be the

development of ‘universal’ detection methods using higher throughput techniques

to avoid replication of isolation efforts.

Routine isolation of plant derived natural products, when polyphenolic

constituents are present generally requires the use of acids.229, 230 Zwitterionic

144 compounds such like cimipronidine are pH-sensitive and, thus, with the addition

of acid may introduce significant variability in the preparative HPLC retention

properties of the zwitterionic constituents as was observed in this semi-

preparative HPLC isolation of cimipronidine. In this instance, the addition of TFA reduced the retention time of the highly polar material. This may also result in a complexation of the zwitterionic components with the polyphenolic constituents found in the polar extracts.202, 231 This possibility presented it self in this study as

depicted in figure 51 which shows the elution of both fukinolic acid and

cimipronidine in fraction 6 (from G15). While this figure does not serve as a

confirmation of complexation, it does provide basis for additional experiments to

determine complexation.

Analytical HPLC evaluation of hydroalcoholic commercial extracts of

C.racemosa has been relatively limited to the detection and quantification of

triterpene glycosides.48, 210 Identification and quantitiation of triterpenoids presents

a challenge in detection of major compounds that are generally considered poor

chromophores, thus the application of HPLC-UV detection is not appropriate.

The recent use of in-line ELSD and MS detection has made the reliable and

robust quantitation of a commercial product available. However, the majority of

these studies only address one class of constituents, i.e., the triterpenes, in a complex matrix including aromatic acids, oils, sugars and resins, which have been reported from hydroalcoholic extracts of Cimicifuga racemosa. In addition,

the With greater serotonergic activity present in the more polar, butanol-soluble

fractions, standardization methods will need to be developed to rapidly separate,

145 quantify and identify known constituents of polar extracts and fractions, as well

as having the ability to separate new structures from these fractions. As with any

multi-component product, which includes all botanicals, the development of

methodology that best correlates to some biological activity is the most desired in

terms of commercially available products, offering the consumer some level of

assurance of quality. Botanical dietary supplement analysis does not follow the

same straight ahead approach as pharma QA/QC models (i.e. single point standard curves) to standardize a product. While achieving this model is not a possibility, using the best components of a pharma QA/QC model, such as standard-spike recoveries, would benefit the analyses of the complex matricies encountered in botanical analytical chemistry

Positive-ion electrospray was shown to be an excellent ionization technique for the mass spectrometric detection of guanidine constituents and polar phenolic acids. Compounds with an odd number (n=1,3,5,7,9…) of nitrogen atoms were detected as even numbered mass protonated molecules.

146 R1 R2 R3 Fukinolic Acid H H OH 34 9

2 5 COOH Cimicifugic Acid A CH3 H OH 1 N 8 1000 d B Cimicifugic Acid C H CH OH i 3

6 c 7 A 1 d Cimicifugic Acid F H CH3 H 900 2 HN NH c ci gi A fu c mV i 800 i O l d A c i i o c n

m R i

A 1

700 k O c Ci Fu

d F COOH ugi i HO f

600 ) R i c 2 COOH 1 c ( i A d i

e R d

c 3 m i 500 n i ci Ac C d ugi A i c f i i c i c on

400 l ul OH mi pr ru er i i of C m

300 Fe s I Ci

200

100

0

0.00 5.00 10.00 15.00 20.00 25.00 30.00 35.00 40.00 45.00 50.00 55.00 60.00 65.00 min

140.00 1000.00 Phenolics Triterpene glycosides 120.00 800.00 and ??? 100.00

600.00 80.00

V V m m 60.00 400.00 40.00

200.00 20.00

0.00 0.00

0.00 5.00 10.00 15.00 20.00 25.00 30.00 35.00 40.00 45.00 50.00 55.00 60.00 65.00 10.00 20.00 30.00 40.00 50.00 60.00 70.00 Minutes Minutes Isoferulic acid in the butanol-soluble fraction Isoferulic acid in the clinical extract Method B Method A

Relative Polarity of C. racemosa Constituents

Resins unknown known phenolic phenolic triterpenes and ??? compounds compounds

Figure 45. HPLC-ELSD trace of the BuOH-soluble fraction (01-75-15), from which 1 was isolated (solvent A: 0.05% TFA/5% MeOH/H2O, solvent B: 0.05% TFA/MeOH; elution gradient: 0.0 min A=97%, 50.0 min. A=25%, 61.0 min A=25%, 62.0 A=10%), Retention times: 34.587-ferulic acid, 35.584-isoferulic acid, 39.309-fukinolic acid, 42.632-cimicifugic acid A, 43.263-cimicifugic acid B, 45.913-cimicifugic acid F. Additional HPLC-ELSD traces are present to make evident the difference in polarity between the clinical extract and BuOH-soluble

fraction.

147 Cimpronidine (1) signature Fukinolic acid (2) signature

10 9 8 7 6 5 4 3 2 1 ppm

Figure 46. P6 (from G15)1H-NMR spectrum (500 MHz) in MeOH-d4. Potential complexation of cimipronidine and fukinolic acid. While not conclusive evidence this is a semi-preparative fraction that was eluted between 1 and 2, approximately 20 minutes after 1 and 10 minutes before 2 in fraction G-15 .

148 6.5 NMR spectroscopy

The relatively simple structure of cimipronidine (1) offered quite a complex

structure elucidation problem. Guanidine compounds have only been

reported within a very limited number of plant genera.206-208 Among the

constituents pictured in figure 47, most have a long aliphatic component, thus

the 1H-NMR data will be of a greater merit in terms of providing information for

elucidating the structure. The principle difficulty with 1 is that with the

guanidine moiety centralized on the ring, the ring protons appear quite broad

in the 1H-NMR spectra, offer little spectral information (i.e. lack of coupling

constants) and do not offer long range HMBC correlations. The reasons for

this may be the following: (i) rapid fluctuations associated with 5-membered

ring dynamics, which are known to occur and which can lead to broadening of

the proton resonances with concomitant introduction of short T2’s that affect

the efficiency of polarization transfer from proton to carbon; (ii) the fact that

2 typical JH, C couplings are generally reduced in magnitude relative to 3-bond

couplings; (iii) for the expected 3-bond correlations C-5/H-2, C-5/H-3, C-4/H-

2, and C-3/H-5 the dihedral angle between these protons and the indicated

carbon atoms in the ring approach a dynamically averaged value of 90o, and,

therefore, expected correlations are too weak to be observed or are

completely absent (effective coupling is zero). This latter alternative is

probably the most reasonable explanation. The guanidine carbon NMR shift

ca. 150 ppm will be the one constant observable trend in these structures.

While a higher field strength instrument will usually offer higher resolution with

149 many structure elucidation issues, they offered no additional benefit in

elucidating 1 and will offer the same frustration with other guanidines. The

higher resolution, improved signal-to-noise ratio and greater sensitivity that

will solve many elucidation problems will offer no benefit with the rapid proton fluctuations relative to the NMR timescale. Solubility is an important challenge with guanidine protons. The use of a variety of NMR solvents with various concentrations of both, acid and base are necessary in confirming the presence of guanidine protons, as was needed for 1 (figure 30). Additionally, the possibility of complexation of nitrogen bearing compounds with substituted phenolics appeared during the isolation of 1. To further resolve the proton signals and for confirmation of complexation with phenolics by spectroscopic techniques, the stoichiometry of the complexes still remains to be determined, and may be easily rectified by qNMR. This complexation may be significant to the biological activity of Black Cohosh.

150 O H N NH N 2 H NH HO p-coumaroyl-agmatine HO

segetalin H

NH O O H H N N O H2N NH HN H O H N OH galegine H N N N 2 H O NH Ph OMe MeO H NH N N NHR O H

caracasanamide G1 (R = iso-C6H15) caracasanamide G5 (R = H) H NNHR H2N NH caracasanamide G3 (R = iso-C6H15) caracasanamide G6 (R = H)

Figure 47. Other guanidine isolates from higher plants

151 7.0 CONCLUSIONS

7.0.1. Review of the pharmacognosy of Black Cohosh

The introductory material (pages 1-38) serves as a comprehensive survey

of the clinical, biological and pharmacological literature to date of Black Cohosh.

7.0.2. Preparation of a biologically, chemically and botanically standardized

extract for clinical trial.

To this end, the use of RAPD-PCR, HPLC-ELSD and numerous bioassay

systems within the Center were used to select a formulation. The formulation is

unique in that the techniques above were used to verify the authenticity of the

plant material, select the best extract type of a number of extracts for a clinical

trial based on quantitation of the active constituents and the biological activity.

The result was:

-75% ethanolic extract

-Chemically standardized to 5% of the active triterpene glycosides as determined

by the UIC Center (actein (26R), actein (26S), 23-epi-26-deoxyactein and 26- deoxyactein)

-Biologically standardized to competitively inhibit 5HT-7 binding of 3H-LSD at 18

µg/ml.

7.0.3. Chemical characterization of the active butanolic Black Cohosh fraction

The initial butanol soluble fraction (figure 10) demonstrated significant

activity in the 5HT7 (IC50 4.78) bioassay used to characterize the anti-climacteric

152 activity of Black Cohosh. The constituents that make up this activity were isolated and elucidated within the center. HPLC-ELSD methodology has been developed to identify the presence of these constituents in the fraction. Isolation work on this fraction led to the first report of an alkaloid from Black Cohosh, a novel cyclic guanidine alkaloid. The compounds identified from this fraction are:

-Cimicifugic acids A, B, C, D, F

-Cimipronidine

-Ferulic acid

-Fukinolic Acid

-Isoferulic acid

7.0.4. Development of preparative and analytical methods

Chromatographic methods comprising non-silica (XAD-2 and CHP20P column chromatography), silica (column chromatography) and RP-HPLC have been developed for the serotonergic constituents from Black Cohosh. The results of the chromatographic experiments have been digitized and deposited on the

Center file server (humulus.pharm.uic.edu) to assist in future isolation efforts on

Black Cohosh. The following methods/experimental information are deposited on

Humulus:

- XAD-2 separation of the butanol-soluble fraction

- MCI gel CHP20P separations of

- TLC fraction control of XAD-2, MCI gel CHP 20P and Sephadex fractions

using SSC7-SSC9, and a variety of detection methods

153 - HPLC-ELSD data for clinical extracts, fractions and pure compounds

7.0.5. Dereplication of Botanical Center isolates

The guanidine compound that was identified (1) has not been previously catalogued in either the Chemical Abstracts or Beilstein databases. Thus, it is a new compound, the first report of an alkaloid from Black Cohosh. The processed spectral data have been digitized and deposited on the Center fileserver

(humulus.pharm.uic.edu) to assist in future structure elucidation efforts on Black

Cohosh. Data on the following chemical classes can be distinguished by comparison with the data herein:

- cyclic guanidines

- fukiic acid esters

- 9,19 cycloartane triterpenes

- cinnamic acid derivatives

7. 1 Summary of Conclusions

The work presented in this dissertation can serve as a reference for producing chemically, botanically and biologically standardized botanical extracts for clinical study. More specifically it can serve as a guide for future production and chemical analysis of a standardized Black Cohosh formulation. The chromatography and elucidation work provided herein provide a general compass for future efforts to isolate guanidine compounds, additional triterpene and phenolic compounds from black cohosh.

154 8.0 Future directions

The Center-developed standardized Black Cohosh extract is currently in a

Phase II clinical study at UIC. Thus, the issue of efficacy surrounding the extract will be resolved on completion of the study. However, other research issues have yet to be addressed:

- Production of novel methods to determine identity, strength and purity of

Black Cohosh commercial products. While current methods are

acceptable, they can be improved by adding spike recoveries, studying

the effects of different matricies and solvents on the major constituents. In

addition to determining what exactly the parameters of a quality

commercial product is on the basis of chemistry and biology. Only extracts

have been clinically trialed, still there are numerous products that are

powdered Black Cohosh, yet no equivalence of powdered material to

extracts exists in the compendial literature.

- Preliminary work provided herein shows the qualitative difference between

the Asian and North American Cimicifuga spp. by HPLC-ELSD. While

standards for C. racemosa are widely available to verify the quality and

quantitate commercial Black Cohosh products, little work has been done

to identify the major constituents in Asian Cimicifuga, which are

purportedly used as economic adulterants. In addition the use of ELSD is

not a reality in all academic or commercial labs that may work with Black

Cohosh, additional low cost techniques need to be developed at the very

155 least to ensure the identity of Black Cohosh genetic material and plant

extracts.

- Based on NMR data, the stereochemistry of actein (R/S), the most

abundant triterpene has not been resolved to the degree of 23-epi-26-

deoxyactein and 26-deoxyactein and should be further studied. Actein is

frequently used as a standard in Black Cohosh chemical analysis.

Mutarotation of this compound translates to the use of an uncalibrated,

variable standard for analysis. The mutarotation will vary in different

solvents, therefore effecting the ratio of R:S isomer.

- The interaction between guanidine compounds and phenolics in Black

Cohosh to form complexes in polar solutes, like water, should be studied.

LC-MS/MS indicated the presence of other guanidine compounds yet to

be isolated and characterized. Determination of the isoelectric points of

the guanidines will assist in determination of new structures.

- As additional bioassays are developed that correlated to the alleviation of

hot flushes and other climacteric symptoms, further bioassay guided

fractionation and structure elucidation efforts will be necessary. Future

studies will need to be conducted to determine if black cohosh actually

reaches serotonin receptors in the brain and has effects on hot flashes in

women.

- Reproducible and robust protocols for DNA isolation from dried root, not

only for Black Cohosh, but for other botanicals, can be developed to make

156 the use of genetic fingerprinting more widely available. This will alleviate much of the concern of adulterated material.

157 9.0 References

1. NIH, Workshop on the Safety of Black Cohosh in Clinical Studies. 2004, National Center for Complementary and Alternative Medicine, NIH Office of Dietary Supplements: Bethesda. p. 40. 2. Winterhoff, H., Butterweck, V., Jarry, H. and Wuttke, W. [Pharmacologic and clinical studies using Cimicifuga racemosa in climacteric complaints]. Wien Med Wochenschr. 2002, 152, 360-3. 3. Seidl, M.M. and Stewart, D.E. Alternative treatments for menopausal symptoms. Systematic review of scientific and lay literature. Can Fam Physician. 1998, 44, 1299-308. 4. Taechakraichana, N., Jaisamrarn, U., Panyakhamlerd, K., Chaikittisilpa, S. and Limpaphayom, K.K. Climacteric: concept, consequence and care. J Med Assoc Thai. 2002, 85 Suppl 1, S1-15. 5. Roemheld-Hamm, B. and Dahl, N.V. Herbs, menopause, and dialysis. Semin Dial. 2002, 15, 53-9. 6. Pick, M. Herbal treatments for menopause. Black cohosh, soy and micronized progesterone. Adv Nurse Pract. 2000, 8, 29-30. 7. Pepping, J. Black cohosh: Cimicifuga racemosa. Am J Health Syst Pharm. 1999, 56, 1400-2. 8. Moyad, M.A. Complementary/alternative therapies for reducing hot flashes in prostate cancer patients: reevaluating the existing indirect data from studies of breast cancer and postmenopausal women. Urology. 2002, 59, 20-33. 9. McKenna, D.J., Jones, K., Humphrey, S. and Hughes, K. Black cohosh: efficacy, safety, and use in clinical and preclinical applications. Altern Ther Health Med. 2001, 7, 93-100. 10. Liske, E. Therapeutic efficacy and safety of Cimicifuga racemosa for gynecologic disorders. Adv Ther. 1998, 15, 45-53. 11. Lieberman, S. A review of the effectiveness of Cimicifuga racemosa (black cohosh) for the symptoms of menopause. J Womens Health. 1998, 7, 525-9. 12. Kronenberg, F. and Fugh-Berman, A. Complementary and alternative medicine for menopausal symptoms: a review of randomized, controlled trials. Ann Intern Med. 2002, 137, 805-13. 13. Portallis, M.I., Focus on hormone replacement research. 2004, New York: Nova Biomedical Publications. xi, 172. 14. American Medical Association., Essential guide to menopause. 1998, New York: Pocket Books. xi, 253. 15. Barrett-Connor, E., Grady, D. and Stefanick, M.L. The Rise and Fall of Menopausal Hormone Therapy. Annu Rev Public Health. 2005, 26, 115- 140.

158 16. Henderson, V.W., Benke, K.S., Green, R.C., Cupples, L.A. and Farrer, L.A. Postmenopausal hormone therapy and Alzheimer's disease risk: interaction with age. J Neurol Neurosurg Psychiatry. 2005, 76, 103-5. 17. Henderson, V.W., Guthrie, J.R., Dudley, E.C., Burger, H.G. and Dennerstein, L. Estrogen exposures and memory at midlife: a population- based study of women. Neurology. 2003, 60, 1369-71. 18. Henderson, V.W., Paganini-Hill, A., Miller, B.L., Elble, R.J., Reyes, P.F., Shoupe, D., McCleary, C.A., Klein, R.A., Hake, A.M., and Farlow, M.R. Estrogen for Alzheimer's disease in women: randomized, double-blind, placebo-controlled trial. Neurology. 2000, 54, 295-301. 19. Liehr, J.G. Genotoxicity of the steroidal oestrogens oestrone and oestradiol: possible mechanism of uterine and mammary cancer development. Hum Reprod Update. 2001, 7, 273-81. 20. Liehr, J.G., Stancel, G.M., Chorich, L.P., Bousfield, G.R. and Ulubelen, A.A. Hormonal carcinogenesis: separation of estrogenicity from carcinogenicity. Chem Biol Interact. 1986, 59, 173-84. 21. Bolton, J.L., Pisha, E., Zhang, F. and Qiu, S. Role of quinoids in estrogen carcinogenesis. Chem Res Toxicol. 1998, 11, 1113-27. 22. Bolton, J.L., Trush, M.A., Penning, T.M., Dryhurst, G. and Monks, T.J. Role of quinones in toxicology. Chem Res Toxicol. 2000, 13, 135-60. 23. Craig, M.C., Maki, P.M. and Murphy, D.G. The Women's Health Initiative Memory Study: findings and implications for treatment. Lancet Neurol. 2005, 4, 190-4. 24. Doty, T.J. Postmenopausal hormone replacement therapy after the Women's Health Initiative. Jaapa. 2005, 18, 30-7; quiz 39-40. 25. Gambrell, R.D., Jr. The women's health initiative reports in perspective: facts or fallacies? Climacteric. 2004, 7, 225-8. 26. Colditz, G.A., Hankinson, S.E., Hunter, D.J., Willett, W.C., Manson, J.E., Stampfer, M.J., Hennekens, C., Rosner, B. and Speizer, F.E. The use of estrogens and progestins and the risk of breast cancer in postmenopausal women. N Engl J Med. 1995, 332, 1589-93. 27. Hahn, R.G. Compliance considerations with estrogen replacement: withdrawal bleeding and other factors. Am J Obstet Gynecol. 1989, 161, 1854-8. 28. Barber, C.A., Margolis, K., Luepker, R.V. and Arnett, D.K. The impact of the Women's Health Initiative on discontinuation of postmenopausal hormone therapy: the Minnesota Heart Survey (2000-2002). J Womens Health (Larchmt). 2004, 13, 975-85. 29. Treatment of menopausal vasomotor symptoms. Med Lett Drugs Ther. 2004, 47, 98-9. 30. Keller, C., Fullerton, J. and Mobley, C. Supplemental and complementary alternatives to hormone replacement therapy. J Am Acad Nurse Pract. 1999, 11, 187-98. 31. Azoulay, C. [Menopause in 2004: "hormone replacement therapy" is not what it used to be anymore]. Rev Med Interne. 2004, 25, 806-15.

159 32. Fabricant, D.S. and Farnsworth, N.R. The value of plants used in traditional medicine for drug discovery. Environ Health Perspect. 2001, 109 Suppl 1, 69-75. 33. Akerele, O. WHO's traditional medicine programme: progress and perspectives. WHO Chron. 1984, 38, 76-81. 34. Akerele, O. Nature's medicinal bounty: don't throw it away. World Health Forum. 1993, 14, 390-5. 35. Farnsworth, N.R. The development of pharmacological and chemical research for application to traditional medicine in developing countries. J Ethnopharmacol. 1980, 2, 173-81. 36. Farnsworth, N.R. The role of ethnopharmacology in drug development. Ciba Found Symp. 1990, 154, 2-11; discussion 11-21. 37. Farnsworth, N.R. Ethnopharmacology and drug development. Ciba Found Symp. 1994, 185, 42-51; discussion 51-9. 38. Eisenberg, D.M., Davis, R.B., Ettner, S.L., Appel, S., Wilkey, S., Van Rompay, M. and Kessler, R.C. Trends in alternative medicine use in the United States, 1990-1997: results of a follow-up national survey. Jama. 1998, 280, 1569-75. 39. Brevort, P. The Booming U.S. Botanical Market: A new Overview. Herbalgram. 1999, 44, 51. 40. The use of black cohosh to treat symptoms of menopause. Fertil Steril. 2004, 81, 27-34. 41. Borrelli, F. and Ernst, E. Cimicifuga racemosa: a systematic review of its clinical efficacy. Eur J Clin Pharmacol. 2002, 58, 235-41. 42. Lehmann-Willenbrock, E. and Riedel, H.H. [Clinical and endocrinologic studies of the treatment of ovarian insufficiency manifestations following hysterectomy with intact adnexa]. Zentralbl Gynakol. 1988, 110, 611-8. 43. Duker, E.M., Kopanski, L., Jarry, H. and Wuttke, W. Effects of extracts from Cimicifuga racemosa on gonadotropin release in menopausal women and ovariectomized rats. Planta Med. 1991, 57, 420-4. 44. Stoll, W. Phytopharmacon influences atrophic vaginal epithelium: double- blind study -Cimicifuga vs. estrogenic substances. Therapeuticum. 1987, 1, 23-31. 45. Stefan, H. An essay on the manifestations and therapy of hormone-related female biopathic syndrome. Ringelh. Biol. Umsch. 1959, 10, 149-152. 46. Treatment of menopause-associated vasomotor symptoms: position statement of The North American Menopause Society. Menopause. 2004, 11, 11-33. 47. Liu, J., Burdette, J.E., Xu, H., Gu, C., van Breemen, R.B., Bhat, K.P., Booth, N., Constantinou, A.I., Pezzuto, J.M., Fong, H.H., Farnsworth, N.R., and Bolton, J.L. Evaluation of estrogenic activity of plant extracts for the potential treatment of menopausal symptoms. J Agric Food Chem. 2001, 49, 2472-9. 48. Li, W., Chen, S.-N., Fabricant, D., Angerhofer, C., Fong, H.H.S., Farnsworth, N.R. and Fitzloff, J.F. High-performance liquid chromatographic analysis of Black Cohosh constituents with in-line

160 evaporative light scattering and photodiode array detection. Analytica Chimica Acta. 2002, 471, 61-75. 49. Kennelly, E.J., Baggett, S., Nuntanakorn, P., Ososki, A.L., Mori, S.A., Duke, J., Coleton, M. and Kronenberg, F. Analysis of thirteen populations of black cohosh for formononetin. Phytomedicine. 2002, 9, 461-7. 50. Gleason, H.A. and Cronquist, A., Manual of vascular plants of northeastern United States and adjacent Canada. 2nd ed. 1991, Bronx, N.Y.: New York Botanical Garden. lxxv, 910. 51. Gleason, H.A. and Cronquist, A., Manual of vascular plants of Northeastern United States and adjacent Canada. 1963, Princeton, N.J.,: Van Nostrand. li, 810. 52. Cronquist, A., An integrated system of classification of flowering plants. 1981, New York: Columbia University Press. xviii, 1262. 53. Bakker, F.T., Culham, A., Gomez-Martinez, R., Carvalho, J., Compton, J., Dawtrey, R. and Gibby, M. Patterns of nucleotide substitution in angiosperm cpDNA trnL (UAA)-trnF (GAA) regions. Mol Biol Evol. 2000, 17, 1146-55. 54. Greuter, W., International code of botanical nomenclature : Tokyo code : adopted by the Fifteenth International Botanical Congress, Yokohama, August-September 1993. Regnum vegetabile, v. 131. 1994, Kèonigstein, Germany: Koeltz Scientific Books. xviii, 389. 55. Greuter, W., Report on botanical nomenclature--Saint Louis, 1999 : XVI International Botanical Congress, Saint Louis, Nomenclature Section, 26 to 30 July 1999. Englera, 20. 2000, Berlin: Botanischer Garten und Botanisches Museum Berlin-Dahlem. 253. 56. Ramsey, G.W. A Comparison of Vegetative Characteristics of Several Genera with Those of the Genus Cimicfuga (Ranunculaceae). SIDA. 1988, 13, 57-63. 57. Ramsey, G.W. Morphological Considerations in the North American Cimicifuga (Ranunculaceae). Castanea. 1987, 52, 129-141. 58. Youngken, H., A textbook of pharmacognosy. 4th ed. Vol. xiv. 1936, Philadelphia: P. Blakiston's son and Co. 924. 59. King, J., The American dispensatory. 5th ed. 1859, Cincinnati: Moore Wilstach Keys & Co. 1475. 60. King, J. and Felter, H.W., The American eclectic dispensatory. 1st - ed. 1854, Cincinnati,: Moore Wistach & Keys etc. v. 61. Black Cohosh Rhizome, in American Herbal Pharmacopoeia and Therapeutic Compendium, R. Upton, Editor. 2002, AHP: Santa Cruz. p. 34. 62. Foldes, J. The actions of an extract of C. racemosa. Arzneimittelforschung. 1959, 13, 623-4. 63. Coates, P.M., Encyclopedia of dietary supplements. 2005, New York: Marcel Dekker. xviii. 819. 64. Hernandez Munoz, G. and Pluchino, S. Cimicifuga racemosa for the treatment of hot flushes in women surviving breast cancer. Maturitas. 2003, 44 Suppl 1, S59-65.

161 65. Baier-Jagodinski, G. Praxisstudie mit Cimisan bei klimakterischen Beschwerden, Pramenstruallen syndrom und Dysmenorrhoe. Natur Heilpraxis Naturmedizin. 1995, 48, 1284-88. 66. Wuttke, W., Seidlova-Wuttke, D. and Gorkow, C. The Cimicifuga preparation BNO 1055 vs. conjugated estrogens in a double-blind placebo-controlled study: effects on menopause symptoms and bone markers. Maturitas. 2003, 44 Suppl 1, S67-77. 67. Schotten, E. Experience with the Cimicifuga preparation, Remifemin. Der Landarzt. 1958, 34, 353-4. 68. Kesselkaul, O. [Treatment of climacteric disorders with remifemin]. Med Monatsschr. 1957, 11, 87-8. 69. Nesselhut, T. and Liske, E. Pharmacological measures in postmenopausal women with an isopropanolic aqueous extract of Cimicifuga racemosa rhizoma. Menopause. 1999, 6, 331. 70. Foldes, J. [The effects of an extract from Cimicifuga racemosa]. Arztl Forsch. 1959, 13, 623-4. 71. Starfinger, W. [A contribution to the therapy of the psychovegetative syndrome]. Dtsch Med J. 1960, 11, 308-9. 72. Liske, E., Hanggi, W., Henneicke-von Zepelin, H.H., Boblitz, N., Wustenberg, P. and Rahlfs, V.W. Physiological investigation of a unique extract of black cohosh (Cimicifugae racemosae rhizoma): a 6-month clinical study demonstrates no systemic estrogenic effect. J Womens Health Gend Based Med. 2002, 11, 163-74. 73. Schlidge, E. Essay on the treatment of premenstrual and menopausal mood swings and depressive states. Ringelh Biol Umsch. 1964, 19, 1822. 74. Daiber, W. Klimakterische Beschwerden:ohne Hormone zum Erfolg. Arzt Prax. 1983, 35, 1946-7. 75. Stolze, H. Der andere weg, Klimacterische Beschwerden zu behandlen. Gyne. 1982, 3, 14-6. 76. Vorberg, G. Therapy of climacteric complaints. Zeitschrift fur Allgemeinmedizin. 1984, 60. 77. Warnecke, G. Influence of a phytopharmeaceutical on climacteric complaints. Meizinische Welt. 1985, 36, 871-4. 78. Heizer, H. [Criticism on Cimicifuga therapy in hormonal disorders in women]. Med Klin. 1960, 55, 232-3. 79. Jacobson, J.S., Troxel, A.B., Evans, J., Klaus, L., Vahdat, L., Kinne, D., Lo, K.M., Moore, A., Rosenman, P.J., Kaufman, E.L., Neugut, A.I., and Grann, V.R. Randomized trial of black cohosh for the treatment of hot flashes among women with a history of breast cancer. J Clin Oncol. 2001, 19, 2739-45. 80. Stoll, W. Phytotherapeutikum beeinflusst atrophisches Vaginalepithel. Doppelblindversuch cimicifuga vs ostrogenpraparat. Therapeutikon. 1987, 1, 23-31. 81. Petho, A. Klimakterische beschwerden. Umsteelung einer Hormonbehandlung auf ein pflanzliches Gynakologikum moglich? Arzt Prax. 1987, 38, 1551-3.

162 82. Gorlich, N. Treatment of ovarian disorders in general practice. Arzt Prax. 1962, 14, 1742-3. 83. Brucker, A. Essay on the phytotherapy of hormonal disorders in women. Med Wel. 1960, 44, 2331-3. 84. Mielnik, J. Cimicifuga racemosa in the treatment of neuro vegetative symptoms in women in the perimenopausal period. Maturitas. 1997, 27, 215. 85. Georgiev, D.B., Iordanova, E., Phytoestrogens-the alternative approach. Maturitas. 1997, 27, P309. 86. Huntley, A. and Ernst, E. A systematic review of the safety of black cohosh. Menopause. 2003, 10, 58-64. 87. Russell, L., Hicks, G.S., Low, A.K., Shepherd, J.M. and Brown, C.A. Phytoestrogens: a viable option? Am J Med Sci. 2002, 324, 185-8. 88. Cohen, S.M., O'Connor, A.M., Hart, J., Merel, N.H. and Te, H.S. Autoimmune hepatitis associated with the use of black cohosh: a case study. Menopause. 2004, 11, 575-7. 89. Bouchet, C., Guillemin, F. and Briancon, S. Nonspecific effects in longitudinal studies: impact on quality of life measures. J Clin Epidemiol. 1996, 49, 15-20. 90. Pecins-Thompson, M., Widmann, A.A. and Bethea, C.L. Beta-endorphin, but not oxytocin, substance P or vasoactive-intestinal polypeptide, contributes to progesterone-induced prolactin secretion in monkeys. Neuroendocrinology. 1996, 63, 569-78. 91. Loprinzi, C.L., Pisansky, T.M., Fonseca, R., Sloan, J.A., Zahasky, K.M., Quella, S.K., Novotny, P.J., Rummans, T.A., Dumesic, D.A., and Perez, E.A. Pilot evaluation of venlafaxine hydrochloride for the therapy of hot flashes in cancer survivors. J Clin Oncol. 1998, 16, 2377-81. 92. Loprinzi, C.L., Sloan, J.A., Perez, E.A., Quella, S.K., Stella, P.J., Mailliard, J.A., Halyard, M.Y., Pruthi, S., Novotny, P.J., and Rummans, T.A. Phase III evaluation of fluoxetine for treatment of hot flashes. J Clin Oncol. 2002, 20, 1578-83. 93. Liao, J.F., Jan, Y.M., Huang, S.Y., Wang, H.H., Yu, L.L. and Chen, C.F. Evaluation with receptor binding assay on the water extracts of ten CNS- active Chinese herbal drugs. Proc Natl Sci Counc Repub China B. 1995, 19, 151-8. 94. Jarry, H., Harnischfeger, G. and Duker, E. [The endocrine effects of constituents of Cimicifuga racemosa. 2. In vitro binding of constituents to estrogen receptors]. Planta Med. 1985, 316-9. 95. Jarry, H. and Harnischfeger, G. [Endocrine effects of constituents of Cimicifuga racemosa. 1. The effect on serum levels of pituitary hormones in ovariectomized rats]. Planta Med. 1985, 46-9. 96. Eagon, C.L., Elm, M.S., Eagon, P.K. Estrogenicity of traditional Chinese and Western herbal remedies. Proceeding of the American Association for Cancer Research. 1996, 37, 284.

163 97. Zava, D.T., Dollbaum, C.M. and Blen, M. Estrogen and progestin bioactivity of foods, herbs, and spices. Proc Soc Exp Biol Med. 1998, 217, 369-78. 98. Jarry, H., Metten, M., Spengler, B., Christoffel, V. and Wuttke, W. In vitro effects of the Cimicifuga racemosa extract BNO 1055. Maturitas. 2003, 44 Suppl 1, S31-8. 99. Hooper, L., Thompson, R.L., Harrison, R.A., Summerbell, C.D., Moore, H., Worthington, H.V., Durrington, P.N., Ness, A.R., Capps, N.E., Davey Smith, G., Riemersma, R.A., and Ebrahim, S.B. Omega 3 fatty acids for prevention and treatment of cardiovascular disease. Cochrane Database Syst Rev. 2004, CD003177. 100. Hochberg, R.B., Pahuja, S.L., Zielinski, J.E. and Larner, J.M. Steroidal fatty acid esters. J Steroid Biochem Mol Biol. 1991, 40, 577-85. 101. Seibel, M.M. Treating hot flushes without hormone replacement therapy. J Fam Pract. 2003, 52, 291-6. 102. Liu, J., Burdette, J.E., Sun, Y., Deng, S., Schlecht, S.M., Zheng, W., Nikolic, D., Mahady, G., van Breemen, R.B., Fong, H.H., Pezzuto, J.M., Bolton, J.L., and Farnsworth, N.R. Isolation of linoleic acid as an estrogenic compound from the fruits of Vitex agnus-castus L. (chaste- berry). Phytomedicine. 2004, 11, 18-23. 103. Vallette, G., Vanet, A., Sumida, C. and Nunez, E.A. Modulatory effects of unsaturated fatty acids on the binding of glucocorticoids to rat liver glucocorticoid receptors. Endocrinology. 1991, 129, 1363-9. 104. Ingkaninan, K., von Frijtag Drabbe Kunzel, J.K., AP, I.J. and Verpoorte, R. Interference of linoleic acid fraction in some receptor binding assays. J Nat Prod. 1999, 62, 912-4. 105. Stoll, B.A. Linkage between retinoid and fatty acid receptors: implications for breast cancer prevention. Eur J Cancer Prev. 2002, 11, 319-25. 106. Pink, J.J. and Jordan, V.C. Models of estrogen receptor regulation by estrogens and antiestrogens in breast cancer cell lines. Cancer Res. 1996, 56, 2321-30. 107. Jarry, H., Leonhardt, S., Duls, C., Popp, M., Christoffel, V., Spengler, B., Theiling, K., Wuttke, W. Organ specific effects of C.racemosa in brain and uterus. in 23rd International LOF-Symposium on Phyto-Oestrogens. 1999. Belgium. 108. Liu, Z.P., Yu, B., Huo, J.S., Lu, C.Q. and Chen, J.S. Estrogenic Effects of Cimicifuga racemosa (Black Cohosh) in Mice and on Estrogen Receptors in MCF-7 Cells. J Med Food. 2001, 4, 171-178. 109. Amato, P., Christophe, S. and Mellon, P.L. Estrogenic activity of herbs commonly used as remedies for menopausal symptoms. Menopause. 2002, 9, 145-50. 110. Eagon, P.K., Tress, N.B., Ayer, H.A., Wiese, J.M., Henderson, T., Elm, M.S., Eagon, C.L. Medicinal botanicals with hormonal activity. Proceeding of the American Association for Cancer Research. 1999, 40, 161-2.

164 111. Freudenstein, J., Dasenbrock, C. and Nisslein, T. Lack of promotion of estrogen-dependent mammary gland tumors in vivo by an isopropanolic Cimicifuga racemosa extract. Cancer Res. 2002, 62, 3448-52. 112. Lohning, A., Verspohl, E., Winterhoff, H. Pharmacological studies on the dopaminergic activity of C. racemosa. in 23rd international LOF- symposium on "phyto-oestrogens". 1999. Belgium. 113. Seidlova-Wuttke, D., Jarry, H., Becker, T., Christoffel, V. and Wuttke, W. Pharmacology of Cimicifuga racemosa extract BNO 1055 in rats: bone, fat and uterus. Maturitas. 2003, 44 Suppl 1, S39-50. 114. Nisslein, T., Freudenstein, J., Effects of black cohosh on urinary bone markers and femoral density in an ovx-rat model. Osteoporosis Interantional. 2000. 115. Li, J.X., Kadota,S., Li, H.Y., Miyahara, T., Wu, Y.W., Seto, H., Namba, T., Effects of Cimicifugae rhizoma on serum calcium and phosphate levels in low calcium dietary rats and bone mineral density in ovariectomized rats. Phytomedicine. 1997, 3, 379-385. 116. Li, J.X., Kadota,S., Li, H.Y., Miyahara, T., Namba, T., The effect of traditional medicines on bone resorption induced by parathyroid hormone (PTH) in tissue culture: a detailed study on cimicifuga rhizome. J Trad Med. 1996, 13, 50-8. 117. Johnson, E.B., Muto, M.G., Yanushpolsky, E.H. and Mutter, G.L. Phytoestrogen supplementation and endometrial cancer. Obstet Gynecol. 2001, 98, 947-50. 118. Eagon, C.L., Elm, M.S., Teepe, A.G., Eagon, P.K. Medicinal Botanicals: Estrogenicity in rat uterus and liver. Proceeding of the American Association for Cancer Research. 1997, 38, 293. 119. Einer-Jensen, N., Zhao, J., Andersen, K.P. and Kristoffersen, K. Cimicifuga and Melbrosia lack oestrogenic effects in mice and rats. Maturitas. 1996, 25, 149-53. 120. Liu, Z., Yang, Z., Zhu, M. and Huo, J. [Estrogenicity of black cohosh (Cimicifuga racemosa) and its effect on estrogen receptor level in human breast cancer MCF-7 cells]. Wei Sheng Yan Jiu. 2001, 30, 77-80. 121. Burdette, J.E., Liu, J., Chen, S.N., Fabricant, D.S., Piersen, C.E., Barker, E.L., Pezzuto, J.M., Mesecar, A., Van Breemen, R.B., Farnsworth, N.R., and Bolton, J.L. Black cohosh acts as a mixed competitive ligand and partial agonist of the serotonin receptor. J Agric Food Chem. 2003, 51, 5661-70. 122. Payne, J., Jones, C., Lakhani, S. and Kortenkamp, A. Improving the reproducibility of the MCF-7 cell proliferation assay for the detection of xenoestrogens. Sci Total Environ. 2000, 248, 51-62. 123. Roy, D. and Liehr, J.G. Estrogen, DNA damage and mutations. Mutat Res. 1999, 424, 107-15. 124. Bruggisser, R., Daeniken, K.V., Jundt, G., Schaffner, W. and Tullberg- Reinert, H. Interference of plant extracts, phytoestrogens and antioxidants with the MTT tetrazolium assay. Planta Med. 2002, 68, 445-8.

165 125. Zierau, O., Bodinet, C., Kolba, S., Wulf, M. and Vollmer, G. Antiestrogenic activities of Cimicifuga racemosa extracts. J Steroid Biochem Mol Biol. 2002, 80, 125-30. 126. Lohning, A., Winterhoff, H. Neurotransmitter concentrations after three weeks treatment with Cimicifuga racemosa[abstract]. Phytomedicine. 2000, 7, 13. 127. Borrelli, F., Izzo, A.A. and Ernst, E. Pharmacological effects of Cimicifuga racemosa. Life Sci. 2003, 73, 1215-29. 128. Burdette, J.E., Chen, S.N., Lu, Z.Z., Xu, H., White, B.E., Fabricant, D.S., Liu, J., Fong, H.H., Farnsworth, N.R., Constantinou, A.I., Van Breemen, R.B., Pezzuto, J.M., and Bolton, J.L. Black cohosh (Cimicifuga racemosa L.) protects against menadione-induced DNA damage through scavenging of reactive oxygen species: bioassay-directed isolation and characterization of active principles. J Agric Food Chem. 2002, 50, 7022- 8. 129. Johnson, B.M. and van Breemen, R.B. In vitro formation of quinoid metabolites of the dietary supplement Cimicifuga racemosa (black cohosh). Chem Res Toxicol. 2003, 16, 838-46. 130. Liang, W., Metabolism and pharmacokinetics of active compounds of botanical dietary supplements., in Department of Medicinal Chemistry and Pharmacognosy. 2004, University of Illinois at Chicago: Chicago. 131. Woo, K.C., Park, Y.S., Jun, D.J., Lim, J.O., Baek, W.Y., Suh, B.S. and Kim, K.T. Phytoestrogen cimicifugoside-mediated inhibition of catecholamine secretion by blocking nicotinic acetylcholine receptor in bovine adrenal chromaffin cells. J Pharmacol Exp Ther. 2004, 309, 641-9. 132. Loub, W.D., Farnsworth, N.R., Soejarto, D.D. and Quinn, M.L. NAPRALERT: computer handling of natural product research data. J Chem Inf Comput Sci. 1985, 25, 99-103. 133. Corley, D.G. and Durley, R.C. Strategies for database dereplication of natural products. J Nat Prod. 1994, 57, 1484-90. 134. Wende, K., Mugge, C., Thurow, K., Schopke, T. and Lindequist, U. Actaeaepoxide 3-O-beta-D-xylopyranoside, a new cycloartane glycoside from the rhizomes of Actaea racemosa (Cimicifuga racemosa). J Nat Prod. 2001, 64, 986-9. 135. Linde, H. [Contents of Cimicifuga racemosa. 2. On the structure of actein]. Arch Pharm Ber Dtsch Pharm Ges. 1967, 300, 885-92. 136. Shao, Y., Harris, A., Wang, M., Zhang, H., Cordell, G.A., Bowman, M. and Lemmo, E. Triterpene glycosides from Cimicifuga racemosa. J Nat Prod. 2000, 63, 905-10. 137. Chen, S.N., Li, W., Fabricant, D.S., Santarsiero, B.D., Mesecar, A., Fitzloff, J.F., Fong, H.H. and Farnsworth, N.R. Isolation, structure elucidation, and absolute configuration of 26-deoxyactein from Cimicifuga racemosa and clarification of nomenclature associated with 27- deoxyactein. J Nat Prod. 2002, 65, 601-5.

166 138. Chen, S.N., Fabricant, D.S., Lu, Z.Z., Fong, H.H. and Farnsworth, N.R. Cimiracemosides I-P, new 9,19-cyclolanostane triterpene glycosides from Cimicifuga racemosa. J Nat Prod. 2002, 65, 1391-7. 139. He, K., Zheng, B., Kim, C.H., Rogers, L. and Zheng, Q. Direct analysis and identification of triterpene glycosides by LC/MS in black cohosh, Cimicifuga racemosa, and in several commercially available black cohosh products. Planta Med. 2000, 66, 635-40. 140. Linde, H. [Contents of Cimicifuga racemosa. 5. 27-desoxyacetylacteol]. Arch Pharm Ber Dtsch Pharm Ges. 1968, 301, 335-41. 141. Shibata, M., Sakurai, N. and Onoda, M. [Pharmacological studies on the Chinese crude drug "Shoma". II. Anti-inflammatory action of Cimicifuga rhizoma, Cimicifuga simplex Wormsk (author's transl)]. Yakugaku Zasshi. 1977, 97, 911-5. 142. Chen, S.N., Fabricant, D.S., Lu, Z.Z., Zhang, H., Fong, H.H. and Farnsworth, N.R. Cimiracemates A-D, phenylpropanoid esters from the rhizomes of Cimicifuga racemosa. Phytochemistry. 2002, 61, 409-13. 143. Kruse, S.O., Lohning, A., Pauli, G.F., Winterhoff, H. and Nahrstedt, A. Fukiic and piscidic acid esters from the rhizome of Cimicifuga racemosa and the in vitro estrogenic activity of fukinolic acid. Planta Med. 1999, 65, 763-4. 144. Li, W., Sun, Y., Liang, W., Fitzloff, J.F. and van Breemen, R.B. Identification of caffeic acid derivatives in Actea racemosa (Cimicifuga racemosa, black cohosh) by liquid chromatography/tandem mass spectrometry. Rapid Commun Mass Spectrom. 2003, 17, 978-82. 145. Kusano, A., Seyama, Y., Nagai, M., Shibano, M. and Kusano, G. Effects of fukinolic acid and cimicifugic acids from Cimicifuga species on collagenolytic activity. Biol Pharm Bull. 2001, 24, 1198-201. 146. Watanabe, K., Mimaki, Y., Sakagami, H. and Sashida, Y. Cycloartane glycosides from the rhizomes of Cimicifuga racemosa and their cytotoxic activities. Chem Pharm Bull (Tokyo). 2002, 50, 121-5. 147. Bedir, E. and Khan, I.A. Cimiracemoside a: A new cyclolanostanol xyloside from the rhizome of Cimicifuga racemosa. Chem Pharm Bull (Tokyo). 2000, 48, 425-7. 148. Bedir, E. and Khan, I.A. A new cyclolanostanol arabinoside from the rhizome of Cimicifuga racemosa. Pharmazie. 2001, 56, 268-9. 149. Willaman, J.J. and Li, H.L. Alkaloid-bearing Plants and their contained alkaloids, 1957-1968. Lloydia. 1970, 33S, 1-286. 150. Loser, B., Kruse, S.O., Melzig, M.F. and Nahrstedt, A. Inhibition of neutrophil elastase activity by cinnamic acid derivatives from Cimicifuga racemosa. Planta Med. 2000, 66, 751-3. 151. Struck, D., Tegtmeier, M., Harnischfeger, G. Flavones in extracts of C. racemosa. Planta Med. 1997, 63, 289. 152. MATTHIAS, H., CHRISTIAN, W. and BJORN, B. CYCLOARTANE GLYCOSIDES FROM CIMICIFUGA RACEMOSA. PHARM PHARMACOL LETT. 2001, 11, 15-7.

167 153. Linde, H. [Contents of Cimicifuga racemosa. 3. On the constitution of the rings A, B and C of actein]. Arch Pharm Ber Dtsch Pharm Ges. 1967, 300, 982-92. 154. Linde, H. [the Composition of Cimicifuga Racemosa. 1. On the Isolation of 2 Glycosides]. Arzneimittelforschung. 1964, 14, 1037-40. 155. Kusano, G. and Takemoto, T. [Studies on the constituents of Cimicifuga spp. IX. Structure of Cimigol (author's transl)]. Yakugaku Zasshi. 1975, 95, 1133-7. 156. Kusano, G., Uchida, H., Murakami, Y., Sakurai, N. and Takemoto, T. [Studies on the constituents of Cimicifuga spp. XII. A revised structure of acerinol and the structures of the related compounds (author's transl)]. Yakugaku Zasshi. 1976, 96, 321-5. 157. Kusano, G., Murakami, Y., Sakurai, N. and Takemoto, T. [Studies on the constituents of Cimicifuga spp. XI. Isolation and stereostructures of dahurinol, dehydroxydahurinol, isodahurinol and 25-o-methylisodahurinol (author's transl)]. Yakugaku Zasshi. 1976, 96, 82-5. 158. Kusano, A., Shibano, M., Kusano, G. and Miyase, T. Studies on the constituents of Cimicifuga species. XIX. Eight new glycosides from Cimicifuga simplex Wormsk. Chem Pharm Bull (Tokyo). 1996, 44, 2078- 85. 159. Kusano, G. [Studies on the constituents of Cimicifuga species]. Yakugaku Zasshi. 2001, 121, 497-521. 160. Kusano, A., Shibano, M., Tsukamoto, D. and Kusano, G. Studies on the constituents of Cimicifuga species. XXVIII. Cycloart-7-enol glycosides from the underground parts of Cimicifuga simplex Wormsk. Chem Pharm Bull (Tokyo). 2001, 49, 437-41. 161. Sakurai, N., Inoue, T. and Nagai, M. [Studies on the Chinese crude drug "Shoma." II. Triterpenes of Cimicifuga dahurica Maxim]. Yakugaku Zasshi. 1972, 92, 724-8. 162. Sakurai, N., Nagai, M. and Inoue, T. [Studies on the Chinese crude drug "Shoma". III. Triterpene glycosides from Cimicifuga japonica and structure of 25-anhydrocimigenol arabinoside (author's transl)]. Yakugaku Zasshi. 1975, 95, 1354-60. 163. Sakurai, N., Koeda, M., Inoue, T. and Nagai, M. Studies on the Chinese crude drug "Shoma." VIII. Two new triterpenol bisdesmosides, 3- arabinosyl-24-O-acetylhydroshengmanol 15-glucoside and 3-xylosyl-24-O- acetylhydroshengmanol 15-glucoside, from Cimicifuga dahurica. Chem Pharm Bull (Tokyo). 1994, 42, 48-51. 164. Takemoto, T., Kusano, G. and Yamamoto, N. [Studies on the constituents of Cimicifuga spp. I. Constituents of Cimicifuga acerina. (1)]. Yakugaku Zasshi. 1966, 86, 1006-10. 165. Takemoto, T. and Kusano, G. [Studies on the costituents of Cimicifuga spp. II. Constituents of Cimicifuga acerina. 2. Structure of cimigenol]. Yakugaku Zasshi. 1967, 87, 1569-72.

168 166. Takemoto, T., Kusano, G. and Yamamoto, N. [Studies on the constituents of Cimicfuga spp. 3. Constituents of Cimicifuga acerian. 3. Structure of acerinol]. Yakugaku Zasshi. 1967, 87, 1489-92. 167. Takemoto, T. and Kusano, G. [Studies on the constituents of cimicifuga spp. IV. Constituents of Cimicifuga acerina. (4). Structure of methyl cimigenol]. Yakugaku Zasshi. 1968, 88, 623-6. 168. Takemoto, T. and Kusano, G. [Studies on the constituents of Cimicifuga spp. V. Constituents of Cimicifuga acerina. (5). Structures of 25-O- acetylcimigenol, dehydroxy-15-O-methylcimigenol and 15-O- methylcimigenol]. Yakugaku Zasshi. 1969, 89, 954-8. 169. Takemoto, T., Kusano, G. and Yamamoto, N. [Studies on the constituents of Cimicifuga spp. VII. Structure of cimicifugenol]. Yakugaku Zasshi. 1970, 90, 68-72. 170. Takemoto, T., Kusano, G. and Kawahara, M. [Studies on the constituents of Cimicifuga spp. VI. Structures of 25-O-acetylcimigenoside and 25-O- methylcimigenoside]. Yakugaku Zasshi. 1970, 90, 64-7. 171. Zhang, Q.W., Ye, W.C., Che, C.T. and Zhao, S.X. A new cycloartane saponin from Cimicifuga acerina. J Asian Nat Prod Res. 1999, 2, 45-9. 172. Zhang, Q.W., Ye, W.C., Hsiao, W.W., Zhao, S.X. and Che, C.T. Cycloartane glycosides from Cimicifuga dahurica. Chem Pharm Bull (Tokyo). 2001, 49, 1468-70. 173. Zhu, N., Jiang, Y., Wang, M. and Ho, C.T. Cycloartane triterpene saponins from the roots of Cimicifuga foetida. J Nat Prod. 2001, 64, 627-9. 174. Panossian, A., Danielyan, A., Mamikonyan, G. and Wikman, G. Methods of phytochemical standardisation of rhizoma Cimicifugae racemosae. Phytochem Anal. 2004, 15, 100-8. 175. Onorato, J. and Henion, J.D. Evaluation of triterpene glycoside estrogenic activity using LC/MS and immunoaffinity extraction. Anal Chem. 2001, 73, 4704-10. 176. Takahira, M., Kusano, A., Shibano, M., Kusano, G., Koizumi, K., Suzuki, R., Kim, H.S. and Wataya, Y. Antimalarial activity and nucleoside transport inhibitory activity of the triterpenic constituents of Cimicifuga spp. Biol Pharm Bull. 1998, 21, 823-8. 177. McCoy, J. and Kelly, W. Survey of Cimicifuga racemosa for phytoestrogenic flavonoids. in 212th ACS National Meeting. 1996. Orlando, FL, August 25-29: ACS. 178. Farnsworth, N.R., NAPRALERT Database. 2003, University of Illinois at Chicago: Chicago, Illinois. 179. Woldemariam, T.Z., Betz, J.M. and Houghton, P.J. Analysis of aporphine and quinolizidine alkaloids from Caulophyllum thalictroides by densitometry and HPLC. J Pharm Biomed Anal. 1997, 15, 839-43. 180. Ganzera, M., Dharmaratne, H.R., Nanayakkara, N.P. and Khan, I.A. Determination of saponins and alkaloids in Caulophyllum thalictroides (blue cohosh) by high-performance liquid chromatography and evaporative light scattering detection. Phytochem Anal. 2003, 14, 1-7.

169 181. Kennelly, E.J., Flynn, T.J., Mazzola, E.P., Roach, J.A., McCloud, T.G., Danford, D.E. and Betz, J.M. Detecting potential teratogenic alkaloids from blue cohosh rhizomes using an in vitro rat embryo culture. J Nat Prod. 1999, 62, 1385-9. 182. Baillie, N. and Rasmussen, P. Black and blue cohosh in labour. N Z Med J. 1997, 110, 20-1. 183. Chan, G.M. and Nelson, L.S. More on blue cohosh and perinatal stroke. N Engl J Med. 2004, 351, 2239-41; author reply 2239-41. 184. Edmunds, J. Blue cohosh and newborn myocardial infarction? Midwifery Today Int Midwife. 1999, 34-5. 185. Finkel, R.S. and Zarlengo, K.M. Blue cohosh and perinatal stroke. N Engl J Med. 2004, 351, 302-3. 186. Jones, T.K. and Lawson, B.M. Profound neonatal congestive heart failure caused by maternal consumption of blue cohosh herbal medication. J Pediatr. 1998, 132, 550-2. 187. Rao, R.B. and Hoffman, R.S. Nicotinic toxicity from tincture of blue cohosh (Caulophyllum thalictroides) used as an abortifacient. Vet Hum Toxicol. 2002, 44, 221-2. 188. Potterton, D. More on blue cohosh and perinatal stroke. N Engl J Med. 2004, 351, 2239-41; author reply 2239-41. 189. Wright, I.M. Neonatal effects of maternal consumption of blue cohosh. J Pediatr. 1999, 134, 384-5. 190. Gunn, T.R. and Wright, I.M. The use of black and blue cohosh in labour. N Z Med J. 1996, 109, 410-1. 191. Jackson, B.P. and Snowdon, D.W., Powdered vegetable drugs; an atlas of microscopy for use in the identification and authentication of some plant materials employed as medicinal agents. 1969, New York,: American Elsevier Pub. Co. viii, 203. 192. Black Cohosh-Standards of analysis, quality control and therapeutics, in American Herbal Pharmacopoeia and Therapeutic Compendium, R. Upton, Editor. 2002, AHP: Santa Cruz. p. 37. 193. United States Pharmacopoeia and National Formulary (USP 28- NF 23). in United States Pharmacopeia Convention. 2004. Rockville, MD. 194. Lunn, G. and Schmuff, N.R., HPLC methods for pharmaceutical analysis. 1997, New York: Wiley. 4 v. 195. Neue, U.D., HPLC columns : theory, technology, and practice. 1997, New York: Wiley-VCH. xvi, 393. 196. Burdette, J.E., Chen, S.N., Lu, Z.Z., Xu, H., White, B.E., Fabricant, D.S., Liu, J., Fong, H.H., Farnsworth, N.R., Constantinou, A.I., Van Breemen, R.B., Pezzuto, J.M., and Bolton, J.L. Black Cohosh (Cimicifuga racemosa L.) Protects against Menadione-Induced DNA Damage through Scavenging of Reactive Oxygen Species: Bioassay-Directed Isolation and Characterization of Active Principles. J Agric Food Chem. 2002, 50, 7022- 7028. 197. Xu, H., Fabricant, D.S., Piersen, C.E., Bolton, J.L., Pezzuto, J.M., Fong, H., Totura, S., Farnsworth, N.R. and Constantinou, A.I. A preliminary

170 RAPD-PCR analysis of Cimicifuga species and other botanicals used for women's health. Phytomedicine. 2002, 9, 757-62. 198. Upton, R., Black Cohosh Adulteration, N.R. Farnsworth, Editor. 2002. 199. AHPA Code of Ethics. 2005, AHPA: Silver Spring. p. 5. 200. Fu, R.Z., Wang, J., Sun, Y.R. and Shaw, P.C. Extraction of genomic DNA suitable for PCR analysis from dried plant rhizomes/roots. Biotechniques. 1998, 25, 796-8, 800-1. 201. Burdette, J.E., Estrogenic activity of Botanical Extracts, in Department of Medicinal Chemistry and Pharmacognosy. 2002, University of Illinois at Chicago: Chicago. 202. Sarni-Manchado, P. and Cheynier, V. Study of non-covalent complexation between catechin derivatives and peptides by electrospray ionization mass spectrometry. J Mass Spectrom. 2002, 37, 609-16. 203. Schmuck, C. Self-folding molecules: a well defined, stable loop formed by a carboxylate-guanidinium zwitterion in DMSO. J Org Chem. 2000, 65, 2432-7. 204. Chang, L., Whittaker, N.F. and Bewley, C.A. Crambescidin 826 and dehydrocrambine A: new polycyclic guanidine alkaloids from the marine sponge Monanchora sp. that inhibit HIV-1 fusion. J Nat Prod. 2003, 66, 1490-4. 205. Cummins, D.J., Andrews, C.W., Bentley, J.A. and Cory, M. Molecular diversity in chemical databases: comparison of medicinal chemistry knowledge bases and databases of commercially available compounds. J Chem Inf Comput Sci. 1996, 36, 750-63. 206. Berlinck, R.G. Natural guanidine derivatives. Nat Prod Rep. 1996, 13, 377- 409. 207. Berlinck, R.G. Natural guanidine derivatives. Nat Prod Rep. 1999, 16, 339- 365. 208. Berlinck, R.G. Natural guanidine derivatives. Nat Prod Rep. 2002, 19, 617- 49. 209. Stromeier, S., Phytochemische Untersuchungen und Prüfung auf estrogene Aktivität von phenolischen Inhaltsstoffen aus Cimicifuga racemosa (L.) Nutt. (syn. Actaea racemosa L.), in Pharmazeutische Biologie und Phytochemie. 2003: Münster. p. 198. 210. Fabricant, D., Li, W., Chen, S.N., Graham, J., Fitzloff, J., Fong, H.H.S., Farnsworth, N.R. Geographical and diurnal variations of chemical constituents of C. racemosa (L.) Nutt. in ASP & CRN Botanical Dietary Supplements at a Crossroads. 2001. Asilomar CA. 211. Joint FAO/WHO Codex Alimentarius Commission., Joint FAO/WHO Food Standards Programme., Food and Agriculture Organization of the United Nations. and World Health Organization., Codex alimentarius. 2004, Rome: Food and Agriculture Organization of the United Nations : World Health Organization. v, 51. 212. World Health Organization., WHO monographs on selected medicinal plants. 1999, Geneva: World Health Organization. v. <1-2 >.

171 213. Cao, P., Pu, X.F., Peng, S.L., Zhang, X.R. and Ding, L.S. Chemical constituents from Cimicifuga foetida. J Asian Nat Prod Res. 2005, 7, 145- 9. 214. Chen, S.N., Fabricant, D.S., Pauli, G.F., Fong, H.H. and Farnsworth, N.R. Synthesis of cimiracemate B, a phenylpropanoid found in Cimicifuga racemosa. Nat Prod Res. 2005, 19, 287-90. 215. Ju, J. and Yang, J. [Advance in the research on triterpenoids of tribe Cimicifugeae]. Zhongguo Zhong Yao Za Zhi. 1999, 24, 517-21, 572. 216. Kimura, O., Sakurai, N., Nagai, M. and Inoue, T. [Studies on the Chinese crude drug "Shoma." VI. Shengmanol xyloside, a new genuine natural product of some Cimicifuga glycosides]. Yakugaku Zasshi. 1982, 102, 538-45. 217. Kimura, O., Sakurai, N. and Inoue, T. [Studies on the Chinese crude drug "Shoma." VII. Isolation and determination of genuine natural products, acetyl shengmanol xyloside, 24-O-acetylhydroshengmanol xyloside, and shengmanol xyloside, in Cimicifuga dahurica and the other Cimicifuga plants]. Yakugaku Zasshi. 1983, 103, 293-9. 218. Koeda, M., Aoki, Y., Sakurai, N. and Nagai, M. Studies on the Chinese crude drug "shoma." IX. Three novel cyclolanostanol xylosides, cimicifugosides H-1, H-2 and H-5, from cimicifuga rhizome. Chem Pharm Bull (Tokyo). 1995, 43, 771-6. 219. Li, C.J., Chen, D.H. and Xiao, P.G. [Chemical constituents of traditional Chinese drug "sheng-ma" (Cimicifuga dahurica)]. Yao Xue Xue Bao. 1993, 28, 777-81. 220. Liu, Y., Chen, D., Si, J., Tu, G. and An, D. Cimidahuside 1 and 2, two new cyclolanostanol xylosides from the aerial parts of Cimicifuga dahurica. Nat Prod Res. 2003, 17, 243-6. 221. Liu, Y., Chen, D., Si, J., Tu, G. and An, D. Two new cyclolanostanol xylosides from the aerial parts of Cimicifuga dahurica. J Nat Prod. 2002, 65, 1486-8. 222. Nishida, M., Yoshimitsu, H., Okawa, M., Ikeda, T. and Nohara, T. Two new 15,16-seco-cycloartane glycosides from Cimicifuga rhizome. Chem Pharm Bull (Tokyo). 2003, 51, 1215-6. 223. Nishida, M., Yoshimitsu, H. and Nohara, T. Two new tetranor-cycloartane glycosides from cimicifuga rhizome. Chem Pharm Bull (Tokyo). 2003, 51, 1117-8. 224. Nishida, M., Yoshimitsu, H. and Nohara, T. Three cycloartane glycosides from Cimicifuga rhizome and their immunosuppressive activities in mouse allogeneic mixed lymphocyte reaction. Chem Pharm Bull (Tokyo). 2003, 51, 354-6. 225. Sakurai, N. and Nagai, M. [Chemical constituents of original plants of Cimicifugae rhizoma in Chinese medicine]. Yakugaku Zasshi. 1996, 116, 850-65. 226. Zhang, Q.W., Ye, W.C., Che, C.T. and Zhao, S.X. [Cycloartane triterpenes and glycosides from Cimicifuga acerina]. Yao Xue Xue Bao. 2001, 36, 287-91.

172 227. Zhao, X.H., Chen, D.H., Si, J.Y., Pan, R.L. and Shen, L.G. [Studies on the phenolic acid constituents from Chinese medicine "sheng-ma", rhizome of Cimicifuga foetida L.]. Yao Xue Xue Bao. 2002, 37, 535-8. 228. Zhao, X.H., Chen, D.H., Si, J.Y., Pan, R.L., Shen, L.G. and Chen, D. [Studies on new trierpenoid constituents from the Rhizoma of Cimicifuga foetida]. Zhongguo Zhong Yao Za Zhi. 2003, 28, 135-8. 229. Eldridge, G.R., Vervoort, H.C., Lee, C.M., Cremin, P.A., Williams, C.T., Hart, S.M., Goering, M.G., O'Neil-Johnson, M. and Zeng, L. High- throughput method for the production and analysis of large natural product libraries for drug discovery. Anal Chem. 2002, 74, 3963-71. 230. Molgaard, P., Johnsen, S., Christensen, P. and Cornett, C. HPLC method validated for the simultaneous analysis of cichoric acid and alkamides in Echinacea purpurea plants and products. J Agric Food Chem. 2003, 51, 6922-33. 231. Charlton, A.J., Baxter, N.J., Khan, M.L., Moir, A.J., Haslam, E., Davies, A.P. and Williamson, M.P. Polyphenol/peptide binding and precipitation. J Agric Food Chem. 2002, 50, 1593-601.

173

10.0 Appendices

10.1 Additional Structural Data for Novel Center Isolates.

Structural data of 14 Cimicifuga racemosa novel constituents from silica

fractions (pages 83, 84). Proton NMR data given as; chemical shift (multiplicity, J

value(s) where applicable in Hz, proton position). Carbon NMR data given as;

chemical shift (multiplicity, carbon location).

Cimiracemate A: light brown powder, mp 94–96 °C. 1H NMR spectral data (500

MHz, CD3OD): δ 6.36 (d, 15.9, H-2), 7.61 (d, 15.9, H-3), 7.09 (d, 2.1, H-5a), 6.94 (d, 8.3, H-8a), 7.06 (dd, 2.1, 8.3, H-9a), 4.86 (s, H-1’), 3.63 (s, H-3’), 6.68 (d, 2.0, H-5b), 6.73 (d, 8.0, H-8b), 6.57 (dd, 2.0, 8.0, H-9b), 3.88 (s, MeO-7); 13C NMR

spectra data (125 MHz, CD3OD): δ 168.1 (s, C-1), 115.2 (d, C-2), 147.4 (d, C-3), 128.8 (s, C-4), 114.8 (d, C-5) 148.0 (s, C-6), 151.7 (s, C-7), 112.5 (d, C-8), 123.0 (d, C-9), 68.5 (t, C-1’), 204.5 (s, C-2’), 46.3 (t, C-3’), 126.0 (s, C-4’), 117.6 (d, C- 5’), 146.6 (s, C-6’), 145.7 (s, C-7’), 116.5 (d, C-8’), 122.0 (d, C-9’), 56.4 (q, MeO-

7). UV λmaxMeOH nm (log ): 210.5 (4.26), 291.0 (3.77), 325.0 (3.96). IR νmax NaCl cm−1: 3403, 2940, 1705, 1608, 1512, 1442, 1264, 1162, 1131, 1023.Negative ESI–MS m/z (relative intensity %): 357 (18)[M]−, 193 (100), 178 (20), 163 (90),

149 (25), 134 (48); HR-ESIMS m/z 357.0974 (calc. 357.0974 for C19 H17O7).

Cimiracemate B: light brown powder, mp 86–88.5 °C. 1H NMR spectral data

(500 MHz, CD3OD): δ 6.42 (d, 16.0, H-2), 7.65 (d, 16.0, H-3), 7.21 (d, 1.9, H-5), 6.83 (d, 8.3, H-8), 7.09 (dd, 1.9, 8.2, H-9), 4.86 (s, H-1′), 3.62 (s, H-3′), 6.68 (d, 2.0, H-5′), 6.73 (d, 8.0, H-8′), 6.57 (dd, 2.0, 8.0, H-9′), 3.89 (s, MeO-6); 13C NMR

spectra data (125 MHz, CD3OD): δ 168.3 (s, C-1), 114.5 (d, C-2), 147.4 (d, C-3), 127.6 (s, C-4), 116.5 (d, C-5), 150.9 (s, C-6), 149.4 (s, C-7), 111.7 (d, C-8), 124.3 (d, C-9), 68.5 (t, C-1′), 204.6 (s, C-2′), 46.3 (t, C-3′), 126.0 (s, C-4′), 117.6 (d, C- 5′), 146.6 (s, C-6′), 145.7 (s, C-7′), 116.5 (d, C-8′), 122.0 (d, C-9′), 56.4 (q, MeO- −1 6). UV λmax MeOH nm (log ): 208.5 (4.28), 327.5 (3.92). IR νmax NaCl (cm ):

174

3396, 2930, 1708, 1600, 1519, 1448, 1217, 1172, 1125, 1030. Negative ESI–MS m/z (relative intensity %): 357 (23)[M]−, 313 (10%), 193 (100), 179 (45), 163 (70),

149 (70), 134 (68), HR–ESIMS m/z 357.0974 [M]− (calc. 357.0974 for C19

H17O7).

Cimiracemate C: light brown powder, mp 88–90 °C. 1H NMR spectral data (500

MHz, CD3OD): δ 6.35 (d, 15.9, H-2), 7.59 (d, 16.0, H-3), 7.07 (d, 2.0, H-5), 6.94 (d, 8.4, H-8), 7.06 (dd, 2.0, 8.4, H-9), 4.94 (d, 17.5, H-1′a), 5.00 (d, 17.5, H-1′b), 4.79 (s, H-3′), 6.78 (d, 2.0, H-5′), 6.79 (d, 8.1, H-8′), 6.72 (dd, 2.0, 8.1, H-9′), 3.89 13 (s, MeO-7), 3.33 (s, MeO-3′). C NMR spectra data (125 MHz, CD3OD): δ 168.1 (s, C-1), 115.2 (d, C-2), 147.4 (d, C-3), 128.8 (s, C-4), 114.8 (d, C-5) 148.1 (s, C- 6), 151.7 (s, C-7), 112.5 (d, C-8), 123.0 (d, C-9), 66.9 (t, C-1′), 203.6 (s, C-2′), 88.2 (d, C-3′), 127.9 (s, C-4′), 115.5 (d, C-5′), 147.4 (s, C-6′), 147.0 (s, C-7′),

116.5 (d, C-8′), 120.7 (d, C-9′), 56.4 (q, MeO-7), 57.1 (q, MeO-3′). UV λmax MeOH −1 nm (log): 212.0 (4.35), 292.9 (3.87), 326.0 (4.09). IR νmax NaCl (cm ): 3399, 2927, 1710, 1608, 1515, 1442, 1270, 1156. Negative ESI–MS m/z (relative intensity%): 387 (8)[M]−, 355 (33), 340 (10), 296 (30), 193 (100), 178 (45), 161

(38), 149 (27), 134 (37). HR–ESIMS m/z 387.1073 [M]− (calc. 387.1080 C20

H19O8).

Cimiracemate D: light brown powder, mp 100–102 °C. 1H NMR spectral data

(500 MHz, CD3OD): δ 6.40 (d, 15.9, H-2), 7.63 (d, 16.0, H-3), 7.19 (d, 1.9, H-5), 6.94 (d, 8.1, H-8), 7.06 (dd, 1.9, 8.1, H-9), 4.94 (d, 17.5, H-1′a), 5.00 (d, 17.5, H- 1′b), 4.79 (s, H-3′), 6.79 (d, 1.9, H-5), 6.80 (d, 7.9, H-8′), 6.72 (dd, 1.9, 7.9, H-9′), 13 3.88 (s, MeO-6), 3.35 (s, MeO-3′). C NMR spectra data (125 MHz, CD3OD): δ 168.5 (s, C-1), 114.5 (d, C-2), 147.8 (d, C-3), 127.6 (s, C-4), 116.5 (d, C-5), 150.8 (s, C-6), 149.4 (s, C-7), 111.8 (d, C-8), 124.4 (d, C-9), 66.9 (t, C-1′), 203.7 (s, C-2′), 88.2 (d, C-3′), 127.9 (s, C-4′), 115.5 (d, C-5′), 147.4 (s, C-6′), 145.0 (s,

C-7′), 116.6 (d, C-8′), 120.0 (d, C-9′), 56.5 (q, MeO-6), 57.2 (q, MeO-3′). UV λmax −1 MeOH nm (log): 212.0 (4.35), 326.0 (4.09). IR νmax NaCl (cm ): 3387, 2979, 1710, 1601, 1520, 1481, 1271, 1156, 1029. Negative ESI–MS m/z (relative

175

intensity %): 387 (12)[M]−, 355 (22), 337 (22), 296 (22), 193 (95), 178 (100), 175

(52), 161(40), 149 (20), 134 (40), HR–ESIMS m/z 387.1084 (calc. 387.1080 C20

H19O8).

Cimiracemoside I: pale yellow powder, mp >300 °C (began decomposing at 250 20 1 °C), [α] D -13.13 (c 0.267, CHCl3); H-NMR spectral data (500 MHz, pyridine-

d5): δ 5.08 (d, 7.2, H-7), 4.87 (d, 7.4, H-1'), 4.38 (dd, 4.5, 10.5, H-5'), 4.31 (dd, 6.7, 12.0, H-16), 4.24 (dd, 6.0, 9.0,H-4'), 4.18 (t, 6.9, H-3'), 4.05 (overlapped, H- 2'), 4.04 (d, 10.2, H-26), 3.76 (t, 10.3, H-5'), 3.70 (s,H-24), 3.61 (d, 10.2, H-26), 3.48 (brd, 7.5, H-3), 2.33 (m, overlapped, H-2), 2.26 (overlapped, H-20), 2.11 (brt, 8.6, H-15), 2.09 (m, H-11a),1.96 (dd, 6.3, 11.4, H-15), 1.80 (m, H-6), 1.70 (m, H-1), 1.66 (2H, m, H-12), 1.58 (brd. 13.5, H-22a), 1.56 (t, 13.5, H-17), 1.46 (s, H-27), 1.46 (m, H-6), 1.40 (brt, 14.7, H-22b), 1.35 (s, H-29), 1.30 (m, overlapped, H-2), 1.30 (m, H-1), 1.26 (s, H-18), 1.24 (overlapped, H-5), 1.11 (m, H-11b), 1.10 (s, H-28), 1.04 (s, H-30), 1.00 (d, 6.4, H-21), 0.97 (d, 3.5, H-19), 13 0.46 (d, 3.5, H-20), C-NMR spectral data (500 MHz, pyridine-d5) 149.2 (s, C-8), 113.5 (d, C-7), 107.6 (d, C-1'), 106.2 (s, C-23), 88.1 (d, C-3), 78.7 (d, C-3'), 75.6 (d, C-2'), 74.9 (d, C-16), 71.3 (d, C-4'), 68.0 (t, C-26), 67.2 (t, C-5'), 62.6 (d, C- 24), 62.1 (s, C-25), 56.9 (d, C-17), 49.8 (s, C-14), 44.1 (s, C-13), 43.0 (t, C-15), 42.7 (d, C-5), 40.4 (s, C-4), 37.5 (t, C-22), 32.9 (t, C-12), 30.9 (t, C-1), 29.6 (t, C- 2), 28.3 (t, C-19), 26.9 (q, C-28), 25.8 (q, C-29), 25.3 (t, C-11), 23.7 (s, C-10), 23.7 (d, C-20), 22.9 (q, C-18), 21.8 (t, C-6), 21.0 (s, C-9), 20.8 (q, C-21), 14.3 (q, −1 C-27), 14.3 (q, C-30), IR νmaxNaCl (cm ): 3485, 3409, 2923, 1454, 1379, 1038,

966; HRESIMS m/z 623.3581 (calc. 623.3560 for C35H52O8Na).

20 Cimiracemoside J: pale yellow powder, mp 138-140 °C, [α] D -14.23 (c 0.260, 1 CHCl3); H-NMR spectral data (500 MHz, pyridine-d5): δ 5.36 (brs, H-26), 5.27 (brd, 7.8 H-12), 4.89 (brs, H-), 4.79 (d, 6.8, H-1'), 4.44 (t, 7.8, H-2'), 4.42 (s, H- 15), 4.32 (overlapped, H-4'), 4.31 (overlapped, H-5'), 4.31 (overlap, H-23), 4.17 (overlapped, H-3'), 4.12 (brs, H-24), 3.79 (brd, 11.3, H-), 3.49 (dd, 3.1, 7.9, H-3),

176

2.94 (dd, 9.6, 15.4, H-22a), 2.32 (m, H-2), 2.29 (m, H-11), 2.22 (m, H-7), 2.13 (s, OAc), 1.85 (s, H-27), 1.77 (brd,12.4, H-5), 1.65 (overlapped, H-17), 1.64 (overlapped, H-20), 1.58 (brd. 12.2, H-1), 1.56 (m, H-6), 1.37 (s, H-18), 1.33 (m, H-2), 1.29 (overlapped, H-8), 1.28 (s, H-29), 1.20 (s, H-28), 1.17 (brd, 15.3, H- 22b), 1.12 (m, H-7), 1.11 (brd. 14.4, H-1), 1.03 (m, H-11), 1.01 (s, H-30), 0.94 (d, 6.5, H-21), 0.75 (m, H-6), 0.61 (d, 4, H-19), 0.32 (d, 4, H-19), 13C-NMR spectral data (500 MHz, pyridine-d5) δ 170.6 (s, OAc), 145.8 (s, C-25), 113.2 (t, C-26), 112.3 (s, C-16), 107.4 (d, C-1'), 88.3 (d, C-3), 86.5 (d, C-24), 79.3 (d, C-15), 77.3 (d, C-12), 74.7 (d, C-23), 74.7 (d, C-3'), 73.0 (d, C-2'), 69.5 (d, C-4'), 66.7 (t, C- 5'), 59.6 (d, C-17), 48.4 (s, C-13), 47.2 (d, C-5), 47.2 (d, C-8), 46.1 (s, C-14), 41.3 (s, C-4), 38.5 (t, C-11), 37.5 (t, C-22), 32.4 (t, C-1), 30.9 (t, C-19), 30.1 (t, C-2), 26.8 (s, C-10), 26.0 (t, C-7), 25.7 (q, C-29), 23.9 (d, C-20), 21.7 (q, O-Ac), 20.8 (t, C-6), 20.1 (s, C-9), 19.8 (q, C-21), 18.1 (q, C-27), 15.4 (q, C-30), 12.7 (q, C-18), −1 12.0 (q, C-28), IR νmaxNaCl (cm ): 3468, 2933, 2869, 1731, 1454, 1377, 1240,

1067, 755; HRESIMS m/z 683.3763 (calc 683.3771 for C37H56O10Na).

20 Cimiracemoside K: pale yellow powder, mp 142-143 ˚C, [α] D -59.32 (c 0.147, 1 CHCl3); H-NMR spectral data (500 MHz, pyridine-d5): δ 5.35 (brs, H-26), 5.28 (brd, 7.6, H-12), 4.89 (brs, H-26), 4.84 (d, 7.4, H-1'), 4.42 (s, H-15), 4.34 (dd, 4.8, 9.8, H-5'), 4.30 (d, 8.5, H-23), 4.21 (m, H-4'), 4.18 (brs, H-24), 4.15 (t, 8.4, H-3'), 4.03 (dd, 8.0, 15.4, H-2'), 3.72 (dd, 9.8, 9.1, H-5'), 3.49 (dd, 3.5, 10.8, H-3), 2.94 (dd, 9.5, 16.1, H-22), 2.30 (m, H-2), 2.26 (m, H-11), 2.19 (m, H-7), 2.12 (s, OAc), 1.90 (m, H-2), 1.85 (s, H-27), 1.76 (brd, 12.2, H-5), 1.64 (overlapped, H-17), 1.64 (overlapped, H-20), 1.56 (brt, 12.4, H-1), 1.53 (m, H-6), 1.32 (s, H-18), 1.31 (s, H- 29), 1.30 (overlapped, H-8), 1.20 (s, H-28), 1.16 (brd, 16.1, H-22), 1.10 (m, H-7), 1.08 (brd. 14.4, H-1), 1.04 (s, H-30), 1.02 (m, H-11), 0.95 (d, 6.5, H-21), 0.73 (m, H-6), 0.60 (d, 3.4, H-19), 0.32 (d, 3.4, H-19), 13C-NMR spectral data (500 MHz,

pyridine-d5) δ 170.6 (s, OAc), 145.8 (s, C-25), 113.2 (t, C-26), 112.3 (s, C-16), 107.5 (d, C-1'), 88.3 (d, C-3), 86.5 (d, C-24), 79.3 (d, C-15), 78.6 (d, C-3'), 77.3 (d, C-12), 75.6 (d, C-2'), 74.6 (d, C-23), 71.6 (d, C-4'), 67.1 (t, C-5'), 59.6 (d, C-

177

17), 48.4 (s, C-13), 47.2 (d, C-5), 47.2 (d, C-8), 46.1 (s, C-14), 41.3 (s, C-4), 38.5 (t, C-11), 37.5 (t, C-22), 32.4 (t, C-1), 30.9 (t, C-19), 30.0 (t, C-2), 26.8 (s, C-10), 26.0 (t, C-7), 25.7 (q, C-29), 23.9 (d, C-20), 21.7 (q, OAc), 20.8 (t, C-6), 20.1 (s, C-9), 19.8 (q, C-21), 18.1 (q, C-27), 15.4 (q, C-30), 12.7 (q, C-18), 12.0 (q, C-28), −1 IR νmaxNaCl (cm ): 3425, 2935, 2869, 1732, 1456, 1376, 1233, 1042, 760;

HRESIMS m/z 683.3745 (calc 683.3771 for C37H56O10Na).

20 Cimiracemoside L: white powder, mp 125-128 ˚C, [α] D -41.11 (c 0.450, 1 CHCl3); H-NMR spectral data (500 MHz, pyridine-d5): δ 5.61 (brs, H-4'), 5.42 (brt, 8.4, H-23), 4.82 (d, 7.1, H-1'), 4.48 (brt, 8.1, H-2'), 4.35 (s, H-15), 4.28 (d, 12.6, H-5'a), 4.21 (d, 7.3, H-3'), 3.85 (d, 12.6, H-5'b), 3.53 (dd, 4.3, 11.7, H-3), 3.05 (d, 8.4, H-24), 2.69 (brt 12.2, H-22a), 2.42 (m, H-2a), 2.35 (d, 6.5, H-17), 2.14 (overlapped, H-20), 2.12 (s, OAc), 2.10 (m, H-11a), 2.06 (s, OAc), 1.88 (dd, 4.4, 12.4, H-8), 1.80 (2H, (m, H-12), 1.77 (m, H-22b), 1.62 (overlapped, H-1a), 1.60 (m, H-6a), 1.40 (s, H-27), 1.39 (overlapped, H-5), 1.38 (s, H-18), 1.35 (m, H- 2b), 1.31 (s, H-29), 1.30 (overlapped, H-7a), 1.28 (overlapped, H-1b), 1.27 (d, 6.9, H-21), 1.26 (s, H-26), 1.22 (s, H-28), 1.15 (m, H-11b), 1.14 (overlapped, H- 7b), 1.06 (s, H-30), 0.76 (m, H-6b), 0.59 (d, 3.7, H-19a), 0.32 (d, 3.7, H-19b), 13C-

NMR spectral data (500 MHz, pyridine-d5) δ 220.0 (s, C-16), 170.9 (s, OAc), 170.7 (s, OAc), 107.6 (d, C-1'), 88.8 (d, C-3), 83.0 (d, C-15), 73.2 (d, C-2'), 72.6 (d, C-3'), 72.1 (d, C-23), 72.1 (d, C-4'), 65.2 (d, C-24), 64.4 (t, C-5'), 60.0 (d, C- 17), 58.6 (s, C-25), 48.3 (d, C-8), 47.5 (d, C-5), 46.1 (s, C-14), 41.6 (s, C-13), 41.4 (s, C-4), 37.0 (t, C-22), 33.1 (t, C-12), 32.2 (t, C-1), 30.5 (t, C-19), 30.1 (t, C- 2), 28.0 (d, C-20), 26.8 (s, C-10), 26.7 (t, C-7), 26.0 (t, C-11), 25.7 (q, C-29), 24.7 (q, C-26), 21.2 (q, OAc), 21.0 (t, C-6), 21.0 (q, OAc), 20.4 (q, C-21), 20.1 (s, C-9), −1 19.8 (q, C-18), 19.4 (q, C-27), 15.5 (q, C-30), 12.0 (q, C-28); IR νmaxNaCl (cm ): 3466, 2937, 2871, 1737, 1455, 1376, 1241, 1089, 757; HRESIMS m/z 705.4205

(calc 705.4214 for C39H61O11).

20 Cimiracemoside M: white powder, mp 107-109 °C, [α] D -19.00 (c 0.30, 1 CHCl3); H-NMR data (500 MHz, pyridine-d5) δ 5.41 (overlapped, H-4'), 5.39 (brt,

178

8.4, H-23), 4.90 (d, 7.2, H-1'), 4.39 (s, H-15), 4.38 (overlapped, H-5'a), 4.29 (t, 8.6, H-2'), 4.08 (t, 6.7, H-3'), 3.63 (t, 10.5, H-5'b), 3.55 (dd, 4.0, 12.8, H-3), 3.05 (d, 8.4, H-24), 2.69 (brt, 12.8, H-22a), 2.39 (d, 6.5, H-17), 2.31 (m, H-2a), 2.13 (overlapped, H-20), 2.10 (overlapped, H-7a), 2.10 (m, H-11a), 2.07 (s, OAc), 1.99 (s, OAc), 1.95 (m, H-2b), 1.88 (dd, 4.6, 11.5, H-8), 1.82 (2H, (m, H-12), 1.78 (m, H-22b), 1.60 (overlapped, H-1a), 1.59 (m, H-6a), 1.41 (s, H-27), 1.39 (overlapped, H-5), 1.38 (s, H-18), 1.34 (s, H-29), 1.28 (overlapped, H-1b), 1.27 (d, 6.8, H-21), 1.26 (s, H-26), 1.22 (s, H-28), 1.18 (overlapped, H-7b), 1.15 (m, H- 11b), 1.07 (s, H-30), 0.79 (m, H-6b), 0.60 (d, 3.6, H-19a), 0.33 (d, 3.6, H-19b), 13 C-NMR data (500 MHz, pyridine-d5) δ 220.0 (s, C-16), 170.5 (s, OAc), 170.5 (s, OAc), 107.4 (d, C-1'), 88.5 (d, C-3), 83.0 (d, C-15), 75.8 (d, C-3'), 75.0 (d, C- 2'), 73.2 (d, C-4'), 72.1 (d, C-23), 65.2 (d, C-24), 63.2 (t, C-5'), 60.0 (d, C-17), 58.6 (s, C-25), 48.3 (d, C-8), 47.4 (d, C-5), 46.1 (s, C-14), 41.6 (s, C-13), 41.4 (s, C-4), 37.0 (t, C-22), 33.1 (t, C-12), 32.2 (t, C-1), 30.0 (t, C-2), 30.0 (t, C-19), 28.0 (d, C-20), 26.8 (s, C-10), 26.7 (t, C-11), 26.4 (t, C-7), 25.7 (q, C-29), 24.7 (q, C- 26), 21.1 (t, C-6), 21.0 (q, OAc), 20.9 (q, OAc), 20.4 (q, C-21), 20.1 (s, C-9), 19.8 −1 (q, C-18), 19.4 (q, C-27), 15.4 (q, C-30), 12.0 (q, C-28); IR νmaxNaCl (cm ): 3465, 2933, 2866, 1732, 1370, 1235, 1041, 750; HRESIMS m/z 727.4041 (calc

727.4033 for C39 H60O11Na).

20 Cimiracemoside N: pale yellow powder, mp 172-174 °C, [α] D -70.36 (c 0.367, 1 CHCl3); H-NMR data (500 MHz, pyridine-d5) δ 5.12 (overlapped, H-12), 4.78 (d, 6.8, H-1'), 4.45 (t, 14.5, H-2'), 4.33 (brs, H-4'), 4.31 (d, 11.0.2, H-5'a), 4.24 (d, 6.9, H-3'), 4.24 (brt, 6.9, H-16), 4.07 (d, 10.5, H-26a), 3.80 (d, 11.0.2, H-5'b), 3.68 (s, H-24), 3.63 (d, 10.5, H-26b), 3.44 (dd, 4.2, 13.0, H-3), 2.71 (dd, 8.8, 15.4, H- 11a), 2.28 (m, H-2a), 2.23 (overlapped, H-20), 2.14 (s, H-OAc), 1.88 (m, H-15a), 1.83 (m, H-2b), 1.79 (overlapped, H-17), 1.76 (m, H-15b), 1.60 (m, H-8), 1.58 (overlapped, H-22a), 1.49 (overlapped, H-1a), 1.48 (s, H-27), 1.45 (overlapped, H-22b), 1.42 (s, H-18), 1.38 (m, H-6a), 1.29 (overlapped, H-7a), 1.27 (s, H-29), 1.22 (overlapped, H-5), 1.16 (overlapped, H-11b), 1.12 (overlapped, H-1b), 1.02 (d, 6.3, H-21), 0.96 (s, H-30), 0.89 (overlapped, H-7b), 0.85 (s, H-28), 0.63 (m, H-

179

6b), 0.54 (d, 3.6, H-19a), 0.19 (d, 3.6, H-19b), 13C-NMR data (500 MHz, pyridine- d5) δ 170.7 (s, OAc), 107.5 (d, C-1'), 105.9 (s, C-23), 88.1 (d, C-3), 77.1 (d, C- 12), 74.7 (d, C-16), 74.5 (d, C-3'), 72.9 (d, C-2'), 69.6 (d, C-4'), 68.1 (t, C-26), 66.8 (t, C-5'), 62.5 (d, C-24), 62.2 (s, C-25), 56.2 (d, C-17), 48.8 (s, C-13), 47.8 (s, C-14), 47.0 (d, C-5), 45.6 (d, C-8), 44.1 (t, C-15), 41.2 (s, C-4), 37.5 (t, C-22), 36.6 (t, C-11), 31.9 (t, C-1), 29.8 (t, C-2), 29.5 (t, C-19), 26.7 (s, C-10), 25.7 (q, C-29), 25.6 (t, C-7), 23.3 (d, C-20), 21.7 (q, OAc), 21.3 (q, C-21), 20.3 (t, C-6), 20.1 (s, C-9), 19.6 (q, C-28), 15.3 (q, C-30), 14.3 (q, C-27), 13.5 (q, C-18), IR −1 νmaxNaCl (cm ): 3454, 2935, 2871, 1729, 1455, 1372, 1244, 1070, 1030, 754;

HRESIMS 661.3962 (calc 661.3952 for C37H57O11).

20 Cimiracemoside O: pale yellow powder, mp 143-145 °C, [α] D -60.00 (c 0.160, 1 CHCl3); H-NMR data (500 MHz, pyridine-d5) δ 5.76 (s, H-26), 5.41 (ddd, 5.4, 9.7, 9.7, H-4'), 5.10 (brd, 6.0, H-12), 4.85 (d, 7.3, H-1'), 4.62 (dd, 7.1, 14.3, H-16), 4.34 (dd, 5.5,11.4, H-5'b), 4.28 (dd, 9.1,9.2, H-3'), 4.04 (dd, 8.5, 8.3, H-2'), 3.95 (s, H-24), 3.61 (dd, 10.9, 11.4, H-5'a), 3.43 (dd, 4.0, 11.4, H-3), 2.71 (m, H-11a), 2.24 (overlapped, H-22a), 2.24 (m, H-2a), 2.16 (s, OAc), 1.99 (s, OAc), 1.85 (m, H-2b), 1.80 (overlapped, H-20), 1.79 (s, H-27), 1.79 (s, H-29), 1.78 (overlapped, H-17), 1.75 (m, H-15a), 1.70 (dd, 6.5, 18.0, H-22b), 1.62 (m, H-8), 1.55 (m, H- 15b), 1.51 (overlapped, H-1a), 1.37 (s, H-18), 1.27 (m, H-6a), 1.25 (overlapped, H-7a), 1.21 (overlapped, H-5), 1.21 (overlapped, H-11b), 1.12 (overlapped, H- 1b), 0.98 (s, H-30), 0.98 (d, 6.5, H-21), 0.89 (overlapped, H-7b), 0.80 (s, H-28), 0.67 (m, H-6b), 0.57 (d, 4.5, H-19a), 0.23 (d, 4.5, H-19b), 13C-NMR data (500

MHz, pyridine-d5) δ 170.6 (s, OAc), 170.6 (s, OAc), 107.3 (d, C-1'), 105.8 (s, C- 23), 98.4 (d, C-26), 88.2 (d, C-3), 77.0 (d, C-12), 75.7 (d, C-2'), 75.0 (d, C-3'), 73.1 (d, C-4'), 73.0 (d, C-16), 65.8 (s, C-25), 63.4 (d, C-24), 63.2 (t, C-5'), 56.4 (d, C-17), 48.7 (s, C-13), 47.8 (s, C-14), 46.9 (d, C-5), 45.7 (d, C-8), 43.5 (t, C-15), 41.2 (s, C-4), 37.6 (t, C-22), 36.7 (t, C-11), 31.9 (t, C-1), 29.8 (t, C-19), 29.5 (t, C- 2), 26.7 (s, C-10), 26.0 (d, C-20), 25.6 (t, C-7), 25.6 (q, C-29), 21.6 (q, OAc), 21.0 (q, C-21), 20.9 (q, OAc), 20.4 (t, C-6), 20.1 (s, C-9), 19.5 (q, C-28), 15.3 (q, C- -1 30), 13.5 (q, C-18), 13.1 (q, C-27). IR νmax NaCl (cm ) 3449, 2940, 2869, 1732,

180

1455, 1371, 1241, 1048, 983, 755; HRESIMS m/z 701.3889 (calc 701.3901 for

C39H57O11 [M - H2O + H]+).

20 Cimiracemoside P: pale yellow powder, mp 151-153 °C, [α] D -69.77 (c 0.440, 1 CHCl3); H-NMR data (500 MHz, pyridine-d5) δ 5.11 (brd 6.8, H-12), 4.87 (d, 7.1, H-1'), 4.41 (s, H-24), 4.38 (m, H-5'a), 4.22 (m, H-4'), 4.20 (t, 8.6, H-3'), 4.05 (m, H-2'), 4.05 (brt, 7.7, H-16), 3.76 (9.8, 10.1, H-5'b), 3.48 (brd, 7.0, H-3), 2.75 (m, H-11a), 2.32 (m, H-2a), 2.21 (overlapped, H-22a), 2.16 (s, OAc), 1.90 (overlapped, H-15a), 1.88 (m, H-2b), 1.86 (overlapped, H-17), 1.85 (overlapped, H-20), 1.67 (overlapped, H-22b), 1.65 (s, H-27), 1.64 (overlapped, H-15b), 1.63 (m, H-8), 1.57 (overlapped, H-1a), 1.43 (overlapped, H-6a), 1.33 (s, H-18), 1.33 (s, H-29), 1.27 (overlapped, H-5), 1.25 (overlapped, H-7a), 1.21 (overlapped, H- 11b), 1.14 (overlapped, H-1b), 1.02 (s, H-30), 0.95 (overlapped, H-7b), 0.93 (d, 5.0, H-21), 0.85 (s, H-28), 0.70 (m, H-6b), 0.57 (d, 3.8, H-19a), 0.26 (d, 3.8, H- 13 19b), C-NMR data (500 MHz, pyridine-d5) δ 172.4 (s, C-26), 170.5 (s, OAc), 107.6 (d, C-1'), 106.2 (s, C-23), 88.1 (d, C-3), 78.7 (d, C-3'), 76.8 (d, C-12), 75.6 (d, C-16), 75.4 (d, C-2'), 71.3 (d, C-4'), 67.2 (t, C-5'), 62.7 (d, C-24), 58.6 (s, C- 25), 55.6 (d, C-17), 48.8 (s, C-13), 48.0 (s, C-14), 47.0 (d, C-8), 45.7 (d, C-5), 43.2 (t, C-15), 41.2 (s, C-4), 36.6 (t, C-11), 35.6 (t, C-22), 31.9 (t, C-1), 29.9 (t, C- 2), 29.6 (t, C-19), 26.8 (s, C-10), 25.7 (t, C-7), 25.7 (q, C-29), 25.3 (d, C-20), 21.6 (q, OAc), 20.7 (q, C-21), 20.4 (t, C-6), 20.1 (s, C-9), 19.5 (q, C-28), 15.3 (q, C- -1 30), 13.5 (q, C-18), 11.1 (q, C-27). IR νmax NaCl (cm ) 3423, 2956, 2934, 2870, 1787, 1731, 1454, 1362, 1243, 1041, 968, 753; HRESIMS m/z 675.3758 (calc

675.3744 for C37H55O11).

20 23-epi-26-deoxyactein: colorless needles, mp 251-253 °C, [α] D -61.18 (c 1 0.255, CHCl3-MeOH, 1:1); H-NMR data (500 MHz, pyridine-d5) δ 5.10 (brd, 5.3, H-12), 4.83 (d, 7.2, H-1'), 4.35 (dd, 10.5, 4.2, H-5'a), 4.23 (brd, 6.7, H-16), 4.21 (m, H-4'), 4.14 (t, 8.6, H-3'), 4.05 (d, 10.4, H-26a), 4.04 (s, H-24), 4.01 (t, 7.8, H- 2'), 3.73 (t, 10.2, H-5'b), 3.62 (d, 10.4, H-26b), 3.44 (dd, 3.4, 10.8, H-3), 2.70 (dd, 8.6, 15.5, H-11a), 2.28 H-2a), 2.23 H-20), 2.12 (s, OAc), 1.89 (dd, 6.7, 11.8, H-

181

15a), 1.84 H-2b), 1.78 H-15b), 1.76 (overlapped, H-17), 1.59 (brd, 12.4, H-8), 1.59 (brd, 12.4, H-22a), 1.52 H-1a), 1.45 (s, H-27), 1.44 (brd, 12.4, H-22b), 1.40 (s, H-18), 1.4 H-6a), 1.29 (s, H-29), 1.22 (brd, 10.1, H-5), 1.21 H-7a), 1.17 (brd, 15.5, H-11b), 1.11 H-1b), 1.00 (d, 6.7, H-21), 0.99 (s, H-30), 0.91 H-7b), 0.83 (s, H-28), 0.60 (brd, 13.6, 11.1, H-6b), 0.52 (d, 3.8, H-19a), 0.19 (d, 3.8, H-19b),, 13 C-NMR data (500 MHz, pyridine-d5) δ 170.7 (s, OAc), 107.5 (d, C-1'), 105.9 (s, C-23), 88.1 (d, C-3), 78.7 (d, C-3'), 77.1 (d, C-12), 75.6 (d, C-2'), 74.5 (d, C-16), 71.3 (d, C-4'), 67.2 (t, C-5'), 67.1 (t, C-26), 62.5 (d, C-24), 62.3 (s, C-25), 56.2 (d, C-17), 48.8 (s, C-13), 47.9 (s, C-14), 47.0 (d, C-5), 45.6 (d, C-8), 44.2 (t, C-15), 41.2 (s, C-4), 37.6 (t, C-22), 36.7 (t, C-11), 32.0 (t, C-1), 30.0 (t, C-2), 29.5 (t, C- 19), 26.8 (s, C-10), 25.7 (t, C-7), 25.7 (q, C-29), 23.3 (d, C-20), 21.7 (q, C-21), 21.4 (q, OAc), 20.4 (t, C-6), 20. 2 (s, C-9), 19.7 (q, C-28), 15.3 (q, C-30), 14.3 (q, C-27), 13.5 (q, C-18), FABMS m/z (relative intensity %) 661.4 (0.75), 613.2 (2.0), 460.1 (11), 307.1 (100), 289.1 (43), 219.2 (9), 154.1 (100), 136.1 (100), 107.9

(75), 78.9 (35), 66.3 (27); HRFABMS m/z 683.3779 (calcd 683.3771 for C37

H56O10Na).

20 26-deoxyactein: colorless needles, mp 253-254 °C, [α] D -52.17 (c 0.025, 1 CHCl3); H-NMR data (500 MHz, pyridine-d5) δ 5.11 (dd, 3.6, 8.8, H-12), 4.86 (d, 7.6, H-1'), 4.53 (brdd, 7.2, 14.2, H-16), 4.38 (dd, 11.2, 5.1, H-5'b), 4.24 (m, H-4'), 4.18 (t, 8.7, H-3'), 4.05 (t, 7.8, H-2'), 3.95 (d, 10.0, H-26a), 3.88 (d, 10.0, H-26b), 3.77 (t, 10.0, H-5'a), 3.71 (s, H-24), 3.48 (dd, 4.3, 11.7, H-3), 2.74 (dd, 7.1, 15.9, H-11a), 2.28 (H-2a), 2.19 (brd, 12.4, H-22a), 2.15 (s,OAc), 1.91 (dd, 8.0, 12.6, H- 15a), 1.88 (H-2b), 1.80 (overlapped, H-17), 1.79 (H-20), 1.68 (dd, 6.6, 12.6, H- 15b), 1.64 (dd, 4.7, 14.4, H-8), 1.6 (H-22b), 1.52 (H-1a), 1.47 (s, H-27), 1.44 (H- 6a), 1.37 (s,H-29), 1.35 (s,H-18), 1.3 (H-7a), 1.26 (brd, 12.4, H-5), 1.20 (dd, 3.7, 15.9, H-11b), 1.15 (H-1b), 1.02 (s, H-30), 0.96 (d, 5.7, H-21), 0.95 (H-7b), 0.86 (s, H-28), 0.69 (brd, 12.7, H-6b), 0.60 (d, 4.0, H-19a), 0.25 (d, 4.0, H-19b) 13C-

NMR data (500 MHz, pyridine-d5) δ 170.6 (s, OAc), 107.5 (d, C-1'), 105.9 (s, C- 23), 88.1 (d, C-3), 78.7 (d, C-3'), 77.1 (d, C-12), 75.6 (d, C-2'), 73.0 (d, C-16), 71.3 (d, C-4'), 68.8 (t, C-26), 67.1 (t, C-5'), 63.3 (s, C-25), 63.3 (d, C-24), 56.5 (d,

182

C-17), 48.8 (s, C-13), 48.0 (s, C-14), 47.0 (d, C-5), 45.8 (d, C-8), 43.7 (t, C-15), 41.2 (s, C-4), 36.7 (t, C-11), 36.7 (t, C-22), 32.0 (t, C-1), 29.9 (t, C-2), 29.6 (t, C- 19), 26.8 (s, C-10), 26.0 (d, C-20), 25.7 (t, C-7), 25.7 (q, C-29), 21.0 (q, C-21), 20.7 (q, OAc), 20.5 (t, C-6), 20. 2 (s, C-9), 19.6 (q, C-28), 15.4 (q, C-30), 13.8 (q, C-27), 13.5 (q, C-18), FABMS m/z (relative intensity %) 661.4 (6), 451.3 (25), 307.1 (100), 289.1 (58), 219 (15), 154.1 (100), 136.1 (80), 107.9 (60), 78.9 (60);

HRFABMS m/z 683.3775 (calcd 683.3771 for C37 H56O10Na).

183

10.2 Mass spectral data supporting the presence of additional nitrogenous constituents in Black Cohosh.

Samp le g11 on amide column crjuly 150403 TOF MS ES+ 15.19 100 180 180.1 7.42e3

%

5.64 180.1 3.92 163.1

0 crjuly150403 TOF MS ES+ 27.24 100 342 342.2 1.82e3

%

4.97 342.3

8.25 6.28 342.2 11.81 3.33 137.17.24 20.57 344.3 163.1 208.2 270.2 0 Time 2.00 4.00 6.00 8.00 10.00 12.00 14.00 16.00 18.00 20.00 22.00 24.00 26.00 28.00 30.00

Appendix Figure 1. Additional LC-MS/MS spectral analysis supporting the presence of compounds in Cimicifuga racemosa fraction g11 with an odd number of nitrogens (n=1,3,5,..etc.).

184

Sample g11 on amide column ms/msSample g11 on amide column ms/ms 15-JUL-2004 crjuly150404 119 (16.523) Sm (SG, 1x3.00); Cm (112:127) 3: TOF MSMS 172.10ES+ 70.1025 100 501

%

112.1264

0 crjuly150404 72 (13.992) Sm (SG, 1x3.00); Cm (62:81) 3: TOF MSMS 172.10ES+ 70.1059 100 564

%

172.1561 130.1292

94.1059 112.1177 154.1461 71.0933 113.1096 137.1125

0 m/z 70 80 90 100 110 120 130 140 150 160 170

Appendix Figure 2. High resolution positive-ion electrospray tandem mass spectrum of the protonated molecule of m/z 172.1561, in fraction g11, obtained utilizing a quadrupole time-of-flight mass spectrometer. This mass and fragmentation pattern are similar to cimipronidine.

185

Sample g11 on amide column ms/msSample g11 on amide column ms/ms 15-JUL-2004 crjuly150404 48 (15.528) Sm (SG, 1x3.00); Cm (41:55) 5: TOF MSMS 154.10ES+ 154.1411 100 99.0

70.1059

%

94.0980

95.0904

112.1177

77.0875

0 m/z 65 70 75 80 85 90 95 100 105 110 115 120 125 130 135 140 145 150 155 160

Appendix Figure 3. High resolution positive-ion electrospray tandem mass spectrum of the protonated molecule of m/z 154.1411 from fraction g11 obtained utilizing a quadrupole time- of-flight mass spectrometer. The fragments are similar to cimipronidine.

186

Sample g11 on amide column ms/msSample g11 on amide column ms/ms 15-JUL-2004 crjuly150404 177 (27.079) Sm (SG, 1x3.00); Cm (148:217) 7: TOF MSMS 342.10ES+ 180.1536 100 255

163.1179

%

342.2438

145.0986

85.0697 137.0982 117.1167 301.2376 0 crjuly150404 63 (15.034) Sm (SG, 1x3.00); Cm (54:68) 4: TOF MSMS 180.10ES+ 145.1084 109 100 117.1123

%

163.1179

115.0979

91.0898 180.1426

0 m/z 80 100 120 140 160 180 200 220 240 260 280 300 320 340

Appendix Figure 4. High resolution positive-ion electrospray tandem mass spectrum of the protonated molecules of m/z 342.2438 and 180.1426 obtained utilizing a quadrupole time-of- flight mass spectrometer. The fragmentation pattern varies from the pattern seen with cimipronidine.

187

10.3 Additional Black Cohosh Documentation

Appendix Figure 5. Permit to Collect Cimicifuga species in Great Smoky Mountain National Park (GSMNP). Permits were held by the UIC/NIH Center from 1999 to 2002 to collect in the GSMNP.

188

Appendix Figure 6. Permit to Collect Cimicifuga species in the Blue Ridge Parkway (BRP). Permits were held by the UIC/NIH Center from 1999 to 2002 to collect in the BRP.

189

Appendix Figure 7. Results for the clinical black cohosh extract, screen for heavy metals by USP <561> using Inductively Coupled Plasma-Mass Spectrometry (ICP-MS). The result of 13 ppm is within acceptable limits for total heavy metals set forth by USP of 20 ppm.

190

Appendix Figure 8. Results for the clinical black cohosh extract screen for Lead (Pb) and Cadmium (Cd) using a graphite furnace. The results were below level of quantitation. This indicated no potential toxic concentration of either heavy metal was present in the extract. The lead level at <0.1 ppm is below the California Proposition 65 level of 0.5 ppm of lead daily.

191

Appendix Figure 9. USP pesticide screen of black cohosh extract. No detectable pesticides in the extract were reported.

192

Appendix Figure 9 (continued). USP pesticide screen of black cohosh extract. No detectable pesticides in the extract were reported.

193

Appendix Figure 10. Ames Test screening for mutagenicity of TA98 without metabolic activation (-S9) of Black Cohosh Phase I extract. The result was no mutagenic effect.

194

Appendix Figure 11. Ames Test screening for mutagenicity of TA100 without metabolic acitivation (-S9) of Black Cohosh Phase I extract. The result was no mutagenic effect.

195

Appendix Figure 12. Ames Test screening for mutagenicity of TA98 with metabolic acitivation of Black Cohosh Phase I extract. The result was no mutagenic effect.

196

Appendix Figure 13. Ames Test screening for mutagenicity of TA100 with metabolic activation of Black Cohosh Phase I extract. The result was no mutagenic effect.

197

Appendix Figure 14. Ames Test screening methodology and positive control data.

198

Appendix Figure 15. Material data safety sheet from Pure World Botanicals on clinical black cohosh extract.

199

Appendix Figure 16. Pure World Botanicals certificate of analysis of Clinical Black Cohosh extract.

200

Appendix Figure 17. Batch record of Phase I clinical encapsulation from Pharmavite corporation.

201

Appendix Figure 18. Additional batch record report from Pharmavite corporation for the Phase I clinical capsule.

202

Appendix Figure 19. Mixing formulation record of Phase I clinical capsule from Pharmavite Corporation.

203

Appendix Figure 20. Clinical capsule specifications summary from Pharmavite corporation.

204

11.0 Vita

1.0 Personal Data

Daniel S. Fabricant Program for Collaborative Research in the Pharmaceutical Sciences, UIC/ NIH Center for Botanical Dietary Supplement Research, College of Pharmacy, University of Illinois at Chicago 833 S. Wood Street (M/C 877), Chicago, Illinois 60612 Phone: 312.996.7253 Fax: 312.413.5894 E-mail: [email protected]

2.0 Education

1998-2005 Doctoral Candidate, Pharmacognosy, UIC/NIH Center for Botanical Dietary Supplements Research, Program for Collaborative Research in the Pharmaceutical Sciences (PCRPS), University of Illinois at Chicago, Chicago, Illinois.

1997 B.S., Chemistry, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina.

3.0 Positions Held and Responsibilities

2005 Assistant Director, Research, ConsumerLab.com, Pasadena, Maryland

2000-2005 Research Assistant, NIH/ODS Center for Botanical Dietary Supplement Research. University of Illinois at Chicago. Development of a chemically, biologically and botanically standardized dosage form of Cimicifuga racemosa for use in Clinical Trials. Isolation and structure elucidation of bioactive constituents from polar fractions. HPLC-ELSD and RAPD-PCR method development for identification of botanicals. Procurement and identification of plant samples. Organic natural product synthesis. Consulted with clinical arm on DSHEA for IRB approval. Advisors: Norman R. Farnsworth and Guido F. Pauli

1999- 2001 Production Associate, NAPRALERT database, University of Illinois at Chicago. Evaluation of scientific literature relating to the chemistry, biological activity and traditional uses of natural products, coding of pharmacological activity data into database.

1998 Teaching Assistant, Department of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, University of Illinois at

205

Chicago. Taught over 30 lecture and recitation sections for professional pharmacy medicinal/organic chemistry courses.

1998 Editorial Assistant, WHO Collaborating Centre for Research in Traditional Medicine, University of Illinois at Chicago. Research and editing of WHO monographs on Selected Medicinal Plants, Volume 1.

1997 Research and Development Scientist, Viragen Inc., Hialeah, FL. HPLC and SEC method development for quantification and purification of interferons. ELISA techniques for identification of interferons. Performed cGMP HPLC stability studies of interferons. Supervisor: Duy Nguyen, Ph.D.

1997 Undergraduate Teaching Assistant, University of North Carolina- Chapel Hill. Chemistry 11-Freshman Chemistry Laboratory. Proctored lectures, laboratory experiments and examinations. Supervisor: Don C. Jicha, Ph.D.

1996 Undergraduate Research Assistant, University of North Carolina- Chapel Hill. Protein synthesis and purification. Mentor: Gary J. Pielak, Ph.D.

1994-1995 Summer Intern, Dade County Medical Examiners’ Office, Toxicology Lab, Miami, FL. HPLC, GC method development for detection and quantification of parent compounds and drug metabolites. Mentor: W. Lee Hearn, Ph.D.

4.0 Society Memberships

2003-2005 American Society of Pharmacognosy (ASP)

1998-2002 American Association for the Advancement of Science (AAAS)

1995-2002 American Chemical Society (ACS)

5.0 Consulting Activities

2003 Bio-equivalence studies on generic Misoprostol. Development of HPLC-ELSD, -MS methodology.

2003 United States Pharmacopoeia. Monograph evaluation of Cimicifuga racemosa extracts.

2002 American Herbal Pharmacopoeia Monographs on Cimicifuga racemosa.

206

1998 Tom’s of Maine, Research and development work. Performed stability studies on a variety botanical extracts. Developed formulation and extraction protocols of bulk plant material used for commercial products.

6.0 Additional Training

2003 Food and Drug Law Institute (FDLI)- Dietary Supplements at a Crossroads, January 15-16, Washington D.C.

2002 Food and Drug Law Institute (FDLI)-Introduction to Food Law, January 14-15, Washington D.C., Sponsored by ODS

2000 NCCAM Workshop on RT-PCR of Botanicals, November 2-3, NIEHS, RTP, North Carolina. Sponsored by ODS

7.0 Peer Reviewed Publications

2005 Cimipronidine a novel alkaloid from C. racemosa J Nat Prod (Re- Submitted)

2005 Synthesis of cimiracemate B, a phenylpropanoid found in Cimicifuga racemosa. Chen SN, Fabricant DS, Pauli GF, Fong HH, Farnsworth NR. Nat Prod Res. 19(3):287-90

2005 Marcel Dekker/Office of Dietary Supplements Encyclopedia of Dietary Supplements, New York, Paul Coates, Editor, Black Cohosh by Fabricant DS, Farnsworth NR, pp.41-54

2003 Black cohosh acts as a mixed competitive ligand and partial agonist of the serotonin receptor. Burdette JE, Liu J, Chen SN, Fabricant DS, Piersen CE, Barker EL, Pezzuto JM, Mesecar A, van Breemen RB, Farnsworth NR, Bolton JL. J. Agric. Food Chem. 51(19): 5661- 5670.

2002 A preliminary RAPD-PCR analysis of Cimicifuga species and other botanicals used for women’s health. Xu H, Fabricant DS, Piersen CE, Bolton JL, Pezzuto JM, Fong H, Totura S, Farnsworth NR, Constantinou AI. Phytomedicine 9(8): 757-62.

2002 Black Cohosh: an alternative therapy for menopause? Mahady, GB, Fabricant DS, Chadwick LR, Dietz B. Nutr. Clin. Care 5(6): 283-9.

2002 High-performance liquid chromatographic analysis of Black Cohosh (Cimicifuga racemosa) constituents with in-line evaporative light

207

scattering and photodiode array detection. Li WK, Chen SN, Fabricant D, Angerhofer CK, Fong HHS, Farnsworth NR, Fitzloff JF. Anal. Chim. Acta 471(1): 61-75.

2002 Black Cohosh (Cimicifuga racemosa) protects against menadione- induced DNA damage through scavenging of reactive-oxygen species: Bioassay-directed isolation and characterization of active principles. Burdette JE, Chen S, Lu ZZ, Xu H, White B, Fabricant DS, Liu J, Fong HHS, Farnsworth NR, Constantinou AI, van Breemen RB, Pezzuto JM, Bolton JL. J Agric Food Chem. 50(24): 7022-8.

2002 Cimiracemates A-D, novel phenylpropanoid esters from the rhizomes of Cimicifuga racemosa Chen SN, Fabricant DS, Lu ZZ, Zhang HJ, Fong HHS, Farnsworth NR. Phytochemistry 61(4): 409- 13.

2002 Cimiracemosides I-P, new 9,19-cycloartane triterpene glycosides from C. racemosa Chen SN, Fabricant DS, Lu ZZ, Fong HHS, Farnsworth NR. J. Nat. Prod. 65(10): 1391-7.

2002 The absolute configurations of 26-deoxyactein and 23-epi-26- deoxyactein. Chen SN, Li WK, Fabricant DS, Santarsiero BD, Mesecar A, Fitzloff JF, Fong HHS, Farnsworth NR. J. Nat. Prod. 65(4):601-5.

2001 The Value of Plants Used in Traditional Medicine for Drug Discovery. Fabricant DS, Farnsworth NR. Environ. Health Perspect. 109(S1): 69-75.

2000 Plants Used Against Cancer: An Extension of the Work of Jonathan Hartwell. Graham JG, Fabricant DS, Quinn ML, Farnsworth NR, J. Ethnopharmacology 73 (3): 347-377.

8.0 Poster Presentations

2004 A novel guanidine alkaloid from C.racemosa. Fabricant DS, Chen SN, Nikolic D, Jaki B, Lankin DC, van Breemen RB, Pauli GF, Farnsworth NR, In: 45th Annual ASP meeting, July 23-25; Tuscon, Arizona.

2002 Development of a chemical, biologically and botanically standardized dosage form of Cimicifuga racemosa for use in clinical trials. Fabricant DS, Chen SN, Nikolic D, Burdette JE, Li WK, Fitzloff JF, van Breemen RB, Bolton JL, Fong HHS, Farnsworth, In:

208

NCCAM/ODS Center Directors Meeting 2002, July 8-10; Bethesda, Maryland.

2001 Geographical and seasonal variation of major chemical constituents of Cimicifuga racemosa. Fabricant DS, Chen SN, Li WK, Fitzloff JF, Fong HHS, Farnsworth NR, In: Proceedings ASP/CRN meeting: Botanical Dietary Supplements: Natural Products at a Crossroads, November 8-11; Asilomar, California.

2001 Using RAPD-PCR markers to identify plant species and variants. Xu H, Fabricant DS, Johnson HE, Piersen CE, Bolton JL, Farnsworth NR, Constantinou A, In: Proceedings ASP/CRN meeting: Botanical Dietary Supplements: Natural Products at a Crossroads, November 8-11; Asilomar, California.

2001 High performance liquid chromatographic analysis of Black Cohosh (Cimicifuga racemosa L.) with in-line photodiode array and evaporative light scattering detection. Li W, Chen SN, Fabricant DS, Fong HHS, Farnsworth NR, Fitzloff JF, In: Abstracts 222nd ACS National Meeting, August 26-30, Chicago, Illinois.

9.0 Lectures Presented

2003 UIC/NIH Center for Botanical Dietary Supplements Research Brainstorming Sessions. Food and Drug Law: Dietary Supplements. February 24th .

2002 UIC/NIH Center for Botanical Dietary Supplements Research Brainstorming Sessions. Cimicifuga racemosa: Research Update. October 21st

10.0 Meetings attended

2002 NCCAM/ODS 2nd Annual Center Directors Meeting 2002, July 8-10; Bethesda, Maryland.

2001 ASP/CRN meeting: Botanical Dietary Supplements: Natural Products at a Crossroads, November 8-11; Asilomar, California.

1999 ASP midterm meeting: Botanical Dietary Supplements: May 1-3; Tunica, Mississippi.

1998 39th Annual ASP meeting: July 25-28; Orlando, Florida.

11.0 Honors and Awards

2004 Myron Goldsmith Scholarship

209

2001-2005 College of Pharmacy Dean’s Scholarship Recipient

1994 Dean’s List, Ithaca College

13.0 Volunteer and Other Activities

2000 Co-Coordinator. Norman R. Farnsworth’s 70th birthday celebration at the Drake Hotel, Chicago, Illinois. Raised over $ 25,000 in funds from ticket sales and industrial contributions to cover expenses. Responsible for site-selection, student participation and coordination of international participation.

1998-1999 Graduate Student Council Representative, University of Illinois at Chicago. Participated as a representative for the National Graduate Student Meeting (NAGPS). Co-Chairperson on subcommittee for incorporation of optical and dental health care plans for graduate student health insurance at UIC.

1996-1997 Americorps, University of North Carolina-Chapel Hill. Literacy tutor for migrant workers learning English as a second language (ESOL). Organized Carr Court summer camp for educationally challenged children.

1995-1997 Varsity Letterman, Football, University of North Carolina-Chapel Hill.

210