You are on page 1of 40

AmericanHerbal Pharmacopoeia

and !herapeutic "ompendium


Black Cohosh Rhizome


Actaea racemosa L.
syn. Cimicifuga racemosa (L.) Nutt.
Standards of Analysis, Quality Control,
and Therapeutics

2002
Editor: Roy Upton Herbalist
Associate Editor and Monograph
Development Coordinator
Alison Graff PhD
Research Associate
Diana Swisher BA

Board of Directors
Roy Upton Executive Director
American Herbal Pharmacopoeia™
Santa Cruz, CA

Michael McGuffin President


American Herbal Products Association
Silver Spring, MD

Joseph Pizzorno ND President Emeritus


Bastyr University
Kenmore, WA
Authors Quantitative Standards and Daniel Fabricant BS
Norman Farnsworth PhD
Aviva Romm CPM Herbalist (AHG)
President
International Status
Department of Medicinal Chemistry American Herbalists Guild
History Josef Brinckmann and Pharmacognosy Canton, GA
Research and Development Manager University of Illinois
Michael A Flannery MA MLS Clarissa Smith Herbalist LAc
Traditional Medicinals Inc Chicago, IL
Assistant Professor Seattle, WA
Sebastopol, CA
Associate Director for Historical Dr Christoph Gorkow
Collections Robin Suggs Executive Director
Therapeutics Department of Medical Sciences Yellow Creek Botanical Institute
Lister Hill Library of the Health Bionorica Arzneimittel
Sciences Pharmacokinetics, Pharmacodynamics, Robbinsville, NC
and Clinical Efficacy Neumarkt, Germany
University of Alabama Andrew Weil MD
Birmingham, AL Leonard Wisneski MD FACP Dr Hansjörg Hagels Program in Integrative Medicine
Clinical Professor of Medicine Analytical Development Division School of Medicine
Botanical Identification George Washington University Medical Schaper-Brümmer GmbH & Co KG University of Arizona
Center Salzgitter, Germany Tucson, AZ
Robin Bencie
Washington, DC Tori Hudson ND
Alison Graff PhD Eric Yarnell ND RH (AHG) President
American Herbal Pharmacopoeia™ Medical Indications Supported by Portland Naturopathic Clinic Botanical Medicine Academy
Santa Cruz, CA Traditional and Modern Experience Portland, OR Sisters, OR
Jill Stansbury ND Chair Frank Jaksch PhD President
Macroscopic Identification and Department of Botanical Medicine Chromadex Inc
Commercial Sources and Handling National College of Naturopathic Laguna Hills, CA Final Reviewers
Medicine
Tammi Hartung Herbalist Jim Kroll MS Adriane Fugh-Berman MD
Portland, OR
Desert Canyon Farm and Learning Senior Chemist Assistant Clinical Professor
Center Safety Profile Waters Corporation Department of Medicine
Canyon City, CO Milford, MA Department of Health Care Sciences
James Reinhart RPh George Washington University School of
Microscopic Identification Director of Pharmacy Prof Dr Eckehard Liske
Medicine
Selma Community Hospital International Medical Support Division
Prof Dr Reinhard Länger Washington, DC
Selma, CA Schaper-Brümmer GmbH & Co KG
Institute for Pharmacognosy Salzgitter, Germany Prof Dr Hubertus Jarry
Center of Pharmacy Department of Clinical and
Dr Reinhard März
University of Vienna
Vienna, Austria
Review Committee Department of Medical Sciences
Experimental Endocrinology
University of Göttingen
Erdal Bedir PhD Bionorica Arzneimittel
Göttingen, Germany
Constituents National Center for Natural Products Neumarkt, Germany
Research Fredi Kronenberg PhD
Mythili Nagarajan MS Joe-Ann McCoy MS
School of Pharmacy Richard and Hinda Rosenthal Center for
Research and Development Chemist Research Associate
University of Mississippi Complementary and Alternative
Enzymatic Therapy Clemson University
University, MS Medicine
Green Bay, WI Clemson, SC
Columbia University
Kerry Bone MCPP FNHAA FNIMH Amanda McQuade Crawford Herbalist College of Physicians and Surgeons
Analytical Herbalist MNIMH (AHG) Director New York, NY
MediHerb PTY Ltd Ojai Center of Phytotherapy
Substantiating Laboratories Helene Leonetti MD
Warwick, Queensland Ojai, CA
High Performance Thin Layer Associate Professor
Australia
Chromatography (TLC/HPTLC) Beat Meier PhD Department of Obstetrics and
Kathrin Thiekoetter Mary Bove Herbalist ND Zeller AG, Herbal Medicinal Products Gynecology
Eike Reich PhD Brattleboro Naturopathic Clinic Romanshorn, Switzerland Temple University
CAMAG Brattleboro, VT Philadelphia, PA
Muttenz, Switzerland William Mitchell ND
Francis Brinker ND Seattle, WA
High Performance Liquid Eclectic Institute and
Chromatography (HPLC) Program in Integrative Medicine Prof Dr Adolf Nahrstedt
Paula Brown MS School of Medicine Editor Planta Medica
Ryan Takahashi University of Arizona Pharmaceutical Biology and
Herbal Evaluation and Analysis Tucson, AZ Phytochemistry
Laboratory University of Münster
Chanchal Cabrera Herbalist MNIMH Münster, Germany
Forensics Department (AHG)
British Columbia Institute of Gaia Garden Herbals Gwynn Ramsey PhD
Technology Vancouver, BC Professor of Biology Emeritus
Burnaby, British Columbia Curator, Ramsey-Freer Herbarium
Canada Dr Volker Christoffel Lynchburg College
Department of Medical Sciences Lynchburg, VA
Institute for Nutraceutical Advancement Bionorica Arzneimittel
(INA) Neumarkt, Germany Joseph Romano MA
Methods Validation Program (MVP) Marketing Manager
Apex, NC Steven Dentali PhD Waters Corporation
Dentali Associates Milford, MA
Boca Raton, FL

ISBN: 1-929425-14-7 ISSN: 1538-0297


© Copyright 2002 American Herbal Pharmacopoeia™ Medical Disclaimer Design & Composition
Post Office Box 66809 Scotts Valley, CA 95067 USA The information contained in this monograph represents a syn- Beau Barnett
All rights reserved. No part of this monograph may be repro- thesis of the authoritative scientific and traditional data. All Felton, CA
duced, stored in a retrieval system, or transmitted in any form or efforts have been made to ensure the accuracy of the informa- Cover Photograph
by any means without written permission of the American tion and findings presented. Those seeking to utilize botanicals Black cohosh (Actaea racemosa) © 2002
Herbal Pharmacopoeia™. as part of a health care program should do so under the guid- courtesy of Martin Wall Photography,
ance of a qualified health care professional. Pleasant Garden, NC
Literature retrieval provided by CCS Associates Inc, Mountain
View, CA. Statement of Nonendorsement
The reporting on the use of proprietary products reflects studies
The American Herbal Pharmacopoeia™ is a nonprofit educa- conducted with these and is not meant to be a product endorse-
tional corporation 501(c)(3). All funding of the American Herbal ment.
Pharmacopoeia™ comes from unrestricted donations and grants.
To purchase a subscription or individual monographs, contact
the American Herbal Pharmacopoeia™ • PO Box 5159 • Santa
Cruz, CA 95063 • USA • (831) 461-6318 or visit the AHP web-
site at www.herbal-ahp.org.
N O M E N C L AT U R E and botrus (a bunch), referring to the large raceme of fruits
(follicles). Later another botanist, De Candolle, mistakenly
Botanical Nomenclature referred to it as Macrotys, a name that stuck and was pre-
Actaea racemosa L. ferred by the Eclectics (Lloyd and Lloyd 1931b).
syn. Cimicifuga racemosa (L.) Nutt. According to Native American ethnobotanical litera-
ture, black cohosh was used for a wide range of conditions
Botanical Family by eastern tribes including the Cherokee, Delaware,
Iroquois, Mikmaq (Micmac), and Penobscot. Medicinal
Ranunculaceae
uses among these tribes included its application as a seda-
Definition tive, anodyne, gynecological remedy, treatment for
rheumatic conditions, and treatment for hives (Moerman
Black cohosh consists of the fresh or dried rhizome and 1986). The first reference to black cohosh in published lit-
roots of Actaea racemosa L. conforming to the methods of erature is unclear. Some attribute the initial citation to
identification provided. Leonard Plunkenet as early as the 1680s, but this is not
definitive. A more conclusive listing of black cohosh (as
Common Names Actaea racemosa) occurs in a translation of Carl von Linne’s
United States: Black cohosh (standardized common botanical work of 1749 in which it was classified as a repel-
name), black bugbane, black snakeroot, lant (reducing swelling), diaphoretic, and anodyne, and as
rheumatism weed (McGuffin and others useful for “female debility” (Whitlaw 1829).
2000). The American medical botanist Benjamin Smith
Germany: Traubensilberkerze, Wanzenkraut. Barton gave more extensive treatment to the plant in his
Collections for an Essay Towards a Materia Medica of the
History United States (1810). He acknowledged its medicinal use by
Black cohosh is an indigenous North American medicinal
plant that was widely used by Native Americans and
Eclectic physicians and continues to be utilized by modern
midwives and practitioners of herbal medicine. Botanically,
black cohosh is best known by its former scientific name
Cimicifuga racemosa (L.) Nutt. It has recently been reclassi-
fied into the genus Actaea, where it was originally placed by
Linnaeus (1753) (Compton and others 1998). Actaea comes
from the word aktea, which is Greek for the elder tree
(Sambucus spp.). Presumably it was called such due to the
similarity in leaf morphology found between the two genera
(Ford 1997). Texts from the early 1800s to the present place
black cohosh in the genus Cimicifuga, a name derived from
the Latin cimex (bedbug) and fugare (to drive away) in ref-
erence to a European species in this genus commonly
known as bugbane and used as an insect repellant. This
common name was later misapplied to our domestic
Cimicifuga (Felter and Lloyd 1909; Lloyd 1921). The
species name racemosa is derived from the Latin racemosus,
meaning “full of clusters”, in reference to the large inflores-
cence of many small white flowers.
Throughout American history black cohosh has been
known by several other common names including baneber-
ry, black snakeroot, and rheumatism weed, although the
most widely applied has been black cohosh. According to
Lloyd and Lloyd (1931a), “cohosh” is an Algonquin word
meaning “it is rough” (with hairs) and was originally applied
to the bristly fruit of Ribes lacustre. Its application by whites
to blue cohosh (Caulophyllum thalictroides), species of
Actaea such as red and white baneberry (A. rubra and A.
pachypoda, respectively), as well as black cohosh, none of
which have hairs, has not been explained (Erichsen-Brown
1979; Lloyd and Lloyd 1931b). The noted medical botanist
and naturalist Constantine Rafinesque in 1808 gave black Figure 1 Black cohosh (Actaea racemosa)
cohosh the name Macrotrys from the Greek makros (large) Source: Good, The Family Flora and Materia Medica (1845).

American Herbal Pharmacopoeia™ • Black Cohosh Rhizome • 2002 1


Table 1 Historical timeline of the medical use of black cohosh (Actaea racemosa)
Native American use Black cohosh was widely used by many tribes including the Cherokee, Delaware, Iroquois, Mikmaq, and Penobscot
as a sedative, anodyne, antirheumatic, and general gynecological remedy.
1808 The noted medical botanist Constantine Rafinesque gives black cohosh the name Macrotrys, which is subsequently
changed to Macrotys by the botanist De Candolle.
1810-1830 The medical use of black cohosh by Native Americans is reported in the works of such early American medical
botanists as Benjamin Smith Barton, Jacob Bigelow, Constantine Rafinesque, and Elisha Smith.
1830-1920 Black cohosh is official in the primary list of the United States Pharmacopoeia.
1831 Young publishes a paper on the medical use of black cohosh in the American Journal of Medical Science .
1832 The Thomsonian practitioner Howard reports on the use of black cohosh for the treatment of small pox.
1832-1955 Black cohosh is listed in the United States Dispensatory.
1835 John King introduces the resin preparation of black cohosh, sold as cimicifugin, macrotyn, and macrotin, into
Eclectic medical practice. In addition to its use as a parturient, black cohosh subsequently becomes a popular
Eclectic remedy for use in the treatment of nervous conditions, myalgia, neuralgia, infections, false and true labor
pains, dysmenorrhea, and ammenorrhea.
1980-present The pharmacology and therapeutic efficacy of black cohosh are intensively studied in Germany. Black cohosh is
reclassified from Cimicifuga racemosa (L.) Nutt. back to the original Linnean designation Actaea racemosa L.
2001 Black cohosh is proposed for re-inclusion in the United States Pharmacopeia – National Formulary.

Native Americans and observed that as a sore throat remedy 1990; Lloyd 1912). According to Felter and Lloyd (1898),
“a strong decoction of the root was used, with great benefit, the rise in popularity of the use of black cohosh among the
as a gargle”. Other prominent medical writers of the time Eclectics was due to the medical experience of John King
such as Elisha Smith and Jacob Bigelow also wrote about who began using it in 1832 when few other Eclectics were
black cohosh but they simply reported on the Native uses of doing so. Consequent to King introducing a resin prepara-
the plant, adding no new information regarding its medical tion (sold as cimicifugin, macrotyn, and macrotin) in 1835,
application (Lloyd 1921). It was not until 1831 that black black cohosh was found in general practice by around 1850
cohosh was definitively introduced into medical practice in (Felter and Lloyd 1898). King reported that the effects of
the United States by Young, who published a paper in the black cohosh on the heart and circulatory system were sim-
American Journal of Medical Science. In 1889, Sir Lauder ilar, though less pronounced, to those of Digitalis, stating it
Brunton recommended black cohosh as a stomachic, car- slowed the heart and increased the heart’s contractile force
diac tonic, and expectorant (Macht and Cook 1932). and arterial tension. For use in obstetrical practice, King
In 1832, the Thomsonian practitioner Howard enthusi- considered black cohosh to be a superior and safer oxytocic
astically supported the use of black cohosh (Macrotys) for agent than ergot.
the treatment of small pox. This use was supported by a Dr Based on its widespread use, black cohosh was also
GH Norris 40 years later (1872) in a paper read before the prominent in the 19th century armamentarium of conven-
Alabama State Medical Association in which it was reported tional medical practitioners. It appeared on the secondary
that families consuming Macrotys prophylactically as a tea list of substances in the first United States Pharmacopeia
during a smallpox epidemic remained “absolutely free” of (USP) of 1820, where it was listed as an anti-inflammatory
the disease. Howard noted significant differences between and antispasmodic. It soon rose to the primary list in 1830,
the use of the tea and alcohol extracts. Of particular histori- a position it held until the 10th decennial revision of 1920
cal interest, in 1848, the Committee of the American (Boyle 1991). Black cohosh appeared in the first edition of
Medical Association “uniformly found Macrotys to lessen the United States Dispensatory (USD) in 1833 and
the frequency and force of the pulse, to soothe pain and remained through 1955 for a total of 122 years. The first
allay irritability” and held it to be “the most purely sedative USD stated that “Cimicifuga unites, with a tonic power, the
agent we possess” (Lloyd 1921). property of stimulating the secretions, particularly those of
Black cohosh was a popular Eclectic remedy used in the skin, kidneys, and pulmonary mucous membrane. It is
“muscular pains; uterine pains, with tenderness; false pains; thought by some to have a particular affinity for the uterus.
irregular pains; rheumatism of the uterus; dysmenorrhea” Its common name black snakeroot was probably derived
(Felter and Lloyd 1909). Its reputation as a virtual cure-all from its supposed power of curing the disease arising from
made black cohosh one of the most popularly prescribed the bite of the rattlesnake” (Wood and Bache 1833).
drugs according to one 1912 survey (Crellin and Philpott Carrying forward the traditional Native American use of

2 American Herbal Pharmacopoeia™ • Black Cohosh Rhizome • 2002


black cohosh for women’s ailments and Barton’s use for throat
complaints, current therapeutics finds the plant used “in a
number of preparations for coughs and for gynecological dis-
orders” (Reynolds 1996). In 2001, both the rhizome and the
dry extract of black cohosh were proposed once again for
inclusion in the United States Pharmacopeia – National
Formulary (USPF 27[4] 2001).
Asian species of Actaea (cited as Cimicifuga) are official
in the pharmacopoeias of China and Japan. Actaea racemosa,
however, is not included in modern official Western pharma-
copoeias, although it is listed in the British Herbal
Pharmacopoeia (1996) and a monograph by the World Health
Organization is currently in preparation. Black cohosh is
approved by the German Health Authority (Commission E)
for “premenstrual discomfort, dysmenorrhea, or climacteric
(menopausal) neurovegetative ailments” (Blumenthal and Figure 2a Black cohosh (Actaea racemosa) in flower in its natural
others 1998). habitat
Photograph © 2002 courtesy of Martin Wall Photography, Pleasant Garden, NC

I D E N T I F I C AT I O N
Botanical Identification
Actaea racemosa L. Herbaceous perennial from rhizome.
Stem: Erect, solitary, to 2.5 m tall, glabrous. Leaves: Basal
and cauline, alternate, 2-4-ternately compound, petioles 15-
60 cm long, bases clasping stem; leaflets 20-70; terminal
leaflet of central division 3-lobed, 6-15 cm long, 6-16.5 cm
wide, with 3 prominent veins arising from base; sub-terminal
leaflets with blades ovate-lanceolate to obovate, 4-12 cm long
and 3-8 cm wide; margins toothed to deeply incised; green
above, paler below; glabrous or rarely pubescent along veins
of undersurface. Inflorescence: Terminal panicle of 4-9 slen-
der branches, each 7-60 cm long, pubescent; 1 bract sub-
tending each pedicel. Flowers: Perfect, radially symmetric;
sepals 4, greenish-white, caducous; petals 0; staminodes (1-) 4
(-8), petaloid, cream-colored, 2-3 mm long, clawed, apex
Figure 2b Black cohosh (Actaea racemosa) leaves
bifid; stamens 55-110; pistils 1 (-3), glabrous to pubescent,
Photograph © 2002 courtesy of Martin Wall Photography, Pleasant Garden, NC
ovary superior, style short, stigma 0.5 mm wide. Fruit: Many-
seeded follicle, 5-10 mm long, ovoid, laterally compressed with
curved, stout beak (peristent style), pubescent; seeds hemi-
spheric, brown, scales lacking. Chromosome number: n = 8.
Distribution: Moist deciduous forests, ravines, moist mead-
ows, creek margins, mountainous terrain. Flowers June to
September. Native to eastern North America from Ontario
south to Georgia and west to Missouri (Compton and others
1998; Linnaeus 1753 [original citation]; Ramsey 1965, 1987,
1997).
There are currently two varieties of A. racemosa recog-
nized based on differences in leaf morphology: var. racemosa
and var. dissecta. The former variety has triternate-pinnate
leaves with serrate margins, while the latter has quadriternate-
pinnate leaves that are deeply incised with serrate lobes
(Compton and others 1998). Variety dissecta is only known
from very few herbarium specimens, all of which were col-
lected well over 100 years ago, making this taxon of uncertain
Figure 2c Black cohosh (Actaea racemosa) in flower
taxonomic significance (Ramsey 1997). Photograph © 2002 courtesy of Martin Wall Photography, Pleasant Garden, NC

American Herbal Pharmacopoeia™ • Black Cohosh Rhizome • 2002 3


Table 2 Botanical characteristics distinguishing black cohosh
(Actaea racemosa) from yellow cohosh (Actaea podocarpa syn. Aroma: Slightly sweet and musty in both the fresh and dried
Cimicifuga americana) rhizome and root, pungent and somewhat unpleasant. Taste:
Slightly bitter, acrid, slightly mucilaginous.
Character Black cohosh Yellow cohosh
Powder: Light to dark brown in color with a heavy and dis-
agreeable aroma that fades with age (BHP 1996; Kraemer
1920; Lloyd and Lloyd 1931b; Mansfield 1937; Osol and
Farrar 1947; Sayre 1917; Steinegger and Hänsel 1992;
Fruit Youngken 1930).

Seeds

Pistils, pedicels, and bracts


Figure 3 Freshly harvested rhizome and roots of black cohosh
(Actaea racemosa)
Source: Lloyd and Lloyd (1931a).

Staminodia

Source: Ramsey (1987).

Macroscopic Identification
Black cohosh may be traded as a mixture of whole, broken,
or cut pieces of the rhizome and roots, usually dried but
sometimes fresh. In the retail market it is sold dried, cut and
sifted. The whole rhizome is 2-15.2 cm long and 0.5-3 cm
in diameter, with many tightly clustered upright or upward-
ly curving branches. Externally it is brownish-black, some- Figure 4a Freshly harvested black cohosh rhizome and roots
times drying to a grayish-brown. The entire surface is slight- (Actaea racemosa)
ly annulate due to the many leaf scales. The upper surface Photograph © 2002 courtesy of Martin Wall Photography, Pleasant Garden, NC
has many stem buds and deep, cup-shaped scars from past
stems. The lower surface has many root scars and few to
many wiry roots. The dry rhizome is resinous with a fibrous
texture and horny fracture (hard and uneven), and the fresh
rhizome is brittle and breaks with a nearly clean fracture. In
cross section the rhizome is almost round and has dark
green or brown bark approximately 1 mm thick, dark brown
wood 4-5 mm thick, and a whitish or dark brown and waxy
pith 3-5 mm in diameter. The xylem is visible as wedges
alternating with medullary rays.
The roots are 3-16 cm long and 1-5 mm in diameter,
dark brown externally, and longitudinally wrinkled. When
dry they are brittle with a short fracture. In cross section they
are round or broadly angled with a dark brown to black bark
and light to dark brown or yellowish wood. The wood is
Figure 4b Freshly harvested black cohosh rhizome and roots
arranged in a characteristic cross-like pattern with 4, some-
(Actaea racemosa)
times 2-6, radiating, widely separated strands.
Photograph © 2002 courtesy of Roy Upton, Soquel, CA

4 American Herbal Pharmacopoeia™ • Black Cohosh Rhizome • 2002


Figure 4c Freshly harvested black cohosh rhizome and roots Figure 5a Cut dried black cohosh (Actaea racemosa)
(Actaea racemosa) Photograph © 2002 courtesy of Roy Upton, Soquel, CA
Photograph © 2002 courtesy of Roy Upton, Soquel, CA

Figure 4d Dried black cohosh rhizome and roots (Actaea racemosa) Figure 5b Dried slices of black cohosh (Actaea racemosa)
Photograph © 2002 courtesy of Roy Upton, Soquel, CA Photograph © 2002 courtesy of Roy Upton, Soquel, CA

Figure 4e Freshly harvested yellow cohosh (Actaea podocarpa) Figure 5c Dried slices of sheng ma (Actaea sp.; left) and an adul-
rhizome and roots terant (Serratula sp.; right) of sheng ma
Photograph © 2002 courtesy of Thomas Dadant, Santa Cruz, CA Photograph © 2002 courtesy of David Eagle, Santa Cruz, CA

American Herbal Pharmacopoeia™ • Black Cohosh Rhizome • 2002 5


Microscopic Identification Root: The root is covered by an exodermis of dark brown
Rhizome: Beneath a dark brown exodermis is the colorless papillous cells with a considerably thickened and striated
parenchyma of the cortex. The walls of the cortical cells are outer wall. The cell walls of the cortical parenchyma are
thickened, particularly at the cell corners, and between thickened and the innermost layer of the cortex is a well-
most cells is a small triangular intercellular space. The vas- defined endodermis. Large areas of secondary xylem alter-
cular bundles are arranged radially around the central pith nate with small areas of the primary xylem; the parenchyma
and are separated by broad medullary rays. The vessels of is completely replaced by xylary fibers.
the secondary xylem are grouped with fibers. The vessels are Starch granules are abundant in the water preparation; they
up to 60 µm in diameter, with bordered pits or reticulate are mostly solitary, rarely in aggregates of 2 or 3. Solitary
secondary wall thickenings. In the outermost region of the granules are roundish and up to 10 µm in diameter.
stele, a few fibers and sclereids are embedded in each Powder: Preparation in water: starch. Preparation in chloral
phloem bundle. The phloem regions are elongated with the hydrate: xylary fibers and vessels in longitudinal view;
tissue directly outside of the bundles appearing compressed phloem fibers and sclereids; parenchyma cells.
and different from the surrounding cortical parenchyma.

Exodermis
Fibers
Secondary phloem

Secondary xylem

Pith
5.

1.
6.
Cortex

Endodermis

Xylem
2.

7.

8.

3. Figure 6a Microscopic characteristics of black cohosh rhi-


zome and root (Actaea racemosa)
1. Rhizome (cross section).
2. Root (cross section).
3. Cortical parenchyma of the rhizome (cross section).
4. Phloem fibers in the rhizome (cross section).
5. Phloem tissue with sclereid in the rhizome (longitudinal
view).
6. Vessels with bordered pits and a xylary fiber in the rhizome
(longitudinal view).
7. Root exodermis (cross section).
8. Starch granules.
4. Microscopic drawings courtesy of Reinhard Länger, University of Vienna.

6 American Herbal Pharmacopoeia™ • Black Cohosh Rhizome • 2002


1. 6.

2. 7.

Figure 6b Microscopic characteristics of black cohosh rhizome


and root (Actaea racemosa)
1. Rhizome (cross section). Note small strands of vessels and
fibers and broad medullary rays; in the secondary phloem,
parenchyma predominates with small bundles of fibers exte-
rior to the sieve cells.
2. Rhizome with vessels and fibers on the right and part of a
medullary ray on the left (cross section).
3. Secondary phloem parenchyma of the rhizome (cross sec-
tion).
4. Vessels with bordered pits in the rhizome (longitudinal view).
3. 5. Root (cross section).
6. Root with detail of the endodermis (cross section).
7. Starch granules in the powder.
Microscopic images courtesy of Reinhard Länger, University of Vienna.

4.

5.

American Herbal Pharmacopoeia™ • Black Cohosh Rhizome • 2002 7


COMMERCIAL SOURCES AND Cultivation
HANDLING Black cohosh is grown from rhizome cuttings or seeds and
requires some shading depending on altitude and other
The entire supply of black cohosh comes from the United environmental conditions. If grown from rhizome cuttings,
States. The major producers of black cohosh are Kentucky a plant takes two to three years to become reproductive;
and Tennessee, with additional supplies coming from grown from seed sown in the greenhouse and then planted
Georgia, Ohio, North Carolina, Michigan, South Carolina, out, it takes four to six years; direct-seeded it may take from
Virginia, West Virginia, and Wisconsin. Although there are six to eight years. For more complete information regarding
reports of black cohosh being grown in China and India for cultivation, see Franke and others (2000). Preliminary work
export, the true identity of the cultivated material has not indicates that black cohosh can be propagated successfully
been verified and may well be an Asian species of Actaea using in vitro techniques (Lata and others 2001).
such as A. cimicifuga (syn. Cimicifuga foetida) (HRF and
YCBI 2001). Handling and Processing
In 1998, 600,000-700,000 pounds (272-318 metric tons
Rhizomes with roots may be processed fresh or dried. They
[mt]) of black cohosh were harvested in North America.
should be thoroughly washed directly after harvest and then
Approximately 95% or more of this was wild harvested. As
laid out to dry. Freshly harvested roots should be solid but
much as 60% (320,000 pounds [145 mt]) of total produc-
not woody.
tion in 1998 was exported, and more than 75% of total
exports went to Germany. Exports reportedly increased to Drying
over 400,000 pounds (181 mt) in 1999, and then declined
sharply to 22,046-33,069 pounds (10-15 mt) in 2001. Some Rhizomes with roots are cut and air-dried at 35-45 °C. They
of this decline was due to over-stocking of raw material from are fully dried when they are brittle and snap easily and
the 1998 and 1999 harvests, the remainder of which was when no moisture is evident in cross section, either visibly
used to fill current demand (HRF and YCBI 2001). or to the touch.
Concern over the conservation of black cohosh due to
increasing demand makes this species a good candidate for
Storage
cultivation. Follow general guidelines for storage by packing in airtight
containers protected from light, heat, moisture, and insect
Collection infestation.
Traditionally, black cohosh has been harvested after plants
become reproductive, which occurs anywhere from two to
Adulterants
eight years of age in cultivated plants depending on growing Other species of Actaea, especially yellow cohosh (A.
techniques (see Cultivation). Rhizomes and roots should be podocarpa syn. Cimicifuga americana), have commonly but
harvested in autumn when the plant is dormant. At that unintentionally been mixed with A. racemosa due to the sim-
time the underground portions of the plant have lower ilarity in above-ground appearance between species. The two
moisture content than in other seasons. Fall harvesting also species can be distinguished by differences in their freshly
allows plants to produce mature seeds before being uproot- harvested underground parts: the fresh rhizome of A.
ed. A portion of the rhizome with a visible bud on it should podocarpa has a distinct yellowish hue, whereas that of A.
be left in the ground to re-sprout the following year. There racemosa is black (Figures 4a-e). The rhizomes of both
is no published information on the relationship between species are far more difficult to tell apart when dry because
the constituent profile of the rhizome and its age, growing A. podocarpa darkens upon drying. A. podocarpa has the fol-
conditions, or place of origin, although such studies are lowing other characteristics that distinguish it from A. race-
underway (Fabricant and others 2001). mosa (Table 2): where the base of the stem emerges from the
The impact of harvest on wild populations of black ground, it is slightly grooved to flat on one side rather than
cohosh is currently unknown and sources differ in their completely round (Fletcher, personal communication to
opinion about it. While some maintain that current levels AHP; unreferenced); the adaxial side of the petiole is slight-
of harvest threaten the viability of wild populations, others ly grooved rather than angled or terete; the pistils and fruits
feel that sustainable harvesting is possible at current levels are short stipitate rather than sessile; the fruit beak is slender
of demand. A study of sustainable harvest limits is currently and elongated with a minute tip rather than short and stout
underway (McCoy, personal communication to AHP; with a broad tip; the seeds are scaly; and it blooms approxi-
unreferenced). The regulatory status regarding the trade of mately three weeks later than A. racemosa (Ramsey 1997).
black cohosh is under review by the Convention on The underground portions of baneberry (Actaea pachy-
International Trade of Endangered Species (CITES). poda and A. rubra) occur as occasional adulterants of black
Refraining from harvesting plants until after they have set cohosh supplies. Fruiting plants of baneberry may be distin-
seed and leaving a portion of the rhizome in the ground to guished from black cohosh by their fleshy white or red poi-
resprout are key components to sustainable harvesting. sonous berries which contrast with the dry follicles of black
cohosh (Compton and others 1998; Ford 1997; Ramsey
1997). No information was available on how to distinguish

8 American Herbal Pharmacopoeia™ • Black Cohosh Rhizome • 2002


the underground portions of black cohosh and baneberry and Khan 2000; Shao and others 2000). Actein is a xyloside
from each other. According to one herb dealer, the roots of of acetylacteol that has a 16,23:23,26:24,25-triepoxy side
baneberry are smaller than those of black cohosh and there- chain. Xylosides and arabinosides of cimigenol, which is
fore are not often harvested by wildcrafters (Fletcher, per- 16,23:16,24-diepoxy-9,19-cyclo-9β-lanostane-3,15,25-triol,
sonal communication to AHP; unreferenced). In the Pacific are also present (Bedir and Khan 2000; Corsano 1965;
Northwest, Actaea elata (syn. Cimicifuga elata) is collected Corsano and others 1965; Corsano and Piancatelli 1967;
for medicinal use (Moore 1993). There is no published Shao and others 2000). Cimicifugoside is a xyloside of
information on chemical means of distinguishing the rhi- cimigenol and possesses a 7,8-didehydro-acetylacteol as its
zomes of any of the above species. However, work using aglycone (Corsano and Piancatelli 1967; Kusano and others
random amplified polymorphic DNA (RAPDs) to identify 1977). Other cycloartane-type triterpene glycosides,
plant taxa both at (Xu and others 2001) and below (Novy cimiracemosides E, G, and H, cimiaceroside A, and acetyl
and others 1994) the species level make it likely that such shengmanol xyloside, have also been identified (Bedir and
DNA testing would be useful in the identification of Actaea Khan 2000; Shao and others 2000).
species. The work by Xu and others (2001) was able to dis- *This compound is incorrectly cited in the literature and on commercial
tinguish black cohosh from other medicinal plant species products as 27-deoxyactein. When the carbons in this compound are count-
ed according to the guidelines set by the International Union of Pure and
using RAPDs. Applied Chemistry (IUPAC), its correct name is 23-epi-26-deoxyactein
Various Asian species of Actaea are sold as Chinese (Chen and others 2002).
Cimicifuga, including sheng ma (A. cimicifuga syn. ** Cimiracemoside A was originally identified by Bedir and Khan (2000).
Cimicifuga foetida), xing an sheng ma (A. dahurica syn. C. This compound appears to be the same as the cimiracemoside F and dif-
ferent from the cimiracemoside A described in the work by Shao and oth-
dahurica), and da san ye sheng ma (A. heracleifolia syn. C. ers (2000). Publication priority dictates that the version of cimiracemoside
heracleifolia; also known as guan sheng ma) (McGuffin and A in Bedir and Khan (2000) be accepted.
others 2000). Guang dong sheng ma, a member of the
Serratula genus, is a common substitute for sheng ma in the Aromatic Acids
Asian market. While these species are generally not found as Ferulic acid, isoferulic acid, and acyl caffeic acid. The fol-
adulterants of black cohosh, they are mentioned here due to lowing five hydroxy cinnamic acid esters of fukiic acid and
similarities in medicinal use and the increasing overlap of piscidic acid were isolated from the aqueous ethanolic extract
the Asian and American herb markets. of black cohosh: 2-E-caffeoylfukiic acid (fukinolic acid), 2-E-
feruloylfukiic acid (cimicifugic acid A), 2-E-isoferuloylfukiic
Preparations acid (cimicifugic acid B), 2-E-feruloylpiscidic acid (cimi-
Whole and powdered dried herb, as well as hydroalcoholic cifugic acid E), and 2-E-isoferuloylpiscidic acid (cimicifugic
and dry extracts, are commercially available as singles and in acid F) (Hagels and others 2000; Kruse and others 1999).
combination products. A number of black cohosh extracts are
marketed as yielding 2.5% triterpene glycosides calculated as Flavonoids
27-deoxyactein (correctly named 23-epi-26-deoxyactein, see Three flavonoids, biochanin A, formononetin, and
Constituents). Different manufacturers include varying num- kaempferol, have been reported to occur in black cohosh
bers of compounds in this calculation. Therefore, this decla- rhizome but have subsequently not been detected. The pres-
ration of contents is not sufficient for ensuring that all prod- ence of biochanin A was reported by McCoy and Kelly
ucts are similarly characterized (see Analytical). (1996) but was not found by Hagels and others (2000).
Fluid Extract (1:1): Kaempferol was isolated by Schmitz (1993, cited in Hagels
1:1 g/mL prepared in 60% (V/V) ethanol*. and others 2000) but was later found only in the aerial por-
1:1 g/mL prepared in 71%-78% (V/V) alcohol (NF 1936). tions of the plant, not in the rhizome and roots (Hagels and
Dry Extract (1:1): others 2000). Formononetin was reported to be present by
1:1 g/mL prepared in 40% (V/V) isopropanol*. Jarry and others (1985) and Wagner and Bladt (1996).
Tincture (1:5): However, five recent studies have failed to detect it in crude
1:5 g/mL prepared in 79%-85% (V/V) alcohol (NF 1936). material or preparations (Fabricant and others 2001; Hagels
* Formulation based on the commercial product Remifemin® (Schaper and others 2000; Kennelly and others 2001; McCoy and
and Brümmer), delivering 20 mg of black cohosh per tablet or per 20 Kelly 1996; Struck and others 1997). The presence of these
drops.
three compounds has been considered to be clinically rele-
vant due to their potential phytoestrogen activity. It appears
clear that formononetin is not present in black cohosh. The
CONSTITUENTS absence or presence of biochanin A and kaempferol require
Triterpene Glycosides further confirmation.
Black cohosh contains at least 20 known cycloartane-type Other Constituents
triterpene glycosides. The major constituents are actein
(Corsano and others 1969; Linde 1964, 1967a, 1967b, 1968; Tannins, resins, fatty acids (oleic, linoleic, linolenic,
Radics and others 1975), 23-epi-26-deoxyactein* (Chen and palmitic), starch, and sugars (Beuscher 1996; Harnischfeger
others 2002; Linde 1968) and cimiracemoside A** (Bedir and Stolze 1980; List and Hörhammer 1973).

American Herbal Pharmacopoeia™ • Black Cohosh Rhizome • 2002 9


Discussions of these issues are included below.
As noted in the Constituents section above, the nomen-
clature of a number of compounds in black cohosh have
been cited in the literature inconsistently. Some of these
inconsistencies were reflected in the original INA method
and are pointed out below. More importantly, in the original
method total triterpene glycoside content was calculated
based on response factors that were relative to 23-epi-26-
deoxyactein (27-deoxyactein). This was done due to the gen-
eral lack of availability of the reference standards and for
ease of calculation. More reference standards were included
in the additional validation work and it was determined that
the response factors are specific to each compound and need
to be quantified individually. Additionally, retention times
are not reliable for determining peak identity because they
may vary due to the complexity of the matrix and the nature
of chromatography. The use of known reference standards
or alternative methods of chemical identification, such as
mass spectroscopy, should be employed for peak identifica-
tion. As presented herein, the method is applicable for
ensuring batch-to-batch consistency of a product. It should
only be used to quantify triterpene glycosides in sample
matrices against known reference standards and individually
calculated response factors analyzed under the same chro-
matographic conditions. The INA has revised their method
according to this new information.

High Performance Thin Layer


Chromatography (HPTLC) for the
Identification of Black Cohosh
1) Analysis of Triterpene Glycosides
Sample Preparation
Place 0.5 g of powdered black cohosh rhizome in a round-
bottom flask, add 5 mL methanol, sonicate for 10 minutes,
Figure 7 Primary constituents of black cohosh then filter through filter paper. Wash the filter 4 times with
1 mL methanol. Place the combined filtrates into a 10-mL
volumetric flask and adjust to volume with methanol.
Transfer 1 mL of the solution into a small sample vial. This
is the test solution.
A N A LYT I C A L Standard Preparation
Dissolve 2.5 mg of actein in 10 mL of ethanol and water
The thin layer chromatography (TLC/HPTLC) method (6:4). Dissolve 1 mg of 23-epi-26-deoxyactein in 10 mL of
presented provides a triterpene glycoside and organic acid methanol. These are the standard solutions.
fingerprint of black cohosh developed by CAMAG
(Muttenz, Switzerland). The high performance liquid chro- Reagent Preparation
matography (HPLC) method presented utilizes an evapora- Anisaldehyde-sulfuric acid reagent: 5 mL of 98% H2SO4 are
tive light scattering detector (ELSD) and was initially vali- carefully added to 95 mL of an ice-cooled mixture of 340 mL
dated by the Methods Validation Program (MVP) of the methanol, 40 mL acetic acid, and 2 mL anisaldehyde.
Institute for Nutraceutical Advancement (INA). Its primary Note: This reagent is colorless and should be stored in the refrigerator. If a
color develops the reagent must be discarded.
purpose is for the quantitation of triterpene glycosides in
black cohosh root raw material and extract. The method was Chromatographic Conditions
subjected to further validation by one American Herbal Stationary Phase:
Pharmacopoeia™ collaborating laboratory. This validation HPTLC plates 10 x 10 or 20 x 10 cm silica gel 60 F254
work has provided additional information that must be con- (Merck or equivalent).
sidered by individual laboratories and product marketers Mobile Phase:
when analyzing black cohosh or evaluating products. Ethyl formate:toluene:formic acid (30:50:20).

10 American Herbal Pharmacopoeia™ • Black Cohosh Rhizome • 2002


Sample Application: Results:
2 µL of the test and standard solutions are applied each Compare to the chromatograms provided.
as an 8-mm band with 4 mm distance between bands. Note: The chromatograms were developed on HPTLC plates which allow
Application position should be 8 mm from the lower for better separation, sharper zones, reduced development times, and
require less solvent consumption than standard TLC plates. The method
edge of the plate. can also be run with standard TLC plates adjusting the sample volume as
Development: needed.
10 x 10 or 20 x 10 Twin Trough chamber (CAMAG or
equivalent), pre-saturation for 10 minutes (with filter
paper), 5 or 10 mL, respectively, developing solvent per
trough. Developing distance 70 mm from the lower
edge of the plate. Dry plate in a stream of warm air.
Detection:
Immerse plate in the reagent for one second, dry in a
stream of cold air. Heat plate at 105 ˚C for one minute.
Examine plate under white light.
Results:
Compare to the chromatograms provided.
2) Analysis of Organic Acids
Sample Preparation
Place 0.5 g of powdered black cohosh rhizome in a round-
bottom flask, add 5 mL of ethanol and water (1:1), sonicate
for 10 minutes, then filter through filter paper. Wash the fil-
ter 4 times with 1 mL ethanol (95%) and water (1:1). Place
the combined filtrates into a 10-mL volumetric flask and
adjust to volume with ethanol and water (1:1) and mix well.
Transfer 1 mL of the solution into a small sample vial. This
Figure 8 Anisaldehyde-sulfuric acid reagent, white light
is the test solution.
Standard Preparation Figure 8 HPTLC chromatogram of triterpene glycosides in black
Dissolve 0.5 mg of caffeic acid in 10 mL of methanol. This cohosh (Actaea racemosa)
is the standard solution. Lane 1: Wildcrafted black cohosh rhizome.
Reagent Preparation Lane 2: Actein.
Natural Products Reagent (NP Reagent): Dissolve 0.5 g Lane 3: 23-epi-26-deoxyactein.
diphenylboric acid aminoethylester in 100 mL methanol.
Discussion of Chromatogram:
Chromatographic Conditions 8) Anisaldehyde-sulfuric acid reagent, white light: The standards
Stationary Phase: actein (Lane 2) and 23-epi-26-deoxyactein (Lane 3) show violet
HPTLC plates 10 x 10 or 20 x 10 cm silica gel 60 F254 bands at Rf = 0.41 and 0.38, respectively. Black cohosh rhizome
(Merck or equivalent). (Lane 1) shows several violet bands in the middle of the chro-
Mobile Phase: matogram and some yellow-brown bands near the start line. There
Ethyl formate:toluene:formic acid (30:50:20). are two bands corresponding in color and Rf to actein and 23-epi-
Sample Application: 26-deoxyactein.
2 µL of the test and standard solutions are applied each
as an 8-mm band with 4 mm distance between bands.
Application position should be 8 mm from the lower
edge of the plate.
Development:
10 x 10 or 20 x 10 Twin Trough chamber (CAMAG or
equivalent), pre-saturation for 10 minutes (with filter
paper), 5 or 10 mL, respectively, developing solvent per
trough. Developing distance 70 mm from the lower
edge of the plate. Dry plate in a stream of warm air.
Detection:
a) UV 254 nm.
b) NP reagent: The dried plate is sprayed with NP
reagent and examined under UV 366 nm.

American Herbal Pharmacopoeia™ • Black Cohosh Rhizome • 2002 11


Figure 9 Anisaldehyde-sulfuric acid reagent, white light

Figure 9 HPTLC chromatogram of triterpene glycosides in various


samples of black cohosh (Actaea racemosa), yellow cohosh
(Actaea podocarpa syn. Cimicifuga americana), and Chinese
species of Actaea and Serratula Figure 10 Anisaldehyde-sulfuric acid reagent, white light
Lane 1: Chinese Guang Dong sheng ma rhizome Figure 10 HPTLC chromatogram of triterpene glycosides in black
(Serratula sp.). cohosh (Actaea racemosa) raw material and commercial products
Lane 2: Wildcrafted black cohosh rhizome, sample A.
Lane 1: Wildcrafted black cohosh rhizome.
Lane 3: Chinese sheng ma rhizome (Actaea sp.).
Lane 2: Actein.
Lane 4: Wildcrafted yellow cohosh rhizome. Lane 3: 23-epi-26-deoxyactein.
Lane 5: Wildcrafted black cohosh rhizome, sample B. Lane 4: Combination black cohosh rhizome dry extract and pow-
Lane 6: Actein. dered rhizome product.
Lane 7: 23-epi-26-deoxyactein. Lane 5: Black cohosh rhizome dry extract.

Discussion of Chromatogram: Discussion of Chromatogram:


9) Anisaldehyde-sulfuric acid reagent, white light: The standards 10) Anisaldehyde-sulfuric acid reagent, white light: The standards
actein (Lane 6) and 23-epi-26-deoxyactein (Lane 7) show violet actein (Lane 2) and 23-epi-26-deoxyactein (Lane 3) show violet
bands at Rf = 0.41 and 0.38, respectively. These appear in the black bands at Rf = 0.41 and 0.38, respectively. These appear in the black
cohosh samples in Lanes 2 and 5, with an additional violet band cohosh sample in Lane 1 and in the two commercial preparations in
directly above the Rf of actein. Chinese sheng ma (Lane 3) shows a Lanes 4 and 5. In all samples, two green bands can be observed in
weak band at the Rf of actein and an additional violet band directly the lower Rf region above the start line and one or two violet bands
above it with no band at the position of 23-epi-26-deoxyactein. above the bands of the reference standards. The chromatogram
Yellow cohosh (Lane 4) shows a violet band at the Rf of 23-epi-26- demonstrates that the two preparations (Lanes 4 and 5) contain the
deoxyactein and three additional violet bands below it, with no band same compounds as the raw material (Lane 1), but in differing con-
at the position of actein. Chinese Guang Dong sheng ma (Lane 1) centrations.
shows neither standard. Samples in Lanes 1, 2, 4, and 5 all have two
to three yellow-green bands at and just above the start line.

12 American Herbal Pharmacopoeia™ • Black Cohosh Rhizome • 2002


Figure 12b NP reagent, UV 366 nm

Figures 12a-b HPTLC chromatograms of plant acids in various


samples of black cohosh (Actaea racemosa), yellow cohosh
Figure 11a UV 254 nm Figure 11b NP reagent, UV 366 nm (Actaea podocarpa syn. Cimicifuga americana), and Chinese
species of Actaea and Serratula
Figures 11a-b HPTLC chromatograms of plant acids in black Lane 1: Chinese Guang Dong sheng ma rhizome (Serratula sp.).
cohosh (Actaea racemosa) Lane 2: Wildcrafted black cohosh rhizome, sample A.
Lane 1: Wildcrafted black cohosh rhizome. Lane 3: Chinese sheng ma rhizome (Actaea sp.).
Lane 2: Caffeic acid. Lane 4: Wildcrafted yellow cohosh rhizome.
Lane 5: Wildcrafted black cohosh rhizome, sample B.
Discussion of Chromatograms: Lane 6: Caffeic acid.
11a) UV 254 nm: The standard caffeic acid (Lane 2) gives a weak
dark band at Rf = 0.41. Black cohosh rhizome (Lane 1) shows a weak Discussion of Chromatograms:
band corresponding in color and Rf to caffeic acid, as well as a 12a) UV 254 nm: The standard caffeic acid (Lane 6) is barely visible
weak band at Rf = 0.47 and two other bands near the start line at Rf in this chromatogram at Rf = 0.41. A band at the same Rf as caffeic
= 0.21 and 0.09. acid is in both black cohosh samples (Lanes 2 and 5). The other
species show either no band (Lane 1) or only very weak bands
11b) NP reagent, UV 366 nm: The standard caffeic acid (Lane 2) (Lanes 3 and 4). In the Chinese Guang Dong sheng ma (Lane 1), only
shows a sharp fluorescent blue band at Rf = 0.41. Black cohosh rhi- a single band is visible in the lower Rf region. Characteristic bands
zome (Lane 1) shows several bands, most of them with blue fluo- are seen in the black cohosh samples (Lane 5 and weaker on Lane
rescence. One of the bands corresponds in color and Rf to caffeic 2). At least six bands are visible in the black cohosh sample on Lane
acid. It also shows a green band at Rf = 0.26 and blue bands at Rf = 5. Additional bands are seen in the middle Rf region of the yellow
0.22, 0.12, and 0.09. cohosh sample (Lane 4).

12b) NP reagent, UV 366 nm: The standard caffeic acid (Lane 6)


shows a sharp fluorescent blue band at Rf = 0.41 which appears in
the black cohosh samples (Lanes 2 and 5) but not in any of the other
species (Lanes 1, 3, 4). Chinese Guang Dong sheng ma (Lane 1),
Chinese sheng ma (Lane 3), and both black cohosh samples (Lanes 2
and 5) have a fluorescent blue band at the start position. Chinese
sheng ma and yellow cohosh (Lanes 3 and 4) have characteristic flu-
orescent blue bands near Rf = 0.2, with weaker blue bands below
these. Both black cohosh samples (Lanes 2 and 5) have a character-
istic fluorescent blue band above the start position, with several
weaker bands above these.

Figure 12a UV 254 nm

American Herbal Pharmacopoeia™ • Black Cohosh Rhizome • 2002 13


Discussion of Chromatograms:
13a) UV 254 nm: The standard caffeic acid (Lane 2) shows a weak
dark band at Rf = 0.41. At least six bands, one corresponding to caf-
feic acid and one above it, are visible in the black cohosh rhizome
sample (Lane 1). The two black cohosh preparations (Lanes 3 and 4)
display a similar profile including the presence of caffeic acid and
several bands with poor separation in the lower Rf region. Two of
the bands in the lower Rf region are sharper in the raw material
(Lane 1) compared to the preparations (Lanes 3 and 4), indicating a
higher concentration of compounds in the raw material.

13b) NP reagent, UV 366 nm: The standard caffeic acid (Lane 2)


shows a sharp fluorescent blue band at Rf = 0.41, which also
appears in all of the samples (Lanes 1 and 3-4) along with an addi-
tional weak blue band above it. Two of the bands in the lower Rf
region are sharper in the raw material (Lane 1) compared to the
preparations (Lanes 3 and 4), indicating a higher concentration of
compounds in the raw material.

High Performance Liquid Chromatography


(HPLC) for the Determination of Triterpene
Figure 13a UV 254 nm Glycosides
Reagents
Water, acetonitrile, methanol: HPLC grade
Formic acid: 98%
Ethanol: denatured HPLC grade
Sample Preparation
a) Raw material (rhizome and roots): Accurately weigh 1.0
g (± 0.01 g) of ground plant material and place into a 50-mL
centrifuge tube. Add exactly 40 mL of 50:50 (V/V)
ethanol:water to the tube and shake on an orbital or wrist
action shaker (or equivalent) for 24 hours at room tempera-
ture. Filter, place into an HPLC vial, and cap.
b) Extract: Accurately weigh 300 mg (± 0.01 mg) of pow-
dered extract and place into a 10-mL volumetric flask. Add
approximately 7 mL of methanol and sonicate for 30 min-
utes with frequent shaking. Cool to room temperature,
dilute to volume with methanol, and mix well. Filter, place
into an HPLC vial, and cap.
Standard Preparation
Accurately weigh 5 mg (± 0.01 mg) of 23-epi-26-deoxyactein
(“27 deoxyactein”, available from Addipharma, Hamburg,
Germany and Chromadex Inc, Laguna Hills, CA) and place
into a 10-mL volumetric flask. Add approximately 7 mL of
Figure 13b NP reagent, UV 366 nm methanol and sonicate until completely dissolved. Cool to
room temperature, dilute to volume with methanol, and mix
Figures 13a-b HPTLC chromatograms of plant acids in black well. This is the 500 µg/mL stock standard. Dilute the stock
cohosh (Actaea racemosa) raw material and commercial products standard with methanol to create a minimum of a 4-point
standard calibration curve that includes the stock standard.
Lane 1: Wildcrafted black cohosh rhizome.
Suggested standard dilutions include 1:25, 1:10, and 2:10;
Lane 2: Caffeic acid.
further dilutions may be made for additional data points.
Lane 3: Combination black cohosh rhizome dry extract and pow-
The AOAC International recommends a minimum of six
dered rhizome product.
data points for accurately determining the linear response of
Lane 4: Black cohosh rhizome dry extract.
a single analyte (Weirmont 1985).
Note: Refrigerate all reference standards.

14 American Herbal Pharmacopoeia™ • Black Cohosh Rhizome • 2002


Linearity Range: Detection:**
Prepare a calibration curve with the origin ignored using Richard Scientific/Seder Sedex-75 ELSD
the natural log of peak area versus the natural log of con- (or equivalent)
centration (µg/mL). Perform linear regression analysis on Gas pressure: 2.3 bar
the data. The correlation coefficient must be ≥ 0.995. Temperature: 50 ˚C
Note: Because ELSDs are inherently non-linear, correlation coefficients Gain: 7
may be achieved by plotting peak area versus concentration (µg/mL) and Alltech 5000 ELSD:
fitting to a quadratic equation.
Gas Flow: 1.78 standard liters per minute
Chromatographic Conditions Gas Pressure: 48 psi
Column: Drift Tube Temperature: 40 ˚C
Phenomenex Prodigy ODS (3), 5 µm, 4.6 x 250 mm Low Temperature Adapter: 40 ˚C
or equivalent*. Analog Output: No attenuation
Column Temperature: Injection Volume:
Ambient (27 ± 2 ˚ C). 20 µL.
Mobile Phase: * Also validated using YMC J’ sphere ODS-M80, 4 µm, 4.6 x 250 mm.
Gradient: acetonitrile: 0.1% formic acid. ** Both detectors were used in the validation process.
Minutes % Acetonitrile % Formic Acid Peak Identification:
0 30 70 Peak Triterpene Relative
30 40 60 Number Glycoside Retention Time
60 60 40
1. Cimiracemoside F* 0.551-0.596
70 30 70
2. Epi-actein 0.770-0.820
Flow Rate:
3. Unknown 0.807-0.878
1.0 mL/minute.
4. Unknown 0.861-0.954
5. 26-deoxycimifigoside and

Figure 14 HPLC chromatogram of the triterpene glycosides in black cohosh (Actaea racemosa)*
* The names of the peaks follow the nomenclature provided by the original INA method and some are now out-dated. For correct nomenclature, see Analytical, Peak Identification.

American Herbal Pharmacopoeia™ • Black Cohosh Rhizome • 2002 15


cimiracemoside A* 0.901-0.966 THERAPEUTICS
6. Actein 0.938-0.982
7. 27-deoxyactein* 1.000 Pharmacokinetics
8. Acetyl shengmanol xyloside 1.01-1.04 Pharmacokinetic data on black cohosh are not available.
9. Cimiracemoside C and E 1.06-1.17 Work is underway, however, to quantify the in vitro move-
10. Cimicifugoside 1.15-1.24 ment of isolated actein, deoxyactein, and cimiracemoside
Note: These were the retention times determined in the original INA valida- across caco-2 cell monolayers, a model of the human intesti-
tion. Differences in chromatography and detector parameters, such as a vari- nal mucosa. Preliminary results indicate that the com-
ance in ELSD gas pressure and temperature, will affect retention times.
pounds show a linear transport across the monolayer (Khan
* The original nomenclature used by INA was maintained.
Cimiracemoside F is actually cimiracemoside A as determined by Bedir and and others 2001).
Khan (2001). The identity of the cimiracemoside A listed at peak 5 has not
been definitively determined. The 27-deoxyactein cited is more accurately Pharmacodynamics and Clinical Efficacy
known as 23-epi-26-deoxyactein (Chen and others 2001). Care must be
taken in ascertaining the accurate identity of reference standards purchased. Traditionally, black cohosh has been used for aching mus-
cles and joints, neuralgias, rheumatoid arthritis, dysmenor-
Calculation: rhea, amenorrhea, to prepare for labor (partus preparator),
Measuring the areas under the identified triterpene glyco- and as an aid in relieving pain after birth (Felter 1922; Felter
side peaks, calculate the concentration of the triterpene gly- and Lloyd 1898; King 1855; Scudder 1903). In recent years,
cosides using the following equation: black cohosh has increasingly been recommended for use
in the treatment of the physical and psychological symptoms
of menopause (climacteric complaints) and dysmenorrhea.
Concentration (µg/mL) = exp ln (peak area) – intercept
slope The European literature divides physical menopausal com-
plaints into “neurovegetative” symptoms, including hot
ln peak area = the natural log of the area under the peak flashes, cardiac complaints, fatigue, vertigo, and outbreaks
of interest of sweating, and “somatic” symptoms, including muscle
intercept = the intercept from the linear fit of the cal- pain and spasm, joint pain, urinary incontinence, vaginal
ibration curve dryness, and atrophy of the vaginal epithelium. However,
slope = slope from the linear fit of the calibration various authors group symptoms differently, even when
curve referring to the same assessment scales (eg Kupperman
Note: While this calculation uses the natural log (ln), HPLC chromatog- Menopause Index). The terms used in the individual stud-
raphy software uses the base 10 log. Since the natural log is related to the ies have been retained. Psychological symptoms of
base 10 log by the equation ln / base 10 log = 2.303, the common factor menopause include depression, anxiety, nervousness, irri-
2.303 cancels out and the two expressions are equivalent. This can be used
for automated calibration. tability, forgetfulness, sleep disturbances, and depressed
libido.
Quantitative Standards The decline of estrogen and progesterone levels that
Foreign Organic Matter: accompanies menopause is responsible for many climac-
Not to exceed 2% m/m, determined on 100-500 g dried teric symptoms. During the normal reproductive cycle the
root spread out in a thin layer (BHP 1996). relatively high amounts of estrogen that are present result in
Stem Bases: a relatively low level of luteinizing hormone (LH) and folli-
Not to exceed 5% (BHP 1996). cle-stimulating hormone (FSH). A result of this is a prolifer-
Total Ash: ation of the vaginal and uterine epithelium and mainte-
Not to exceed 10%, determined on 1 g of powdered nance of bone density. Problems that may accompany
dried root evenly distributed in a crucible (BHP 1996). decreased levels of estrogen during and after menopause
Acid-insoluble Ash: include hot flashes, mood swings, sleep disturbances, bone
Not to exceed 4%, calculated with reference to 100 g of loss, vaginal atrophy, and a decline in libido.
dried root (BHP 1996). The exact mechanism by which black cohosh elicits its
Water-soluble Extractive: effects on menopausal symptoms has not been elucidated.
Not less than 10% (BHP 1996)*. While some have proposed that its effectiveness is mediated
* Determined on 5 g of powdered dried root macerated with 100 mL chlo-
by an inhibitory effect on the hypothalamus (Einer-Jensen
roform water (2.5 mL chloroform mixed with 900 mL water and diluted with and others 1996) or an effect on neurotransmitters (Löhning
water to 1000 mL) for 24 hours shaking frequently the first 6 hours. Let stand and others 1999; Löhning and Winterhoff 2000; Seidlová-
18 hours. Filter rapidly, evaporate 20 mL of filtrate to dryness in a tared, flat-
bottom, shallow dish at 105 ˚C to constant weight. Calculate the percentage
Wuttke and Wuttke 2000), others have suggested that it has
of water-soluble extractive with reference to the dry herb (BP 1993). a direct estrogenic effect. These latter researchers suggest
that black cohosh contains phytoestrogens, estrogen-like
compounds found in plants (Jarry and others 1999; Kruse
and others 1999; Seidlová-Wuttke and Wuttke 2000; Wuttke
and others 2000). To date, no known phytoestrogens have
been unequivocally identified in black cohosh (see
Constituents). Kruse and others (1999), however, reported

16 American Herbal Pharmacopoeia™ • Black Cohosh Rhizome • 2002


an estrogenic effect of the caffeic acid derivative fukinolic fibroids (Grady and others 1995). The association of such
acid isolated from black cohosh (see Hormonal and Central risks with HRT underscores the value of identifying alterna-
Nervous System Effects). An estrogenic activity of black tive, non-hormonal therapies. More work needs to be done to
cohosh is supported by some preclinical and clinical trials determine whether black cohosh has estrogenic effects, and if
and not by others. Some of the variation in preclinical so, on which tissues. Long-term studies are lacking. If black
results may be attributable to differences in the black cohosh extract is estrogenic and competes with estradiol more
cohosh product used, though this is untested. In the clinical strongly at ERβ than ERα receptors, it may elicit many of the
trials, almost all of which used the same product, it appears beneficial effects of estrogen without the accompanying risks
that the variation in results may have begun to be resolved and hence be of great benefit as a substitute for HRT.
based upon a broader understanding of the diversity of estro- However, in pharmaceutical research, no risk-free SERM has
gen receptors (see Barnes 1998) and the hypothetical selec- yet been identified.
tive binding of compounds in black cohosh to some of these Most pharmacological research has focused on the use of
receptors and not to others. black cohosh for menopausal symptoms. When evaluating
There are at least two classes of estrogen receptors: the data from the studies presented, it should be noted that a
alpha receptors (ERα) and beta receptors (ERβ). The ERα placebo effect is common in studies of menopausal symp-
are concentrated in the reproductive organs, the pituitary, toms. To date, two placebo-controlled studies investigating
and the kidneys, while the ERβ are found mainly in the the use of black cohosh extract in menopausal women have
brain, bone, vasculature, and prostate. Estrogenic effects found a large placebo effect (Jacobson and others 2001; Stoll
mediated by the ERα receptors include the suppression of 1987). It is therefore extremely important that clinical studies
LH and FSH, as well as the proliferation of the vaginal and are double-blind and placebo-controlled, especially when
uterine epithelium. Estrogenic effects mediated by the ERβ they involve self-reported symptom endpoints. Most of the
receptors include the prevention of bone loss. Some studies conducted to date do not meet these criteria, creating
researchers have suggested that black cohosh may primarily the possibility of misleading positive effects following treat-
affect the ERβ found in the bone and vasculature, rather ment with black cohosh extract.
than the ERα found in the pituitary and reproductive
organs (Jarry and others 1999; Seidlová-Wuttke and others Effects on Menopausal and Premenstrual
2000; Wuttke and others 2000). Such selectivity in phytoe- Symptoms
strogens has been documented before (Kuiper and others Human Clinical Studies
1998) and has caused some researchers to refer to phytoe- Of the studies available on menopausal symptoms, two have
strogens as selective estrogen receptor modulators been randomized, double-blind, and placebo-controlled
(phytoSERMs) (Jarry and others 1999). In addition, phytoe- (Jacobson and others 2001; Stoll 1987). One of these
strogens may act variously as estrogen agonists or antago- addressed treatment-induced menopausal symptoms in breast
nists, depending upon dose and to which receptor type they cancer survivors (Jacobson and others 2001). An additional
bind. The results of preclinical research addressing these study was open-label and placebo-controlled (Földes 1959),
hypotheses, however, are mixed. while another was double-blind, randomized, and Good
Of theoretical importance in this controversy is the Clinical Practice (GCP)-compliant (Liske and others 2000).
safety of black cohosh in women with estrogen-sensitive One older case series investigated the effect of black cohosh
conditions, such as breast cancer. As the medical use of hor- on premenstrual symptoms (Görlich 1962), while two looked
mone therapies has increased over the years, the dangers of at effects on dysmenorrhea and menstrual irregularity
long-term treatment with estrogen have become more obvi- (Görlich 1962; Heizer 1960).
ous. In the 1950s and 1960s, estrogen replacement became Most clinical research on black cohosh has been done
the main treatment for menopause. Not until many years using the commercial preparation Remifemin® (Schaper and
later were the rising rates of endometrial cancer tied direct- Brümmer, Germany). Remifemin® comes in liquid (ethanol
ly to estrogen replacement therapy. Hormone replacement extract, 60% by volume) and tablet form (isopropanol extract,
therapy (HRT), a combination of estrogen and progestin, 40% by volume). Earlier studies employed a previous formu-
was developed to counteract the dangerous effect of treat- lation of Remifemin® at dosages reported to be equivalent to
ment by estrogens alone. There is evidence, however, that 48-140 mg of dried herb daily (Boblitz and others 2000),
HRT, particularly at high doses and in long-term treatment, though the individual studies do not provide full characteri-
may be linked to higher rates of cancer as well, particularly zation of the product used. The current recommended ther-
breast cancer (Beral and others 1997; Dupont and Page apeutic dosage of the liquid is 20 drops twice daily and that of
1991; Henrich 1992; Schairer and others 2000; Steinberg the tablet is one tablet twice daily. Both dosages of the current
and others 1991). Along with the possible dangers for a preparation correspond to 40 mg of dried herb daily. A recent
healthy woman, HRT is also contraindicated for many study reported that both the previous and current formula-
women with specific health conditions. These include tions have equivalent clinical efficacy at their respective
breast cancer, endometrial cancer, malignant melanoma, dosages, supporting the lower dosage recommended for the
unexplained bleeding, liver disease, gallbladder disease, current formulation (Liske and others 2000). One of the
edema, severe diabetes mellitus, endometriosis, venous recent clinical studies uses the black cohosh extract
thrombosis, thrombophlebitis, pancreatitis, and uterine Klimadynon®/Menofem® (58% by volume, Bionorica AG

American Herbal Pharmacopoeia™ • Black Cohosh Rhizome • 2002 17


Germany), administered at two capsules daily, a dosage resulting in withdrawal from treatment; another woman
equivalent to 40 mg dried herb daily. This product contains complained of headaches but continued treatment.
the black cohosh extract BNO 1055 (Bionorica AG) which The effectiveness of black cohosh in treating hot flashes
is used in several of the preclinical studies. Many of the clin- in breast cancer survivors has recently been studied in a ran-
ical studies compare the effects of black cohosh extract with domized, double-blind, placebo-controlled trial (Jacobson
standard therapeutic dosages of estrogen. and others 2001). Black cohosh extract (Remifemin®, 1
There have been many standard scales used by black tablet twice daily for 60 days, equivalent to 40 mg daily of
cohosh researchers to assess the severity of menopausal dis- dried herb; n = 42) or placebo (n = 43) was administered to
orders. These include the Kupperman Menopause (KM) 85 women. Fifty-nine of the women were concurrently using
Index (outdated, although still occasionally used), the the estrogen antagonist tamoxifen (n = 29 in the black
Clinician’s Global Impression scale (CGI), the Profile of cohosh group; n = 30 in the placebo group). Patients
Mood States scale (POMS), the Hamilton Anxiety scale assessed the intensity of their hot flashes (mild, moderate, or
(HAM-A), and the Self-assessment Depression scale (SDS). severe) for the three days prior to treatment, on days 27-30,
The KM Index lists both psychological and neurovegetative and again on days 57-60. Additional outcome measures used
symptoms of menopause which patients are asked to self- to assess overall health and well-being included a Visual
assess. A score lower than 15 indicates that treatment was Analogue scale (VAS) and an unspecified menopausal symp-
successful. The CGI scale, filled out by the physician, assess- tom index using a Likert scale, both of which were scored by
es the degree of severity of a disease before, during, and after patients before and after treatment. Serum levels of FSH and
treatment. The POMS scale is a self-evaluation scale that LH were also monitored in selected patients (FSH: n = 16 in
gives a quantitative measure to the influence of therapy on black cohosh group, n = 17 in placebo group; LH: n = 9 in
mood state. The HAM-A scale, filled out by the physician, both groups). Over the study period, the median number
measures both psychological and somatic anxiety, producing and intensity of hot flashes decreased by approximately 27%
a total score reflecting a patient’s degree of anxiety. The SDS in both the black cohosh and placebo groups. No significant
scale is sometimes used in conjunction with the HAM-A and difference between groups was observed even after adjusting
is filled out by the patient. It includes 20 depression criteria for differences in tamoxifen usage, indicating that black
measured by frequency and is useful in quantifying depres- cohosh was no more effective than placebo in the treatment
sive states. of hot flashes in this study. Neither the VAS nor the
In order to test possible alternatives to long-term estro- menopausal symptom index found a significant effect of
gen therapy, Stoll (1987) performed a randomized, double- black cohosh compared to placebo, except with respect to
blind, placebo-controlled study investigating the effects of the symptom of sweating, which showed a significantly
conjugated estrogens and black cohosh extract on greater improvement in the black cohosh group (P = 0.04).
menopausal symptoms in 80 women (ages 46-58 years). No significant changes in serum FSH and LH levels were
Twenty-nine patients received 0.625 mg of conjugated estro- recorded. Small sample sizes and the confounding effect of
gen, 26 received black cohosh extract (Remifemin®, 4 tamoxifen, known to cause hot flashes, may have influenced
tablets, each containing 2 mg of extract), and 20 were given these results. The authors stated that the short duration of
a placebo, each daily for 12 weeks. Outcome measures were their study might have limited its findings (Jacobson and oth-
the KM Index, the HAM-A scale, and the vaginal maturity ers 2001).
index (VMI, a measure of estrogenic effects on the vaginal In this study, adverse events occurred in a total of 13
epithelium). Physical and psychological symptoms were test- patients. Ten “minor” events included constipation, weight
ed every four weeks and the VMI was measured before and gain, endometrial hyperplasia, another unspecified endome-
after treatment. The conjugated estrogens showed no differ- trial condition necessitating dilatation and curettage, indi-
ence compared to placebo although they were administered gestion, and vaginal bleeding in the black cohosh-tamoxifen
at a standard dose used in estrogen supplementation thera- group; arrhythmia and cramping in the black cohosh-no
py. After 12 weeks, the KM Index among patients treated tamoxifen group; and swollen finger and abdominal rash in
with Remifemin® had dropped below 15 (P < 0.001, com- the placebo-tamoxifen group. Endometrial hyperplasia is a
pared to placebo). On the HAM-A scale, scores in the risk factor for endometrial cancer. Two serious adverse
Remifemin® group also dropped dramatically compared to events, an endometrial condition necessitating a hysterecto-
those in the other groups (P < 0.001). At the end of treat- my and breast cancer recurrence, were reported in the black
ment, only patients in the Remifemin® group showed a sig- cohosh-tamoxifen treatment group. Tamoxifen is known to
nificant increase in the number of estrogenized cells of the cause endometrial abnormalities. The patient with recurrent
vaginal epithelium (P < 0.01). Minor adverse events were breast cancer had an unreported increase in carcinoembry-
reported by 12 (46%) patients in the Remifemin® group, onic antigen before entering the study. An appendectomy
including headache, weight problems, mastalgia, a stimulant was reported in the placebo-tamoxifen group. According to
effect, heaviness of the legs, and an increase in symptoms the researchers, none of the adverse events appeared to be
existing prior to treatment. In those receiving conjugated related to treatment.
estrogens, one patient reported weight problems and anoth- Wuttke and others recently performed a randomised,
er tachycardia. In the placebo group, two women had strong double-blind, placebo-controlled, GCP-compliant study to
adverse events, including headache and weight problems, investigate the clinical effects of black cohosh extract

18 American Herbal Pharmacopoeia™ • Black Cohosh Rhizome • 2002


(Klimadynon®/Menofem®, equivalent to 40 mg dried herb nal cytology were reported. The authors interpreted these
daily for 12 weeks) on menopausal complaints, hormone results as confirming the clinical equivalency of the two
levels (estradiol, LH, FSH), and bone metabolism in 80 dosage forms for relieving menopausal symptoms and as evi-
postmenopausal women (Gorkow, Marz, and Christoffel, dence that the black cohosh product used does not have an
personal communication to AHP; unreferenced). estrogenic effect on hormone levels and vaginal tissue.
Preliminary unpublished findings indicate that there was a When the subgroup of premenopausal women was ana-
significant reduction in the frequency of waking during lyzed, the higher dose was correlated with a significantly
night in the black cohosh group compared to placebo (P = greater reduction in the KM Index compared to the low
0.04). The placebo response rate exceeded 50% in some cli- dose (P < 0.032). During the first three months, 19 unspec-
macteric complaints. No change in estradiol levels and a ified mild to moderate adverse events were reported, 12 at
decrease in FSH levels were observed in the black cohosh the higher dose and 7 at the lower dose. These affected the
group. In addition, there was a significant increase in a gastrointestinal tract (5), the central nervous system (5), the
marker for osteoblast activity (P = 0.01) and a significant breasts or female genitals (5), or remained unclassified (4).
increase in two bone turnover indices in the black cohosh One patient experienced metrorrhagia, though a biopsy
group (P ≤ 0.007), indicating a protective effect of black showed an inactive endometrium.
cohosh on bone tissue. Further evaluation of these data is In 1988, Lehmann-Willenbrock and Riedel conducted
needed following their publication. an open, randomized, comparative study of 60 women
Földes (1959) conducted a placebo-controlled, open- under the age of 40 who had undergone hysterectomies, had
label, crossover study on 41 women complaining of at least one ovary intact, and complained of menopausal
menopausal symptoms. The women were first given an symptoms. The patients were randomly divided into four
unidentified placebo for an unspecified period of time. groups (n = 15) and treated daily over six months with either
Black cohosh extract was then administered at one tablet tid estriol (Ovestin®, 1 mg), conjugated estrogens (Presomen®,
(Remifemin® , equivalency unknown) until it became effec- 1.25 mg), estrogen-gestagen sequential therapy
tive (at approximately one week), at which point the dosage (Trisequens®, one tablet containing 2 mg estradiol and 1 mg
was reduced by an unspecified amount. Symptoms were norethindrone acetate), or black cohosh extract
alleviated more frequently during the treatment phase (Remifemin® four tablets daily, each containing 2 mg of
(improvement, 31 patients; no improvement, 10) compared extract). Efficacy was measured by a modified KM Index
to the placebo phase (improvement, 4 patients; no improve- that included trophic disorders of the genitals. In addition,
ment, 37). Three patients complained of stomach pains LH and FSH levels were monitored. Patients were tested at
while taking the extract. baseline and after 4, 8, 12, and 24 weeks of therapy. The
In a recent randomized, double-blind, GCP-compliant modified KM Index decreased significantly (P < 0.01) in all
study, 152 women (ages 42-60 years) with menopausal four groups with no significant differences between groups.
symptoms were treated for three or six months with two dif- Hormonal levels were not significantly affected in any of the
ferent doses of black cohosh extract (Remifemin®) equiva- treatment groups. After six months of therapy, most patients
lent to either 39 mg (n = 74, three months; n = 57, six were symptom-free. Adverse events were not discussed.
months) or 127 mg (n = 75, three months; n = 59, six In an uncontrolled, open, prospective study by Pethö
months) dried herb daily (Liske and others 2000). (1987), 50 women who had previously received at least 1-2
Treatment effect was assessed using the KM Index, SDS, Gynodian® Depot injections (a combination of estradiol
CGI, and by measuring serum levels of LH, FSH, sex hor- and prasteron, an androgen) for menopausal symptoms were
mone-binding globulin (SHBG), prolactin, and 17-β-estra- tested to see if black cohosh extract (Remifemin®, two
diol, as well as changes in vaginal cytological parameters tablets twice daily for six months, equivalency unknown)
(degree of proliferation of the vaginal epithelium [Schmitt] could adequately replace hormone therapy. Hormone injec-
and karyopyknotic and eosinophil indices). Differences tions were to be given only if the black cohosh preparation
between dosage groups at various time points were analyzed alone was not satisfactory at controlling the symptoms.
statistically, but statistical analyses of changes in outcome Patients were checked by a physician after 2, 4, and 6
measures over the treatment period were not presented. months of treatment. Outcome measures were the number
Both doses showed similar effects in all outcome measures. of hormone injections needed, the KM Index, and the sub-
A reduction in the KM Index was apparent after two weeks, jective statements of the patients regarding their physical
three months, and six months; 70%-72% and 90% of the and psychological symptoms. Over the six-month testing
patients were considered to be “responders” at three and six period, 28 (56%) of the 50 patients requested no supple-
months, respectively (KM Index < 15). Results for the SDS mental hormones, 21 (42%) requested a single injection,
were similar, with a reduction from scores indicative of mild and 1 (2%) patient requested two injections. The KM Index
depression to normal scores (median 36-37). After three and decreased significantly from a mean of 17.6 to 9.2 over the
six months of treatment, global efficacy (CGI = 3.1) was course of the study (P < 0.001). Neurovegetative symptoms
good to very good in 80% and 90%, respectively, of the were rated by the patients as “moderately pronounced”
patients in each group. After six months of treatment, glob- before and “mild” afterwards. Psychological and somatic
al tolerability (CGI = 3.2) was good to very good in 100% of symptoms were mild at the beginning and had almost dis-
the patients. No changes in serum hormone levels or vagi- appeared after treatment. The physician rated the therapeu-

American Herbal Pharmacopoeia™ • Black Cohosh Rhizome • 2002 19


tic success as excellent (all-round improvement) for 21 black cohosh extract (Remifemin® tablets, equivalent to 136
patients, good (evident change for the better) for 20, and mg dried herb daily) given daily for approximately three
light (further treatment advised) for 9 patients. No adverse months to 28 postmenopausal women had a good to very
events occurred during treatment. good effect on 10 neurovegetative symptoms in 80% of the
In 1985, Warnecke conducted a randomized, open, patients and did not alter endometrial thickness, vaginal cell
comparative study to test the effectiveness of black cohosh status, or serum hormone levels (LH, FSH, prolactin, estra-
extract on the physical and psychological symptoms associ- diol). No adverse events were observed during treatment.
ated with menopause. The effect of black cohosh was com- Mielnik (1997) found that significant clinical improvement
pared with hormonal treatment and a psychotropic drug (KM Index < 10) occurred after one month in 26 (76%) of
(diazepam). Sixty patients (ages 45-60) with menopausal 34 menopausal women given an uncharacterized black
symptoms were allocated to three (n = 20) treatment groups. cohosh extract (4 mg daily for six months). All 34 women
One group received 40 drops of black cohosh extract twice had a KM Index > 20 at the beginning of treatment. Four of
daily (Remifemin®, equivalency unknown), another the women showed no improvement and four dropped out
received 0.625 mg daily of conjugated estrogens, and the of the study and began HRT. No adverse events were
third received 2 mg of diazepam daily. These treatments observed. In the study of Georgiev and Iordanova (1997),
were administered over 12 weeks, and patients were tested subjective symptoms evaluated using the KM Index and
before start of therapy and after 2, 4, and 12 weeks of treat- HAM-A scale were found to improve in 45 (90%) of 50 post-
ment. Outcome measures included a modified KM Index, menopausal women following administration of black
HAM-A, SDS, and CGI. Estrogenic stimulatory effects on cohosh extract (characterization and dosage undisclosed) for
the vaginal epithelium were measured using the karyopy- three months. The VMI was increased in 40% of the
knosis and eosinophil indices. At the end of therapy, using patients, while endometrial thickness did not change in any
all four symptom scales, a “highly significant” or “good” patients. Adverse events were not discussed.
effect of treatment on menopausal symptoms was seen In an older case series, Görlich (1962) reported on the
equally in all three treatment groups (P-values not given, effects of black cohosh extract (Remifemin®) in 258 women
though statistical tests were performed). After the fourth with various gynecological complaints including primary
week, vaginal cytology indices revealed a “trend” towards and secondary amenorrhea, polymenorrhea, menorrhagia,
estrogenic stimulation in both the black cohosh extract and metrorrhagia, dysmenorrhea, premenstrual symptoms, ovu-
estrogen groups. Adverse events were not discussed. lation pain, menopausal disorders, and “uterine hypopla-
In the early to mid 1980s, two other uncontrolled open sia”, as well as pregnancy. Evaluation criteria and dosage
studies were conducted on patients with menopausal symp- were largely unspecified. Overall, the extract alleviated the
toms (Daiber 1983, n = 36; Vorberg 1984, n = 50). In both majority of menstrual irregularities unless they were caused
studies, patients were treated with 40 drops of black cohosh by other primary disorders (eg hypo- and hyperthyroidism,
extract twice daily for 12 weeks (Remifemin® , equivalency hypertension). Patients with premenstrual symptoms were
unknown). The results were measured using the KM Index, given one tablet or 20 drops of the extract three times daily
the CGI scale, and in the Vorberg study only, the POMS (tid) for 12-14 days before menstruation. In 41 of the 49 sub-
scale. According to all three scales, all symptoms improved jects suffering from premenstrual symptoms, symptoms
significantly in the 12 weeks of treatment (P < 0.001). The including mood swings, impaired performance, circulatory
Daiber study did not discuss adverse events. In the Vorberg disorders, and headaches were eliminated. In 24 cases of
study, four women reported mild gastrointestinal distur- dysmenorrhea, 16 patients given 10 drops or 1/2 tablet of
bances in the early stages of therapy, although this did not extract tid (between days 10-20 of cycle) were better able to
interfere with treatment. tolerate cramping. Görlich also noticed a significant
Stolze (1982) conducted a multicenter, observational, improvement in ovulatory pain following treatment with the
cohort study on 704 women suffering from climacteric com- extract. Symptoms disappeared in 35 (85%) of the 41
plaints. The study investigated the effectiveness of black women treated for menopausal symptoms. In 18 cases of
cohosh extract (Remifemin®, 40 drops twice daily for six to morning sickness, black cohosh extract controlled vomiting;
eight weeks, equivalency unknown) on neurovegetative and antiemetic drugs were used in only two cases. The success-
psychological menopausal symptoms. Symptoms were eval- ful treatment of threatened miscarriage was reported in six
uated after 4, 6, and 8 weeks of treatment, both by the physi- women with a history of miscarriage. They took black
cian and the patient. Of the 629 evaluable patients, 75% cohosh extract (25 drops hourly until bleeding disappeared
were symptom-free or showed improvement in symptoms followed by 20 drops four times daily for four to six weeks)
after 4 weeks, while 80% showed an improvement in symp- and reported that no miscarriages occurred during treat-
toms after 6 and 8 weeks of treatment. Among the total ment. Adverse events were not discussed.
patient population, 93% reported no adverse events, while In two additional older case series, black cohosh extract
7% noted short-lived unspecified ones (mostly slight stom- (Remifemin®) was given to women with natural or surgical-
ach complaints) that did not interfere with treatment. ly-induced menopausal symptoms. In a study of Heizer
Three recent observational studies investigated the use (1960), among 66 menopausal women with intact uteri, 31
of black cohosh extract for alleviating menopausal symp- (47%) showed a positive response to the extract (30 drops or
toms. Nesselhut and Liske (1999) found that a high dose of 1-2 tablets tid for 2-18 months, equivalency unknown),

20 American Herbal Pharmacopoeia™ • Black Cohosh Rhizome • 2002


Table 3 Summary of results from clinical studies on the treatment of menopausal complaints1 with black cohosh extract
Reference Study Product, daily dosage, Outcome Results
design, n2 and treatment duration measure
Stoll 1987 DBRPCT Remifemin®;4 tablets, KMI, HAM-A Alleviation of symptoms (KMI, P < 0.001, com-
n = 26 each containing 2 mg pared to placebo; HAM-A, P < 0.001, compared
extract3; 12 weeks to placebo and estrogen groups).

Jacobson and DBRPCT Remifemin®; 2 tablets, Patient self- No change in median number and intensity of hot
others 2001 n = 42 equivalent to 40 mg assessment; flashes; no change compared to placebo except
dried herb4; 60 days VAS, MSS for symptom of sweating (VAS, MSS, P = 0.04).

Földes 1959 Open, PCTX, Remifemin®, 3 tablets3; Patient self- Greater alleviation of symptoms in black cohosh
n = 41 unspecified assessment group compared to placebo.

Liske and others DBR, GCP Remifemin®, equivalent KMI, SDS, Alleviation of symptoms in both groups (KMI <
2000 n = 57-59 to 39 and 127 mg dried CGI 15 in 90% women; SDS reduced to median of
herb; 6 months 36-37; global efficacy good to very good in 90%
patients). Results similar after 3 months.

Lehman- R, Open Remifemin®, 4 tablets, KMI Alleviation of symptoms in the black cohosh,
Willenbrock and n = 15 each containing 2 mg ovestin, presomen, and trisequens groups (P < 0.01).
Riedel 1988 extract3; 6 months

Pethö 1987 Open Remifemin®, 4 tablets3; 6 KMI, patient Alleviation of symptoms (P < 0.001). Self-
n = 50 months self- assessment indicated that symptoms decreased
assessment from moderate or mild to mild or almost gone.

Warnecke 1985 R, Open Remifemin®, 80 drops3; KMI, HAM-A, Alleviation of symptoms in the black cohosh,
n = 20 12 weeks SDS, CGI conjugated estrogen, and diazepam groups.

Daiber 1983 Open Remifemin®, 80 drops3; KMI, CGI Alleviation of symptoms (P < 0.001).
n = 36 12 weeks

Vorberg 1984 Open Remifemin®, 80 drops3; KMI, CGI, Alleviation of symptoms (P < 0.001).
n = 50 12 weeks POMS

Stolze 1982 Open Remifemin®, 80 drops3; Physician and Alleviation of symptoms in 80% of patients after
n = 629 6-8 weeks patient self- 6 and 8 weeks.
assessment

Nesselhut and Open Remifemin® tablets, unspecified Good to very good alleviation of 10 menopausal
Liske 1999 n = 28 equivalent to 136 mg symptoms in 80% of patients.
dried herb; 3 months

Mielnik 1997 Open Uncharacterized black KMI Alleviation of symptoms in 76% of patients
n = 34 cohosh extract, 4 mg after 1 month (KMI < 10).
daily; 6 months

Georgiev and Open Uncharacterized black KMI, HAM-A Alleviation of symptoms in 90% of patients.
Iordanova 1997 n = 50 cohosh extract, dose
unspecified; 3 months

Görlich 1962 Case series Remifemin® tablets, unspecified Alleviation of symptoms in 85% of patients.
n = 41 dose/duration unspecified

Heizer 1960 Case series Remifemin®, 90 drops unspecified Alleviation of symptoms in 47% of patients with
n = 66 or 3-6 tablets3; 2-18 months intact uteri and in 35% with hysterectomies.

Kesselkaul 1957 Case series Remifemin®, 60 drops3; unspecified Alleviation of symptoms in 95% of patients.
n = 63 2 weeks

Key: DBRPCT = double-blind, randomized, placebo-controlled trial; DBR = double-blind, randomized; R = randomized; PCT = placebo-controlled; X = cross over;
GCP = Good Clinical Practice compliant; CGI = Clinician’s Global Impression scale; HAM-A = Hamilton Anxiety scale; KMI = Kupperman Menopause Index;
MSS = unspecified menopausal index using the Likert scale; POMS = Profile of Mood States scale; SDS = Self-Assessment Depression scale; VAS = Visual Analogue scale.
1 Menopausal symptoms were natural, surgically induced by hysterectomy (Lehman-Willenbrock and Reidel 1988; Heizer 1960), or induced by breast cancer treatment

(Jacobson and others 2001).


2 Sample size in the black cohosh treatment group.

3 The dried herb equivalents were in the range of 48-140 mg daily (Boblitz and others 2000).

4 Some patients in each group took tamoxifen concurrently.

American Herbal Pharmacopoeia™ • Black Cohosh Rhizome • 2002 21


while 35 (53%) required hormone therapy in addition to the toms in patients treated with black cohosh extract compared
extract. Among 23 women who had had hysterectomies, 15 to placebo (Stoll 1987), as did one open placebo-controlled
(65%) required hormone therapy. In this same study, black study (Földes 1959). However, one other double-blind,
cohosh extract was not effective in treating menstrual irreg- placebo-controlled study found no effect of black cohosh
ularities (n = 16) or pain (n = 5). Adverse events were not dis- compared to placebo, except with respect to the symptom of
cussed, though general tolerability was reported to be good. sweating (Jacobson and others 2001). This study, however,
Kesselkaul (1957) found that black cohosh extract (20 drops was on breast cancer survivors and the effects of black cohosh
tid, equivalency unknown) alleviated menopausal symptoms may have been confounded with those of tamoxifen, an anti-
after two weeks of treatment in 60 (95%) patients. Three estrogenic compound. The other studies that were not place-
patients complained of gastrointestinal complaints during bo-controlled support the effectiveness of black cohosh in
treatment. treating menopausal symptoms, though the possibility of a
In addition to the clinical studies presented above, two placebo effect in these studies cannot be ruled out and the
studies have investigated the effects of a combination prod- assessment criteria of the older studies cannot be relied
uct containing extracts of black cohosh and St. John’s wort upon.
(Hypericum perforatum) on psychological and neurovegeta- Few studies have investigated the efficacy of black
tive symptoms associated with menopause. The product cohosh extract in treating conditions other than menopausal
used, Remifemin® Plus (Schaper and Brümmer), was stan- complaints, and those that exist are uncontrolled case series.
dardized to 1 mg triterpene glycosides calculated as 27- The case series by Görlich (1962) reported efficacy of black
deoxyactein (correctly named 23-epi-26-deoxyactein) and cohosh in treating premenstrual symptoms. Of the two case
0.25 mg total hypericin per coated tablet. One trial used a series looking at the effect of black cohosh on menstrual
randomized, double-blind, placebo-controlled design to irregularities and dysmenorrhea, one reported positive effects
study the efficacy of the preparation in the treatment of pro- (Görlich 1962), while the other showed no effect (Heizer
nounced psychological symptoms associated with 1960). Görlich (1962) reported a beneficial effect of black
menopause such as anxiety, lack of motivation, depressed cohosh in the treatment of morning sickness and threatened
mood, nervousness, and irritability (Boblitz and others miscarriage, though data are insufficient to accurately assess
1999). For six weeks, 87 women were administered efficacy.
Remifemin® Plus (1 tablet twice daily) and 92 patients were
given placebo. Treatment efficacy was evaluated using the Hormonal and Central Nervous System Effects
KM Index. The decrease in the KM Index following active Human Clinical Studies
treatment was significantly greater than that in the placebo Conflicting evidence exists with regard to the hormonal
group (P < 0.001). Six minor adverse events were reported in effects of black cohosh extract. In a 1991 placebo-controlled
five patients in each treatment group without differences in study, Düker and others tested the ability of black cohosh
frequency or severity. extracts to reduce gonadotropin secretion in 110 menopausal
The other trial was an observational cohort study of 911 women with climacteric symptoms. Half the patients were
pre-, peri-, and postmenopausal women (Liske and others treated with black cohosh extract (Remifemin®, two tablets
1997). Patients received either two (n = 619, 68%) or four (n daily, each containing 2 mg of dried extract, yielding 8 mg of
= 292, 32%) tablets daily for 12 weeks. Treatment efficacy native extract daily), while the other half received a placebo.
was evaluated using the CGI scale and patient self-assess- A blood sample was drawn after two months of treatment and
ment of seven psychovegetative symptoms. Patients were LH and FSH levels were tested with a radioimmunoassay. In
evaluated prior to treatment and at 4 and 12 weeks. Over the the black cohosh patients, mean LH levels were reported to
12-week period, significant improvement (P < 0.001) in hot be significantly less than in the placebo group (P < 0.05).
flashes, depression, and insomnia was experienced by 77%, Mean FSH levels remained similar in both groups.
75%, and 72%, respectively, in the 811 patients assessed on According to the researchers, these results indicate that black
all three dates. Change in CGI scale over the treatment peri- cohosh may have an estrogenic effect. Because baseline lev-
od indicated that drug efficacy was good or very good in 41% els of hormones were not measured, it is not known whether
and 40% of the patients, respectively. A stronger effect was the groups were comparable at the beginning of the study,
observed at the higher dosage. The medication was tolerated making the value of the results questionable.
very well by 99% of the patients. Adverse events that were Seven of the human clinical studies on the effect of
likely to be correlated with therapy included mild nausea black cohosh extract on menopausal symptoms (presented in
and moderate diarrhea; adverse events possibly correlated more detail above) also investigated the extract’s estrogenic
with the medication included dry skin, itching skin, and dry effects (Georgiev and Iordanova 1997; Jacobson and others
tongue. 2001; Lehmann-Willenbrock and Riedel 1988; Liske and
Summary others 2000; Nesselhut and Liske 1999; Stoll 1987;
In summary, clinical evidence provides support for the use Warnecke 1985; see Table 4). The extract had no effect on
of black cohosh extract in the treatment of menopausal hormone levels in the four studies investigating this parame-
symptoms (Table 3). One double-blind placebo-controlled ter (Jacobson and others 2001; Lehmann-Willenbrock and
study found a significant improvement of menopausal symp- Riedel 1988; Liske and others 2000; Nesselhut and Liske
1999). This is in contrast to the study of Düker and others

22 American Herbal Pharmacopoeia™ • Black Cohosh Rhizome • 2002


(1991) which found a decrease in LH levels. Among the five Vaginal cytology did not change following treatment with
studies looking at black cohosh treatment effects on vaginal the dichloromethane fraction. The black cohosh methano-
and/or uterine tissue, three found positive proliferative lic extract was further fractionated by the same research
effects on vaginal tissue (Stoll 1987; Georgiev and Iordanova group in an effort to identify the active compound(s) (Jarry
1997: Warnecke 1985) and two did not (Liske and others and others 1985). The water phase was inactive. Three
2000; Nesselhut and Liske 1999), while two found no pro- active compounds were isolated from the chloroform phase,
liferative effects on the endometrium (Georgiev and including a compound thought to be formononetin, a
Iordanova 1997; Nesselhut and Liske 1999). Unfortunately, known phytoestrogen. The presence of formononetin in
all of the studies to date have been for 6 months or less, black cohosh has since been disputed. Although this com-
whereas at least 12 months is necessary to reveal an estro- pound did bind to estrogen-receptors, it did not have an LH-
genic effect on vaginal or uterine tissue. suppressive effect compared to the dichloromethane frac-
tion or vehicle.
Animal and In Vitro Studies
Düker and others (1991) tested the ability of a lipophilic
Several animal and in vitro studies have investigated the
black cohosh extract (8 mg extract daily) and its uncharac-
potential estrogenic activity of black cohosh extract by mea-
terized fractions to lower LH levels in ovx rats. All test sub-
suring its effects on serum hormone level, various tissues and
stances were dissolved in a vehicle (miglyol). Mature ovx rats
cell lines, receptor binding, and gene regulation. Other stud-
received seven injections of either the extract, one of its eight
ies have tested black cohosh’s effects on neurotransmitters.
fractions, or the vehicle alone at 12-hour intervals over three
The findings of this research are mixed. A number of the
and one-half days. Three hours after the seventh injection,
studies reported reflect preliminary findings from work pub-
the first of 10 blood samples was drawn, followed by the
lished as abstracts, mostly in conference proceedings.
other 9 at 20-minute intervals. The lipophilic extract showed
Because removal of ovaries results in cessation of estro-
a strong suppressive effect on LH levels in the rats. Four of
gen production, ovariectomized (ovx) rats are used as an ani-
the fractions showed significant LH-suppressive effects (P <
mal model for menopause. As estrogen production ceases in
0.01, compared to vehicle). The researchers went on to test
women, the serum concentration of FSH and LH greatly
the ability of the fractions to compete in vitro with 17-β-
increases, while secretion of prolactin by the pituitary
estradiol and vehicle for estrogen receptor binding sites. Of
decreases. If estrogen or estrogen-like substances are intro-
the four fractions that lowered LH levels in vivo, one showed
duced exogenously, LH and FSH secretion is reduced while
no receptor binding activity, while three showed activity
prolactin secretion is stimulated. The administration of
comparable to that of estradiol. Another fraction showed pos-
estrogen or estrogen-agonistic substances to ovx rats is
itive receptor binding but did not lower LH levels in vivo
expected to cause similar estrogenic effects. Many of the ani-
under the dosage regime administered. Given as a single
mal studies use an increase in uterine weight as an indicator
injection, however, it did have an LH-lowering effect similar
of an estrogenic effect of black cohosh extract. Other
to that of estradiol in vivo. The researchers suggest that this
researchers have pointed out that uterine weight is a poor
fraction contains estrogenic compounds which may be
indicator of unopposed estrogenic endometrial effects such
metabolized so quickly that only a transient LH suppressive
as hyperplasia (Johnson and others 2001).
effect is exerted. Thus the endocrine effect could be missed
Effects on Hormone Levels in the chronic administration of the fraction, but not in the
In a vehicle-controlled experiment designed to test for estro- acute administration.
genic effects of black cohosh extract, Jarry and In a dimethyl-benzanthracene (DMBA) animal model
Harnischfeger (1985) studied the effect of the extract on LH (see Effects on Estrogen-sensitive Tumors and Cell Lines),
secretion in ovx rats. They used black cohosh liquid extract black cohosh extract (Remifemin®) po at 1x, 10x, and 100x
(Remifemin®) evaporated or lyophilized to dryness and the human therapeutic dose showed no estrogen agonistic
reconstituted in a solution of water and 1,2 propanediol effects on hormone levels (LH, FSH, prolactin)
(70/30 V/V) to a concentration of 40 mg dried extract per (Freudenstein and others 2000).
mL injection vehicle. The extract or the vehicle alone was Löhning and others (1999) measured the effect of black
administered twice daily at 0.3 mL/animal ip for 1, 3, or 14 cohosh extract (uncharacterized) on prolactin secretion in
days. Each treatment group consisted of eight rats. After pituitary cell cultures. They found that both basal and thy-
treatment, the rats were sacrificed and serum levels of the rotropin-releasing hormone (TRH)-stimulated prolactin
pituitary hormones LH, FSH, and prolactin were measured secretion was significantly reduced by dosages of 10 and 100
in trunk blood using radioimmunoassay. Only under the µg/mL. This effect was reversed by the dopamine (D2/D4)-
three-day treatment period did rats treated with the extract antagonist haloperidol, suggesting dopaminergic activity of
have significantly lower LH levels than vehicle-treated rats the extract.
(P < 0.01). Prolactin and FSH levels were not affected under
Effects on Uterine, Vaginal, and Bone Tissue
any treatment regime. The dichloromethane fraction of the
A positive estrogenic effect of black cohosh extract on vagi-
extract (= 27 mg dry residue ip per animal), but not the
nal or uterine tissue has been reported in four preclinical
ethanol or water fractions, had LH-suppressive effects. The
studies. Using mice as a model, Liu and others (2001b)
non-hydrolyzed (glycosilated) dichloromethane fraction was
reported an increase in uterine weight with increasing dose
more effective than the hydrolyzed (aglycone) fraction.

American Herbal Pharmacopoeia™ • Black Cohosh Rhizome • 2002 23


of black cohosh extract (uncharacterized) (75, 150, 300 sitive Tumors and Cell Lines), black cohosh extract
mg/kg ig for 14 days). Treatment with the extract also signif- (Remifemin®) po at 1x, 10x, and 100x the human therapeu-
icantly prolonged the number of days of estrus (P < 0.05 at tic dose showed no estrogenic influence on histologically
the 300 mg/kg dose). The authors interpreted this as evi- analyzed uterine tissue (Freudenstein and others 2000).
dence of an estrogenic effect. It should be noted that other Einer-Jensen and others (1996) similarly found no
researchers have pointed out that uterine weight is a poor estrogenic effect of a black cohosh dry extract (50% ethano-
indicator of unopposed estrogenic endometrial effects such lic extract) in immature female mice and ovx rats. The mice
as hyperplasia (Johnson and others 2001). (n = 215) were divided into groups of 10 and given 6, 60, or
In 1997, Eagon and others tested an extract of black 600 mg/kg daily of the extract orally for 3 days. A placebo
cohosh root (characterization and dose undisclosed) for control group received 1 mL/kg of suspension vehicle and a
estrogenicity in ovx rats. The extract was added to a standard positive control group received 0.4 mg/kg estradiolbenzoate
liquid rat diet and administered for three weeks. Changes in sc. Uterine weight was measured after sacrifice on day 4. In
uterine tissue growth and uterine c-myc mRNA levels the same study, 47 ovx rats were divided into groups of 12,
(increased by estrogens) were tested. Hepatic estrogenic each receiving either 6, 60, or 600 mg/kg daily sc of the
response was also measured by testing for changes in serum extract for three days. Vaginal smears were taken on day 4.
ceruloplasmin levels and liver ceruloplasmin mRNA. The There were no signs of uterotrophic or vaginotrophic effects
extract did have a positive effect on uterine tissue growth, in either the mice or the rats.
but did not effect a change in uterine c-myc mRNA levels.
Receptor Binding Effects
While it did raise the serum ceruloplasmin level, it did not
Following their in vivo study demonstrating the LH-sup-
affect the liver ceruloplasmin mRNA levels. These
pressive effects in ovx rats of black cohosh extract and its
researchers concluded that black cohosh root extract did
fractions, Jarry and others (1985) tested the estrogen recep-
show signs of measurable estrogenic activity.
tor-binding effects of the various fractions. They found that
In the Allan-Doisy test, doses of 0.3 mL twice daily of
the dichloromethane (lipophilic) extract and various frac-
black cohosh extract (Remifemin®) induced estrus in eight
tions of the chloroform phase competed with radio-labelled
out of nine ovx rats; lower doses did not have this effect
estrogen for binding sites on estrogen receptors isolated from
(Földes 1959). This was taken as evidence of an estrogenic
rat uteri. In a follow-up study, the same research group
effect. Twelve three-week-old female mice were given 0.2
(1999) found that the lipophilic fraction of black cohosh
mL of the extract for four days and were sacrificed on day 6.
extract (BNO 1055) had significant estrogen receptor bind-
Uterine weight was increased in the treatment group com-
ing activity, while the hydrophilic fraction did not. The in
pared to the controls. The results from this study were not
vitro work of Düker and others (1991), presented above,
evaluated statistically.
showed a positive estrogen receptor binding effect of some
Knüvener and others (2000) tested black cohosh
of the fractions of the lipophilic extract of black cohosh. In
ethanolic extract (50%; 50-600 mg daily po for three days)
contrast to the above three studies, Liu and others (2001a)
for estrogenic activity in ovx mice. Placebo controls received
found no estrogen receptor-binding activity of a black
water and positive controls received mestranol or 17-β-estra-
cohosh extract.
diol. Both positive controls induced estrus in the mice,
while black cohosh extract was ineffective. An estrogenic Effects on Estrogen-regulated Genes
effect on vaginal cytology was seen in the black cohosh Jarry and others (1999) attempted to differentiate between
group. When co-administered, black cohosh extract (250 the potential ERα- and ERβ-binding effects of black cohosh
mg/kg) significantly antagonized the estrogenic effects of extract. They investigated the effects of black cohosh extract
mestranol. (BNO 1055) on the bone, brain (both containing a pre-
In contrast to the findings of the above studies, three dominance of ERβ), and uterine tissue (containing mostly
other studies found no estrogenic effect of black cohosh ERα) from ovx rats. Over a three-week period, the rats were
extract on vaginal or uterine tissues. Wuttke and others treated with 60 mg/rat sc or ip of the extract (a
(2000) tested the effect of black cohosh extract (BNO 1055) dichloromethanol phase of the dried extract was used to
on uterine tissue, bone loss, and liver and lipid metabolism. enrich lipophilic compounds) or 8 µg/rat of 17-β-estradiol;
Ovariectomized rats were treated with estradiol or BNO both preparations were dissolved in 5% cremophor. A con-
1055 for six hours, seven days, or three months (dose and trol group received only 5% cremophor. Following sacrifice,
route unspecified). Whereas estradiol caused tissue prolifer- the brains, uteri, and femoral arteries were removed from
ation in the uterus, black cohosh extract did not. However, the animals and assays investigating ERα- and β-regulated
the extract reportedly mimicked the effects of estradiol in gene activation were conducted. Both black cohosh extract
preventing and opposing the development of osteoporosis. It and estradiol reduced serum LH levels (P < 0.08 and P <
also displayed beneficial effects on the liver and lipid metab- 0.01, respectively, compared to controls) and activated estro-
olism. The authors suggested that the results support the gen-regulated genes in both brain and bone tissue (P < 0.05,
hypothesis that black cohosh acts as a SERM, since the both treatment groups). However, only estradiol activated
uterus contains predominantly ERα sites, while bone has a estrogen-regulated genes in the uterus, causing an increase
high concentration of ERβ sites. in uterine weight (P < 0.01). The activity of the extract in
In a DMBA animal model (see Effects on Estrogen-sen- brain and bone tissue and not in uterine tissue suggests that

24 American Herbal Pharmacopoeia™ • Black Cohosh Rhizome • 2002


a compound in the extract, at the doses given, binds prefer- with the extract and the anti-estrogenic tumor-inhibiting
entially to ERβ. effect of tamoxifen was increased by co-incubation with the
Seidlová-Wuttke and others (2000) performed another extract. Zava and others (1998) found that incubation of T-
study designed to differentiate between potential ERα and 47D breast cancer cells with a 1/500 dilution of black cohosh
ERβ effects of black cohosh extract. After administration of ethanolic extract (50%, 2:10 g/mL) had no effect on cell pro-
estradiol (3.5 µg/rat for seven days or three months) or the liferation (statistical tests not reported). Also using the T-47D
black cohosh extract (BNO 1055, 62.5 mg/rat daily for seven cell line and the same black cohosh extract, Dixon-Shanies
days or three months, route unspecified), gene expression in and Shaikh (1999) reported a 22% and 71% decrease in cell
the uterus (ERα), bone, liver, and the aorta (ERβ) was growth by the extract at concentrations of 0.1 and 1.0, respec-
examined. While the black cohosh treatment mimicked the tively (P < 0.01). Oketch-Rabah and others (2002) found that
effects of estradiol in the bone, liver, and aorta, it had no neither the hexane, ethyl acetate, nor water extracts of black
effect in the uterus. The above results support the possibili- cohosh at concentrations of 0-20 µg/mL had any effect on
ty that black cohosh may act as a SERM, selectively target- the growth of estrogen-sensitive breast cancer cells. As pre-
ing ERβ. sented above, they also found that the extracts did not regu-
Oketch-Rabah and others (2002) tested black cohosh late estradiol-regulated genes found in ER-expressing breast
hexane, ethyl acetate, and water extracts (concentrations not cancer tissue. As noted above, another study found that black
specified) for estrogenicity using several assays. None of the cohosh extract did not stimulate progesterone receptor
extracts enhanced estrogen-inducible alkaline phosphatase mRNA or the estrogen-inducible gene presenelin-2 in S30
enzyme activity in Ishikawa cells. In addition, none of the breast cancer cells (Liu and others 2001a).
extracts modulated estrogen receptor function as evaluated In contrast to such reports suggesting that black cohosh
using the ERE luciferase reporter assay nor did they have a extract does not have a cell proliferative effect on estrogen-
regulatory effect on genes that are known to be regulated by receptor-positive breast cancer cells, two other studies (Liu
estradiol in estrogen receptor positive breast cancer cells and others 2001b; Löhning and others 1998) found a cell-
(ERα, PgR, pS2). In a study by Liu and others (2001a), black proliferative effect of the extract in MCF-7 cell lines. Liu
cohosh methanolic extract did not stimulate progesterone and others (2001b) found that, compared to a blank control,
receptor mRNA or the estrogen-inducible gene presenelin-2 both black cohosh extract and estradiol shortened the dou-
in S30 breast cancer cells or endometrial (Ishikawa) cells. bling time of MCF-7 breast cancer cell growth. They also
noted a significant increase in estrogen receptors in the
Effects on Estrogen-sensitive Tumors and Cell Lines
MCF-7 cells compared to the control (P < 0.01). Löhning
Freudenstein and others (2000) studied the effect of an iso-
and others (1998) found that the 50% or 96% ethanolic
propanolic extract of black cohosh (Remifemin®) on estro-
extract of black cohosh at dosages of 0.1-10 µg/mL and
gen receptor-positive breast tumors in young female rats.
0.001-1 µg/mL, respectively, stimulated MCF-7 cell growth,
Five to nine weeks after experimental tumor induction with
while a high dose (100 µg/mL) of the 50% extract did not.
DMBA, the ovaries of the rats were removed. Beginning at
The cell-proliferative effect of the extract was abolished in
day 10 after the operation, one group received the synthetic
the presence of the estrogen receptor antagonist IC 182,780.
estrogen mestranol (450 µg/kg daily), three groups received
Kruse and others (1999) isolated fukinolic acid, a caffe-
black cohosh extract po (doses comparable to 1x, 10x, and
ic acid ester, from black cohosh rhizome and tested its estro-
100x the human clinical dose), while the control group
genicity on MCF-7 cell proliferation. At doses of 5 x 10-7 and
received no treatment. After another seven weeks, the ani-
5 x 10-8 M, fukinolic acid increased cell proliferation up to
mals were sacrificed and tumor number and size were
119.7% ± 5.8 (P < 0.05) and 126.3% ± 5.1 (P < 0.001),
recorded, plasma hormone levels were analyzed, and the
respectively; the effect at these doses was equivalent to that of
uterine tissue was examined. After ovariectomy, the tumors
estradiol at 10-10 M. Higher doses (5 x 10-9 M) of fukinolic
regressed in all animals, presumably due to the lack of
acid were ineffective.
endogenous estrogen production. Mestranol caused a new
increase in tumor size that was not found in either the black Central Nervous System Effects
cohosh or control groups. At death, no difference in tumor In vivo and in vitro studies suggest that black cohosh extract
size or number existed between the black cohosh and con- may have a dopaminergic effect. An acute dose (25-100
trol groups. mg/kg po) given to mice was found to decrease body tem-
Nesselhut and others (1993) found no cell-proliferative perature in a dose-dependent manner and prolonged keta-
effect of black cohosh in the breast cancer cell line MD-435. mine-induced sleeping time (Löhning and others 1999).
The MCF-7 and T-47D human breast cancer cell lines are The highest dose had an effect comparable to that of the D2-
characterized by estrogen-dependent cell proliferation. agonist bromocriptine (5 mg/kg ip). These effects were like-
Using MCF-7 cells and dilutions of black cohosh extract ly mediated by dopamine D2 receptors, since they were
(Remifemin®), Freudenstein and Bodinet (1999) found that blocked by pretreatment with sulpiride. Löhning and
cell proliferation was “completely” and “significantly” inhib- Winterhoff (2000) went on, in a controlled study, to measure
ited by dilutions of 10-3 (approximately 100 µg/mL) and 10- neurotransmitter levels in the striatum, hippocampus, and
6, respectively (statistical tests not reported). The cell-prolif- hypothalamus of mice after they were pretreated with black
erative effect of estradiol was antagonized by co-incubation cohosh extract for 21 days. In the striatum, both serotonin

American Herbal Pharmacopoeia™ • Black Cohosh Rhizome • 2002 25


Table 4 Summary of results from clinical studies on the effect of black cohosh extract on hormone levels and vaginal and endometrial cell
status in menopausal1 women
Reference Study Product and daily Treatment Estradiol3 FSH3 LH3 Vaginal cell Endometrial
design, n2 dosage duration proliferation3 thickness3
Stoll 1987 DBRPCT Remifemin®, 4 tablets 12 weeks _ _ _ !* _
n = 26 each containing 2 mg
extract4

Jacobson and DBRPCT Remifemin®, 2 tablets, 60 days _ " " _ _


others 2001 n = 42 equivalent to 40 mg
dried herb4, 5

Liske and others DBR, Remifemin®, 6 months " " " " _
2000 GCP equivalent to 39 mg
n = 57-59 dried herb

Remifemin®, equivalent " " " " _


to 127 mg dried herb

Lehman- R, Open Remifemin®, 4 tablets, 6 months _ " " _ _


Willenbrock and n = 15 each containing 2 mg
Riedel 1988 extract4

Warnecke 1985 R, Open Remifemin®, 80 drops4 _ _ _ ! _


n = 20

Nesselhut and Open Remifemin® tablets, 3 months " " " " "
Liske 1999 n = 28 equivalent to 136 mg
dried herb

Georgiev and Open Unidentified black 3 months _ _ _ ! (in 40% of "


Iordanova 1997 n = 50 cohosh extract, dose treated
unspecified women)

Düker and others PCT Remifemin®, 2 tablets, 2 months _ "5 #* 5 _ _


1991 n = 55 each containing 2 mg
extract4, 6

Key: DBRPCT = double-blind, randomized, placebo-controlled trial; DBR = double-blind, randomized; R = randomized; PCT = placebo-controlled; GCP = Good Clinical Practice
compliant; - = no data; ‚ #= decrease; ! = increase; " = no change; * = P < 0.05.
1 Menopausal symptoms were natural, surgically induced by hysterectomy (Lehman-Willenbrock and Reidel 1988), or induced by breast cancer treatment (Jacobson and others
2001).
2 Sample size in the black cohosh group.

3 Unless otherwise noted, results in placebo-controlled trials express differences in change over time of the parameter in the treatment group compared to placebo; in uncon-
trolled trials, results express change over time of the parameter between baseline and end of treatment.
4 The dried herb equivalents were in the range of 48-140 mg daily (Boblitz and others 2000).

5 Some patients in each group took tamoxifen concurrently.

6 The change in hormone levels reported in this study was based on a comparison between the black cohosh and placebo groups after two months of treatment; baseline hor-
mone levels were not measured.

26 American Herbal Pharmacopoeia™ • Black Cohosh Rhizome • 2002


and dopamine metabolism were significantly reduced in the perhaps in part due to the selectivity of the tissues studied.
treatment group compared to the control group. The Such mixed results, combined with the difficulty in extrap-
authors suggest that such a non-estrogenic effect of black olating from preclinical data to activity in human tissues,
cohosh on neurotransmitters may be responsible for the make the interpretation of these findings problematic.
botanical’s effectiveness in reducing the incidence of hot Three of four studies found an LH- or prolactin-suppressive
flashes in women, a mechanism also suggested by Seidlová- effect of black cohosh extract in animals (Düker and others
Wuttke and Wuttke (2000). 1991; Jarry and Harnischfeger 1985; Löhning and others
Such effects on dopamine and serotonin suggest a 1999). Of the seven preclinical studies looking at the effect
monoamine oxidase inhibition (MAO) activity. Because of black cohosh extract on uterine or vaginal tissue, four
MAO inhibitors are used as antidepressants, Löhning and found positive estrogenic effects (Eagon and others 1997;
Winterhoff (2000) studied black cohosh for an antidepres- Földes 1959; Knüvener and others 2000; Liu and others
sant effect in female mice using the tail-suspension test. The 2001b) while three did not (Einer-Jensen and others 1996;
extract increased mobility time, suggesting an antidepres- Freundenstein and others 2000; Wuttke and others 2000).
sant effect that the authors suggest may explain the use of These data are difficult to evaluate given that it has been
black cohosh in treating the psychological symptoms of pointed out that uterine weight is a poor surrogate for
menopause. endometrial effects including hyperplasia (Johnson and oth-
ers 2001).
Summary
Three estrogen receptor-binding studies (Düker and
In summary, more work is needed to clarify the mechanisms
others 1991; Jarry and others 1985, 1999) have shown that
of action of black cohosh. Based on the accumulated data,
black cohosh extract competes for binding with estradiol,
it cannot be conclusively determined if black cohosh elicits
while one study had negative results (Liu and others 2001a).
estrogenic activity. A number of studies report an estrogenic
Definitive data regarding the effect of black cohosh extract
effect while an almost equal number do not. Clinical data
on estrogen-regulated genes and estrogen-sensitive breast
regarding the potential estrogenic effects of black cohosh are
cancer tumors and cell lines are lacking. While two studies
inconclusive. One of the five studies that measured the
found that black cohosh did not regulate estrogen-regulated
effect of black cohosh on serum hormone levels found a sig-
genes found in breast or endometrial tissues (Liu and others
nificant LH-suppressive effect (Düker and others 1991;
2001a; Oketch-Rabah and others 2002), two other studies
placebo-controlled) though that study suffered from an
found that the extract regulated estrogen-sensitive genes in
inadequate experimental design. Of the six clinical studies
some tissues but not in others (Jarry and others 1999;
measuring the effects of black cohosh treatment on vaginal
Seidlová-Wuttke and others 2000). These latter studies,
cell status, only one found a significant increase in cell pro-
along with a study by Wuttke and others (2000), found that
liferation (Stoll 1987; placebo-controlled), while two found
black cohosh extract may elicit an estrogenic activity with a
increases that were not analyzed statistically (Georgiev and
stronger affinity to tissues rich in ERβ sites, such as bone,
Iordanova 1997; Warnecke 1985; both open and uncon-
rather than tissues rich in ERα sites, such as the uterus,
trolled). Two studies followed endometrial thickness as an
lending support to the SERM hypothesis. Six of the eight
outcome measure and both found no effect of black cohosh
studies on breast cancer found no cell-proliferative effect of
on this parameter, although statistical analyses were not pre-
black cohosh (Dixon-Shanies and Shaik 1999; Freudenstein
sented (Georgiev and Iordanova 1997; Nesselhut and Liske
and Bodinet 1999; Freudenstein and others 2000; Nesselhut
1999). Jacobsen and others (2001) reported endometrial
and others 1993; Oketch-Rabah and others 2002; Zava and
hyperplasia as an adverse event in one patient in the black
others 1998). Another study found a dopaminergic effect of
cohosh-tamoxifen treatment group and noted other
black cohosh, suggesting that its use against hot flashes and
endometrial abnormalities in two other patients in the same
the psychological symptoms of menopause may be related
group (Jacobson, personal communication to AHP; unrefer-
to effects on neurotransmitters rather than an estrogenic LH-
enced). In making an overall evaluation of the clinical data
suppressive effect (Löhning and Winterhoff 2000).
available on the estrogenic effects of black cohosh, it should
be noted that many of the studies might not have been of Other Effects
sufficient duration to determine efficacy in some of the para-
meters reported. For example, the two studies reporting no Cardiovascular Effects
changes in endometrial thickness were of short duration The resinous chloroform fraction of black cohosh extract
(three months) and are of little relevance in evaluating estro- (1-10 mg/kg) had a dose-dependent hypotensive effect in
genic activity in this tissue, as a minimum of 12 months of unanesthetized rabbits and urethanized cats. No such effect
therapy is required before any such changes can be expect- was found in dogs or humans. In humans, black cohosh
ed. Summarizing the present clinical data, a conclusive extract was found to increase peripheral blood flow at doses
determination regarding an estrogenic effect of black of 0.5 mg/kg (Genazzani and Sorrentino 1962). In an ex
cohosh cannot be made. vivo study using rat aortic strips, fukinolic acid caused a
Preclinical studies have given mixed results with respect vasorelaxant effect in strips experimentally contracted with
to the estrogenic effects of black cohosh extract, perhaps in norepinephrine, while cimicifugic acids A, B, and E and
part due to differences in the product used in research and fukiic acid did not (Noguchi and others 1998).

American Herbal Pharmacopoeia™ • Black Cohosh Rhizome • 2002 27


Anti-inflammatory Effects Nervous System and Musculoskeletal Indications
In an in vitro study by Löser and others (2000), fukinolic Some of the earliest recorded indications of black cohosh
acid was shown to be a strong inhibitor of neutrophil elas- among the Eclectics included rheumatic pain, nervous ten-
tase which may explain the anti-inflammatory activity his- sion, and muscular tightness (Scudder 1903). It was consid-
torically attributed to various species of Actaea (syn. ered particularly effective for nervous people with heavy-
Cimicifuga), including black cohosh which was a primary feeling limbs and uterine cramps that radiated across the
remedy for rheumatism. pelvis and down the thighs (Harper-Shove 1952), syndromes
which the Eclectics referred to as “rheumatic neuralgia”.
Other Species of Actaea
Hysterical convulsions, chorea, epilepsy, and particularly
Asian species of Actaea (A. cimicifuga [syn. A. foetida], A.
postpartum seizure were among the traditional indications
dahurica, and A. heracleifolia) have been used as analgesics,
for black cohosh. The Physiomedicalist William Cook
anti-inflammatories, antipyretics, and anticonvulsants
(1869) recommended black cohosh for nervous excitation
(Yamahara and others 1985; Yen 1992), actions that are con-
and agitation suggesting that it be combined with lady’s slip-
sistent with the use of A. racemosa among herbalists.
per (Cypripedium spp.) and skullcap (Scutellaria lateriflora)
for neuralgic pain; with prickly ash bark (Xanthoxyllum sp.)
Conclusion
and poke berry (Phytolacca sp.) for rheumatism; and with
There is evidence from double-blind placebo-controlled blue cohosh (Caulophyllum thalictroides) for hysteria and
clinical trials for a beneficial effect of black cohosh in the general spasms. Finley Ellingwood (1919) reported black
treatment of menopausal complaints. Less well-designed tri- cohosh to be specific for muscle aches and pains and for hys-
als offer additional positive evidence; however, the possibil- teria presenting a flushed face, restlessness, and excitement.
ity of a placebo effect occurring in the black cohosh treat- King recommended minute doses (1/4-1/2 drop) of the satu-
ment groups in these studies cannot be ruled out. Evidence rated tincture in the ear or eye every two hours for pains of
in support of the use of black cohosh for premenstrual symp- neuralgic origin (Felter and Lloyd 1898). Modern practi-
toms and dysmenorrhea is lacking from the scientific litera- tioners continue to use black cohosh for nervous conditions
ture. Human, animal, and in vitro studies give mixed results associated with muscular pain, including fibromyalgia, and
regarding the estrogenic effects of black cohosh extract. for seizures and allaying spasmodic, irritable coughs.
Studies on the effects of black cohosh extract on vaginal or However, its most widespread use as a nervine among
endometrial tissue have been of too short a duration to rule herbalists and naturopathic physicians has been for pre-
out the possibility of estrogenic stimulation of these tissues. menstrual anxiety with muscle tension and headache and
Therefore, long-term, double-blind, placebo-controlled hormone-related anxiety and depression, as may occur pre-
studies need to be conducted in order to determine whether menstrually and perimenopausally.
black cohosh possesses any of the same risks as pharmaceu-
tical HRT. Clinical data regarding other medicinal uses of Cardiovascular Indications
black cohosh are lacking. Black cohosh was used as a cardiotonic by Eclectic physi-
cians, specifically for cases of fatty degeneration of the heart,
Medical Indications Supported by Clinical rheumatic carditis, and pericarditis and was also used for
Trials angina and cardiac arrhythmia due to excessive nervous
stimulation of the heart. William Cook (1869) recommend-
Black cohosh extract, equivalent in dosage and characteri-
ed the rhizome for relaxing nervous stimulation of the heart
zation to that shown to be effective in clinical trials, is indi-
and lungs in cases of hypertension, asthma, and pertussis
cated for use in the treatment of menopausal complaints
and reported that black cohosh can be used to slow the pulse
including hot flashes, outbreaks of sweating, and anxiety.
and improve capillary circulation. Black cohosh is less com-
monly used in these ways at the present time and substanti-
Medical Indications Supported by Traditional
ation for these indications are lacking.
and Modern Experience
The traditional Eclectic literature reports on the use of black Use in Infections
cohosh as a mild stomach tonic, gastric bitter, and alterative The Eclectics used black cohosh for serious infectious dis-
(White 1903) with a reported affinity for the nervous, mus- eases including pertussis, scarlet fever, smallpox, lung infec-
culoskeletal, and especially female reproductive systems. A tions, and especially postpartum infections. It was indicated
summary of traditional indications includes: nervous disor- for the acute onset of febrile illnesses, especially when asso-
ders such as chorea, epilepsy, myalgia, neuralgia, hysteria, ciated with chills and muscle aching (Ellingwood 1919).
depression, nervousness and anxiety, and sciatica; cardiovas- Tincture of black cohosh, often combined with cramp bark
cular disorders such as arrhythmia and hypertension due to (Viburnum opulus), has also been employed for flu and per-
vascular or nervous tension; various infectious diseases; and tussis in children by modern practitioners (Romm, personal
gynecological disorders such as amenorrhea, dysmenorrhea, communication to AHP; unreferenced). In his Medical
leukorrhea, atony of the uterus, cramps during pregnancy, Botany, Griffith (1847) reported black cohosh to promote
premature labor, postpartum hemorrhage or pains, and secretions of skin, kidney, and lungs.
pelvic pain. Black cohosh was also used traditionally as a par-
tus preparator and for pain of the prostate and testes.

28 American Herbal Pharmacopoeia™ • Black Cohosh Rhizome • 2002


Gynecological Indications labor was followed by severe uterine hemorrhage during
Uterine Indications birth.
The Eclectics used black cohosh extensively for gynecolog- Today, black cohosh is used by midwives for leg cramps,
ical problems including amenorrhea, dysmenorrhea, leuko- hypertension, and spasmodic cough during pregnancy, as a
rrhea, uterine cramps, prolapse of the uterus, and cervical partus preparator for women who have previously had diffi-
excoriation (Ellingwood 1919; Felter and Lloyd 1898; Jones cult labors, and in cases of prolonged or delayed labor. In
and Scudder 1858). King recommended a vaginal douche these latter cases its relaxing effect on the uterus is thought
of black cohosh and Geranium spp. infusion for cases of to allow for proper, coordinated uterine contractions. For
leukorrhea, uterine prolapse, and vaginal laxity (King 1855). these purposes, black cohosh is often used in combination
He recommended a typical dose of black cohosh to be 1 tsp with other herbs. Some midwives also use black cohosh in
of a mixture of 10 drops to 3.5 mL of “specific macrotys” (a threatened miscarriage with premature labor contractions
black cohosh product developed by Lloyd and of undis- with or without bleeding and consider it a safer agent than
closed characterization) per 4 oz of water or 1.5-7.5 mL of terbutaline (breathine). For this indication, it is often used
the fluid extract (1:1)*. To induce labor, King recommend- in combination with cramp bark (Viburnum opulus) and
ed dosing with 1/16 oz of the powdered rhizome in water or wild yam (Dioscorea spp.) (2.5-5 mL up to three times daily
1.5 mL of the tincture every 15-20 minutes until labor or 2.5 mL every 30 minutes with active presenting symp-
begins (Felter and Lloyd 1898). toms). This is continued for up to three days after symptoms
* It should be noted that 7.5 mL of a 1:1 fluid extract is equivalent to 7500 have subsided (Romm 2001).
mg of black cohosh dried herb, a dose substantially higher than the 40-80
mg typically used in clinical trials. In general, the dosages of black cohosh Menopausal Indications
cited in the Eclectic medical literature and those used by modern herbal Based primarily on modern German research (see
practitioners are substantially higher than dosages used in clinical trials. Pharmacodynamics and Clinical Efficacy), many naturo-
Black cohosh is thought to be a true amphoteric uterine pathic physicians and herbalists are now using black cohosh
tonic, relaxing uterine tone when excessive and spastic, yet to allay hot flashes, correct menopausal bleeding irregulari-
increasing tone when lax and atonic. As such, black cohosh ties, sleep disturbances, and nervous complaints such as anx-
has been used both to relax the uterus in order to halt pre- iety and depression (Hobbs 1998). It should be noted that
mature labor or prevent miscarriage (Felter 1922) and to long-term use for the treatment of menopausal symptoms is
promote the menses (emmenagogue) and facilitate labor a new indication compared to the short-term use of black
(partus preparator) (Culbreth 1917; Ellingwood 1919; Jones cohosh employed traditionally by herbal practitioners.
and Scudder 1858). Black cohosh is said to be a “sedative
emmenagogue” that promotes blood flow when uterine ten- Actions
sion, cramps and congestion are hindering flow, but will not Pharmacological: Mechanisms of action have not been
induce excessive flow when used to improve uterine tone. determined. Estrogenic agonism, antagonism, and no estro-
Black cohosh was also considered specific for ovarian pain genic effects have been reported in clinical and preclinical
(Felter 1891). studies.
Indications in Pregnancy Traditional: antispasmodic, anxiolytic, emmenagogue
The Eclectics recommended the use of black cohosh in late (when menstruation is inhibited due to uterine tension),
pregnancy as a uterine tonic and partus preparator nervine, partus preparator, uterine relaxant, uterine tonic.
(Ellingwood 1919; Felter 1891; Felter and Lloyd 1898;
Scudder 1903). King (1855) reported black cohosh to be a Substantiation for Structure and Function
partus accelerator in cases of stalled labor due to uterine Claims
atony. Black cohosh is believed to act on the uterus increas-
Based on a review of the currently available data, substanti-
ing muscle tone, but decreasing the pain of labor by pro-
ation for the development of a structure and function claim
moting more efficient, well-orchestrated contractions
is lacking.
(Ellingwood 1919). King (1855) considered black cohosh to
be of value in differentiating between false and true labor Dosages
pains with false labor pains dissipating and true labor pains
increasing upon its use. He also considered it the best and Dried rhizome and root:
safest agent for relieving after-pains, as did other Eclectics 1 g up to three times daily (Osol and Farrar 1947).
(Bartholow 1882; Felter 1891). Cook (1869) reports black Tincture (1:10):
cohosh to be indicated for rigidity of the cervical os during 0.4 mL daily (40%-60% alcohol V/V) (Blumenthal and
labor. He also recommended black cohosh combined with others 2000).
wake robin (Trillium sp.) and lady’s slipper for after-pains. Fluid Extract (1:1):
There was one report among the Eclectics of the “free 20 drops twice daily (60% ethanol V/V; equivalent to 40
use” of black cohosh during pregnancy being correlated mg dried herb daily).*
with “premonitions of abortion”; the author considered * Dosage based on research using the commercial product Remifemin®
(Schaper and Brümmer) or Klimadynon® (Bionorica AG, contains the
such cases “exceptions” (Cook 1869). Ellingwood (1919) extract BNO 1055).
noted six cases in which the use of black cohosh during

American Herbal Pharmacopoeia™ • Black Cohosh Rhizome • 2002 29


S A F E TY P R O F I L E cation to AHP; unreferenced).
In the study of Jacobson and others (2001), breast can-
Side Effects cer recurrence was reported in one woman taking black
Taken in doses equivalent to those used in clinical trials, cohosh with tamoxifen. The patient had an unreported
black cohosh appears to be very well tolerated. A detailed increase in carcinoembryonic antigen before entering the
reporting of side effects, when available, accompanies each study.
study presented under Therapeutics. Among the minor Modern herbalists have observed that some clients get
adverse events reported, the most frequent was mild gas- severe frontal headaches taking black cohosh preparations
trointestinal upset (average of 5.6% of subjects across five at various dosages (Anderson-Geller and Winston, personal
studies) (Földes 1959; Jacobson and others 2001; Kesselkaul communications to AHP; unreferenced), some occasionally
1957; Liske and others 2000; Vorberg 1984). Among the at tincture doses greater than 20 mL in a 24-hour period or
other minor adverse events reported were headache, vertigo, even a single dose greater than 3-5 mL (McQuade
weight gain, mastalgia, heavy feeling in the legs, and a stim- Crawford, personal communication to AHP; unreferenced).
ulant effect (Jacobson and others 2001; Stoll 1987). In the The dosages used by modern herbalists tend to be much
study by Jacobson and others (2001), minor adverse events higher than those used in clinical trials. A single anecdotal
occurred in 8 of 42 subjects in the black cohosh treatment report was received from a woman who in one day con-
group (n = 6 with, and n = 2 without tamoxifen) and in 2 of sumed 400 mg of a black cohosh extract characterized to
43 subjects in the placebo-tamoxifen group. In the Stoll 2.5% 27-deoxyactein. She experienced a sense of well being
(1987) study, minor adverse events occurred in 12 subjects and broke out in a rash (Upton, personal communication to
in the black cohosh treatment group and 2 each in the AHP; unreferenced).
placebo and conjugated estrogen groups. In the unpub-
lished study by Wuttke and others, the incidence of non-seri- Contraindications
ous adverse events was similar between the black cohosh, Based on the available data, no conclusive determination
estrogen, and placebo groups (Gorkow, Marz, and can be made at this time (see Precautions).
Christoffel, personal communication to AHP; unrefer-
enced). Three studies reported no adverse events in the Precautions
black cohosh treatment groups (Mielnik 1997; Nesselhut Both estrogenic and anti-estrogenic activities have been
and Liske 1999; Pethö 1987) and six made no mention of reported for black cohosh and no long-term clinical studies
adverse events (Daiber 1983; Georgiev and Iordanova 1997; exist. Historically, black cohosh was used for relatively short
Görlich 1962; Heizer 1960; Lehmann-Willenbrock and periods of time. The long-term use of it for the management
Reidel 1988; Warnecke 1985). of menopausal symptoms is a new indication. Therefore,
Other reported side effects that were variously classified women using black cohosh for long-term management of
as adverse events or outcome measures and which may have menopausal symptoms or those with a history of breast can-
been due to an estrogen stimulatory effect include vaginal cer (see Carcinogenicity) should do so only under the super-
bleeding and endometrial hyperplasia (Jacobson and others vision of a qualified health care professional. Black cohosh
2001; Liske and others 2000). Endometrial hyperplasia is a should not be used during pregnancy except under the
risk factor for endometrial cancer. In the Jacobson and oth- supervision of a qualified health care professional (see
ers (2001) study, vaginal bleeding and endometrial hyper- Pregnancy, Mutagenicity, and Reproductive Toxicity).
plasia occurred once each in the black cohosh-tamoxifen There appears to be no indication for long-term use of black
group. In this same study, two unspecified endometrial con- cohosh in children. According to the forthcoming World
ditions in the black cohosh-tamoxifen group resulted in a Health Organization (WHO) monograph on black cohosh,
dilatation and curettage and a hysterectomy (Jacobson, per- there is no therapeutic rationale for the use of black cohosh
sonal communication to AHP; unreferenced). As stated pre- in children. According to traditional and contemporary
viously, the effects of black cohosh are confounded with herbalists, however, black cohosh may be safely used for the
those of tamoxifen in this study, making the findings unreli- short-term treatment of acute respiratory infections in chil-
able; tamoxifen is known to cause endometrial abnormali- dren (see Medical Indications Supported by Traditional and
ties. The authors concluded that it was not likely that any of Modern Experience). The German Commission E
the adverse events found in the study were related to treat- (Blumenthal and others 1998) recommends that treatment
ment. In the study by Liske and others (2000), a biopsy with black cohosh not exceed six months. This is based on
showed an inactive endometrium in a patient presenting the current standard of practice in Germany that recom-
metrorrhagia. In the unpublished study of Wuttke and oth- mends the reassessment every six months of the need for
ers, vaginal bleeding occurred at least twice as frequently in pharmacological treatment of menopausal symptoms.
the black cohosh treatment group compared to the placebo.
The authors noted that the more frequent occurrence of Interactions
vaginal bleeding and the non-significant increase in vaginal No clinical interactions have been reported. In one in vitro
cell proliferation and endometrial thickness may be inter- study, black cohosh extract potentiated the anti-estrogenic
preted as a moderate estrogenic effect of black cohosh effect of tamoxifen in breast cancer cell lines and antago-
extract (Gorkow, Marz, and Christoffel, personal communi-

30 American Herbal Pharmacopoeia™ • Black Cohosh Rhizome • 2002


nized the cell-proliferative effects of estradiol (Freudenstein Lactation
and Bodinet 1999). However, a clinical trial investigating Data regarding the effects of black cohosh use during lacta-
the effect of black cohosh in breast cancer survivors found tion on infants or milk supply are lacking. Based on a review
no apparent interaction between tamoxifen and black of the available literature and the experience of modern
cohosh as indicated by similar serum hormone levels in practitioners, no adverse effects are to be expected. There
tamoxifen users taking black cohosh and those not taking appears to be no indication for long-term use of black
black cohosh (Jacobson and others 2001). In rats, black cohosh in lactating mothers.
cohosh extract enhanced the effects of mestranol on induc-
tion of ovulation (Löhning and others 2000). The relevance Carcinogenicity
of such in vitro data to humans is not known.
Endometrial hyperplasia, a precancerous condition, was
Pregnancy, Mutagenicity, and Reproductive reported in one out of 29 subjects taking black cohosh and
tamoxifen concurrently (Jacobson and others 2001). This
Toxicity condition is a known side effect of tamoxifen, so its occur-
Black cohosh should not be used during pregnancy except rence cannot necessarily be attributed to black cohosh. One
under the supervision of a qualified health care profession- animal study found no effect of black cohosh on estrogen-
al. Black cohosh was traditionally used by the Eclectics as a receptor-positive tumors in rats (Freudenstein and others
partus preparator and for uterine cramping and is still used 2000). Seven studies investigated the effect of black cohosh
in this way by midwives today. One early case series on breast cancer cell lines. Six of these found no cell-prolif-
(Görlich 1962) and contemporary use by midwives indicate erative effect (Dixon-Shanies and Shaik 1999; Freudenstein
that black cohosh may be used early in pregnancy as an anti- and Bodinet 1999; Liu and others 2001a; Nesselhut and oth-
abortifacient (Romm, personal communication to AHP; ers 1993; Oketch-Rabah and others 2002; Zava and others
unreferenced). In contrast to such uses, two anecdotal 1998), whereas two did (Liu and others 2001b; Löhning and
reports among the Eclectics cited the rare occurrence of others 1998). The relevance of such in vitro data to effects
threatened abortion or hemorrhage following the use of in human tissues is unknown and requires further testing.
black cohosh in pregnancy or labor, respectively (Cook
1869; Ellingwood 1919; see Medical Indications Supported Influence on Driving
by Traditional and Modern Experience). According to the Based on current knowledge, no negative effects are to be
forthcoming WHO monograph on black cohosh, the use of expected when black cohosh is taken at the suggested
black cohosh during pregnancy or lactation is contraindi- dosages.
cated. This is a general policy of WHO when safety data are
lacking regarding herb use during pregnancy and lactation Overdose
(Mahady and others 2001).
King’s American Dispensatory notes that impaired vision and
There have been two reports of birth defects or injury
dilation of the pupil have been known to occur with large
associated with maternal use of black cohosh. It is not possi-
doses (undefined) of black cohosh (Felter and Lloyd 1898).
ble to identify black cohosh as the causative agent in either
The 22nd edition of The Dispensatory of the United States of
case. In the first case, according to a letter to the New
America states that “In overdoses it [black cohosh] is said to
Zealand Medical Journal, a pregnant woman gave birth to a
cause general relaxation, vertigo, tremors, decided reduc-
baby with basal ganglia and parasagittal hypoxic injury after
tion of the pulse; occasionally it causes vomiting, but its
using a combination of black and blue cohosh
emetic action is never violent…In large doses it produces
(Caulophyllum thalictroides) to induce labor (there may
giddiness, with intense headache and prostration” (Wood
have been other botanicals in the combination) (Gunn and
and others 1937). The German reference Hager’s
Wright 1996). The woman reportedly had a normal preg-
Handbuch reports large doses of black cohosh to be 5 g of
nancy and birth. According to the primary reporters, a con-
the dried drug or 12 g of the fluid extract (List and
stituent of blue cohosh, caulosaponin, was likely to have
Hörhammer 1973).
been responsible for this effect due to its reported ability to
cause coronary blood vessel constriction and direct myocar-
Treatment of Overdose
dial toxicity. In a study of prenatal influences on fetal devel-
opment and survival, researchers followed 3200 pregnan- No data available.
cies; three of the mothers used black cohosh (dose and dura-
tion unspecified). In one of these cases, the child was born Toxicology
with an unspecified malformation (Mellin 1964). Female Wistar rats received 250, 1800, and 5000 mg/kg
The Ames test showed no evidence of a mutagenic Remifemin® granulate daily by oral gavage over a 26-week
potential of an isopropanolic extract of black cohosh equiv- period. The highest dose corresponds to 500 times the
alent to 30.3 mg. In comparison to the negative control human therapeutic dose. After an eight-week post-treatment
used, the extract showed no genotoxic effects (Beuscher observational phase, no toxic effects were observed in the
1996; Boblitz and others 2000). treatment group compared to control (Korn 1991). In the
study by Schindler (1992), the minimum lethal dose of
black cohosh (preparation unknown) following single doses

American Herbal Pharmacopoeia™ • Black Cohosh Rhizome • 2002 31


was reported to be > 500 mg/kg ip in mice, > 1000 mg/kg po ments. Action: Estrogen-like action; luteinizing hormone
in rats, and > 70 mg/kg in guinea pigs. Following subchron- suppression; binding to estrogen receptors (Blumenthal and
ic administration (30 days) of the preparation, the minimum others 1998).
lethal dose was > 6 mg/kg po in guinea pigs and > 10 mg/kg
Sweden
ip in mice.
Classified as a natural remedy intended for self-medication,
Genazzani and Sorrentino (1962) reported on the min-
requiring advanced application for marketing authorization
imum lethal dose of actein as follows: > 70 mg/kg iv single
(DeSmet and others 1993; Zhang 1998).
dose in rabbits; > 500 mg/kg ip for 10 days in mice; > 1000
mg/kg gastric single dose in rats. The conclusion of these Switzerland
researchers was that actein was not acutely toxic and that no Black cohosh single-ingredient medicines are classified by
effects on heart or spontaneous movement in animals was the Interkantonale Kontrollstelle für Heilmittel (IKS-List D)
observed. as non-prescription drugs with sale limited to pharmacies
and drugstores (Morant and Ruppanner 2001-2).
Classification of the American Herbal Products Indications: Menopausal complaints, hot flashes, sweating,
Association sleep disorders, nervousness, and mood disorders (climac-
The Botanical Safety Handbook of the American Herbal teric complaints).
Products Association (AHPA) assigns black cohosh the fol- United Kingdom
lowing classifications (McGuffin and others 1997): Occurs in the British Herbal Pharmacopoeia (BHP 1996).
Class 2b: Herbs not to be used during pregnancy unless oth- Black cohosh dried rhizome and root is an herbal medicine
erwise directed by an expert qualified in the use of the indicated for conditions capable of self diagnosis as specified
described substance.* in the General Sale List, Schedule 1 (medicinal products
Class 2c: Herbs not to be used while nursing unless other- requiring a full product license), Table A (for internal or
wise directed by an expert qualified in the use of the external use); 200 mg maximum single dose (Bradley 1992;
described substance.** GSL 1984-94). Exemptions from licensing are contained in
section 12 of the 1968 Medicines Act (Zhang 1998).
Notice: Emmenagogue, uterine stimulant. Indications: Menopausal disorders, premenstrual com-
Editor’s note: Occasional gastrointestinal discomfort may plaints, dysmenorrhea, uterine spasm (Bradley 1992).
occur. Large doses may cause vertigo, headache, nausea, Actions: Anti-inflammatory (BHP 1996), antirheumatic,
impaired vision, vomiting, and impaired circulation. An endocrine (pituitary, estrogen-mimetic) activity, emmena-
estrogenic effect and lowering of blood pressure have been gogue (Bradley 1992).
recorded (McGuffin and others 1997).
* See Pregnancy, Mutagenicity, and Reproductive Toxicity. WHO
** See Lactation. Black cohosh rhizome is the subject of a forthcoming mono-
graph by the World Health Organization (Mahady and oth-
ers 2001).
I N T E R N AT I O N A L S TAT U S
United States
Regulated as a dietary supplement. Black cohosh dried rhi-
zome and root, both whole and powdered, and black cohosh
dry extract have been proposed for inclusion in the United
States Pharmacopeia – National Formulary (USPF 27 [4]
2001).
Canada
Black cohosh will be listed as a Natural Health Product
under new regulations if a health claim is made or if it is
provided in specific dosage forms. Crude herbs in bulk form
will remain regulated as “foods”. When identified as a
Traditional Herbal Medicine, black cohosh preparations are
regulated as non-prescription over-the-counter drugs requir-
ing pre-market registration and assignment of a drug identi-
fication number (HC 1995, 2001; Zhang 1998).
Germany
The fresh or dried rhizome with attached roots is an
approved non-prescription drug for oral use according to the
German Commission E Monographs (Blumenthal and oth-
ers 1998). Indications: Premenstrual disorders, dysmenor-
rhea, or climacteric [menopausal] neurovegetative ail-

32 American Herbal Pharmacopoeia™ • Black Cohosh Rhizome • 2002


American Herbal Pharmacopoeia™ • Black Cohosh Rhizome • 2002 33
REFERENCES
Barnes S. 1998. Phytoestrogens and Cimicifuga racemosa (L.) Nutt. Materia Medica Therapeutics L. Nature 194(May):544-5.
breast cancer. Baillière’s Clin Isolation, structure elucidation and Pharmacognosy. 11th ed. Georgiev DB, Iordanova E. 1997.
Endocrin Metab 12(4):559-78. and absolute configuration of Chicago: Ellingwood’s Phytoestrogens—the alternative
Bartholow R. 1882. A Practical 26-deoxyactein and clarification Therapeutist. 564 p. approach [abstract]. Maturitas
Treatise on Materia Medica and of nomenclature associated with Einer-Jensen N, Zhao J, Andersen 27(Suppl.):213. Abstract nr
Therapeutics. New York: D. 27-deoxyactein. J Nat Prod. KP, Kristoffersen K. 1996. P309.
Appleton & Co. 662 p. Forthcoming. Cimicifuga and Melbrosia lack Görlich N. 1962. Treatment of ovari-
Barton BS. 1810. Collections for an Compton JA, Culham A, Jury SL. oestrogenic effects in mice and an disorders in general practice.
Essay Towards a Materia Medica 1998. Reclassification of Actaea rats. Maturitas 25:149-53. Arztl Prax 34:1742-3.
of the United States. 3rd ed. to include Cimicifuga and Erichsen-Brown C. 1979. Medicinal Grady D, Gebretsadik T, Kerlikowske
Volume 1. Philadelphia: Edward Souliea (Ranunculaceae): phy- and Other Uses of North K, Ernster V, Petitti D. 1995.
Earle. 67 p. Reprinted:1900. logeny inferred from morpholo- American Plants. New York: Hormone replacement therapy
Bedir E, Khan IA. 2000. gy, nrDNA ITS, cpDNA, trn L-F Dover. 512 p. and endometrial cancer risk: a
Cimiracemoside A: a new sequence variation. Taxon Fabricant D, Li W, Chen SN, meta-analysis. Obstet Gynecol
cyclolanostanol xyloside from 47:593-34. Graham J, Fizloff JF, Fong 85(2):304-13.
the rhizome of Cimicifuga race- Cook WMH. 1869. The Physio- HHS, Farnsworth NR. 2001. Griffith RE. 1847. Medical Botany:
mosa. Chem Pharm Bull Medical Dispensatory: A Geographical and diurnal varia- or Descriptions of the More
48(3):425-7. Treatise on Therapeutics, tion of chemical constituents of Important Plants Used in
Beral V, Bull D, Doll R, Key T, Peto Materia Medica, and Pharmacy, Cimicifuga racemosa (L.) Nutt. Medicine, with Their History,
R, Reeves G. 1997. Breast can- in Accordance with the [abstract]. In: Botanical dietary Properties, and Mode of
cer and hormone replacement Principles of Physiological supplements: natural products at Administration. Philadelphia:
therapy: collaborative reanalysis Medication. Cincinnati: WMH a crossroads; 2001 Nov 8-11; Lea & Blanchard. 704 p.
of data from 51 epidemiological Cook. 832 p. Reprinted:1985. Asilomar CA. Washington, DC: Gunn T, Wright IMR. 1996. The use
studies of 52,705 women with Corsano S, Piancatelli G, Panizzi L. American Society of of black and blue cohosh in
breast cancer and 108,411 1969. Sulla Acteina, principio Pharmacognosy & Council for labour. New Zealand Med J
women without breast cancer. attivo dell’ Actea racemosa. Gazz Responsible Nutrition. p 34. 109:410-1.
Lancet 350(9089):1047-59. Chim Ital 99:915-32. Abstract nr P:12. Hagels H, Baumert-Krauss J,
Beuscher N. 1996. European phy- Corsano S, Mellor JM, Ourisson G. Felter HW. 1891. Eclectic medi- Freudenstein J. 2000.
totherapy: Cimicifuga racemosa 1965. The structure of cines.—I. Macrotys. Medical Composition of phenolic con-
L.—black cohosh. Quart Rev cimigenol. Chem Comm 10:185- Gleaner 3(5):109-11. stituents in Cimicifuga racemosa
Nat Med (spring):19-27. 6. Felter HW, Lloyd JU. 1898. King’s [abstract]. International
Blumenthal M, Busse WR, Hall T, Corsano S. 1965. Research on the American Dispensatory. 18th ed. Congress and 48th annual meet-
Goldberg A, Grünwald J, structure of cimigenol. Gazz Volume 1-2. Portland: Eclec ing of the Society for Medicinal
Riggins CW, Rister RS, editors. Chim Ital 95(1-2):117-26. Med. 2172 p. Reprinted:1983. Plant Research (GA) and 6th
1998. The Complete German Corsano S, Piancatelli G. 1967. Felter HW, Lloyd JU. 1909. King’s International Congress on
Commission E Monographs: [Title unavailable]. Ric Sci American Dispensatory. 19th ed. Ethnopharmacology of the
Therapeutic Guide to Herbal 37(4):366-9. Volume 1-2. Cincinnati: Ohio International Society for
Medicines. Translator: Klein S. Crellin JK, Philpott J. 1990. Herbal Valley. 2172 p. Ethnopharmacology (ISE);
Boston: Integ Med Comm. 685 Medicine Past and Present. Felter HW. 1922. The Eclectic Zurich. Abstract nr P1B/03.
p. Translation of: German Volume 2. Durham (NC): Duke Materia Medica, Pharmacology Harnischfeger G, Stolze H. 1980.
Kommission E. Univ Pr. 549 p. and Therapeutics. Volume 1. Well-proved active substances
Blumenthal M, Goldberg A, Culbreth DMR. 1917. A Manual of Portland: Eclec Med. 743 p. derived from natural products:
Brinckmann J. 2000. Herbal Materia Medica and Reprinted:1985. black snakeroot. Notabene
Medicine: Expanded Pharmacology. 6th ed. Portland: Földes J. 1959. Die Wirkungen eines Medici 10:446-50.
Commission E Monographs. Eclec Med. 627 p. Extraktes aus Cimicifuga race- Harper-Shove F. 1952. Prescriber &
Newton (MA): Integ Med Reprinted:1983. mosa. Arzneimittelforschung Clinical Repertory of Medicinal
Comm. 519 p. Daiber W. 1983. Klimakterische 13(12):623-4. Herbs. 2nd ed. Essex: CW
Boblitz N, Schrader E, Henneicke- Beschwerden: ohne Hormone Ford BA. 1997. Actaea. In: Flora of Daniel. 228 p.
von Zepelin HH, Wüstenberg P. zum Erfolg! Arzt Prax 35:1946-7. North America Editorial Heizer H. 1960. Kritisches zur
1999. Benefit of a fixed drug De Smet PAGM, Keller K, Hänsel R, Committee, editors. Flora of Cimicifuga—Therapie bei hor-
combination containing St. Chandler RF, editors. 1993. North America. Volume 3. New monalen Störungen der Frau.
John’s wort and black cohosh for Adverse Effects of Herbal Drugs. York: Oxford Univ Pr. p 181-3. Med Klin 55(6):232-33.
climacteric patients—results of a Volume 2. Berlin: Springer. 348 Franke R, Schenk R, Abel G. 2000. Henrich JB. 1992. The post-
randomised clinical trial. In: p. Cimicifuga racemosa (L.) menopausal estrogen/breast can-
Proceedings of the 6th annual Dixon-Shanies D, Shaikh N. 1999. Nuttal—ein Kandidt für cer controversy. JAMA 268:1900-
symposium on complementary Growth inhibition of human Inkulturnahme. Z Arzn Gew 2.
health care; 1999 Dec 2-4; breast cancer cells by herbs and 5(2):62-7. Hobbs C. 1998. Black cohosh: a
Exeter (UK). Exeter: Univ phytoestrogens. Oncol Rep Freudenstein J, Bodinet C. 1999. woman’s herb comes of age.
Exeter. p 3. 6(6):1383-7. Influence of an isopropanolic Herbs For Health
Boblitz N, Liske E, Wüstenberg P. Düker EM, Kopanski L, Jarry H, aqueous extract of Cimicifugae March/April:38-41.
2000. Black cohosh: efficacy Wuttke W. 1991. Effects of racemosae rhizoma on the prolif- Jacobson JS, Troxel AB, Evans J,
effect and safety of Cimicifuga extracts from Cimicifuga race- eration of MCF-7 cells. In: 23rd Klaus L, Vahdat L, Kinne D, Lo
racemosa in gynecology. DAZ mosa on gonadotropin release in international LOF-symposium KMS, Moore A, Rosenman PJ,
140(24):107-14. menopausal women and on “phyto-oestrogens”; 1999 Jan Kaufman EL and others. 2001.
Boyle W. 1991. Official Herbs: ovariectomized rats. Planta Med 15; Belgium. Belgium: Univ Randomized trial of black
Botanical Substances in the 57:420-4. Gent. p 1. cohosh for the treatment of hot
United States Pharmacopeias, DuPont WD, Page DL. 1991. Freudenstein J, Dasenbrock C, flashes among women with a
1820-1990. New Palestine (OH): Menopausal estrogen replace- Niblein T. 2000. Lack of promo- history of breast cancer. J Clin
Buckeye Naturopathic. 77 p. ment therapy and breast cancer. tion of estrogen dependent Oncol 19(10):2739-45.
Bradley PR. 1992. British Herbal Arch Int Med 151:67-72. mammary gland tumors in vivo Jarry H, Harnischfeger G. 1985.
Compendium. Volume 1. Eagon CL, Elm MS, Teepe AG, by an isopropanolic black Studies on the endocrine effica-
Bournemouth: Brit Herb Med Eagon PK. 1997. Medicinal cohosh extract [abstract]. cy of the constituents of
Assoc. 238 p. botanicals: estrogenicity in rat Phytomedicine 7(Suppl 2):13. Cimicifuga racemosa: 1.
Chen SN, Li WK, Fabricant DS, uterus and liver [abstract]. Abstract nr SL-14. Influence on the serum concen-
Santarsiero BD, Mesacar A, Proceed Am Assoc Cancer Res Genazzani E, Sorrentino L. 1962. tration of pituitary hormones in
Fitzloff JF, Fong HHS, 38:293. Abstract nr 1967. Vascular action of acteina: active ovariectomized rats. Planta Med
Farnsworth NR. 2002. Ellingwood F. 1919. American constituent of Actaea racemosa 1(4):46-9.

34 American Herbal Pharmacopoeia™ • Black Cohosh Rhizome • 2002


Jarry H, Harnischfeger G, Düker E. Kuiper GGJM, Lemmen JG, Booth N, Constantinou AI, Goldberg A. 1997. Botanical
1985. Studies on the endocrine Carlsson B, Corton JC, Safe SH, Pezzuto JM, Fong HHS and oth- Safety Handbook. Boca Raton:
efficacy of the constituents of Saag van der PT, Burg van der ers. 2001a. Evaluation of estro- CRC. 231 p.
Cimicifuga racemosa: 2. in vitro B, Gustafsson JA. 1998. genic activity of plant extracts for McGuffin M, Kartesz JT, Leung AY,
binding of constituents to estro- Interaction of estrogenic chemi- the potential treatment of Tucker AO. 2000. Herbs of
gen receptors. Planta Med cals and phytoestrogens with menopausal symptoms. J Agric Commerce. 2nd ed. Silver
4(1):291-356. estrogen receptor β. Food Chem 49(5):2472-9. Spring (MD): American Herbal
Jarry H, Leonhardt S, Duels C, Popp Endocrinology 139(10):4252-63. Liu Z, Yang Z, Zhu M, Huo J. Products Association. 421 p.
M, Christoffel V, Spengler B, Kusano G, Hojo S, Kondo Y, 2001b. Estrogenicity of black Mellin GW. 1964. Drugs in the first
Theiling K, Wuttke W. 1999. Takemoto T. 1977. Studies on cohosh (Cimicifuga racemosa) trimester of pregnancy and the
Organ-specific effects of the constituents of Cimicifuga and its effect on estrogen recep- fetal life of Homo sapiens. Am J
Cimicifuga racemosa (CR) in spp. XIII. Structure of cimicifu- tor level in human breast cancer Obst Gyne 90(7 pt 2):1169-80.
brain and uterus. In: 23rd inter- goside. Chem Pharm Bull MCF-7 cells. Wei Sheng Yan Jiu Mielnik J. 1997. Extract of
national LOF-symposium on 25(12):3182-9. 30(2):77-80. Cimicifuga racemosa in the treat-
“phyto-oestrogens”; 1999 Jan 15; Lata H, Bedir E, Hosick A, Ganzera Lloyd JU. 1912. Vegetable drugs ment of neurovegetative symp-
Belgium. Belgium: Univ Gent. M, Khan I, Moraes RM. 2001. employed by American physi- toms in women in the peri-
p 1. In vitro plant regeneration from cians. J Am Pharm Assoc menopausal period. Maturitas
Johnson EB, Muto MG, leaf-derived callus in black 1(11):1228-41. 27 (Suppl.):215.
Yanushpolsky EH, Mutter GL. cohosh (Cimicifuga racemosa) Lloyd JU. 1921. Origin and History of Moerman DE. 1986. Medicinal
2001. Phytoestrogen supplemen- [abstract]. In: Botanical dietary All the Pharmacopoeia Plants of Native America.
tation and endometrial cancer. supplements: natural products at Vegetable Drugs, Chemicals, Volume 1. Ann Arbor: Regents
Obstet Gyn 98(5):947-50. a crossroads; 2001 Nov 8-11; and Preparations. Cincinnati: Univ Mich Museum Anthropol.
Jones LE, Scudder JM. 1858. The Asilomar, CA. Washington, DC: Caxton. 449 p. 910 p.
American Eclectic Materia American Society of Lloyd JU, Lloyd CG. 1931a. Moore M. 1993. Medicinal Plants of
Medica and Therapeutics. Pharmacognosy & Council for Caulophyllum. Bulletin of the the Pacific West. Santa Fe: Red
Volume 1-2. Cincinnati: Moore, Responsible Nutrition. p 33. Lloyd Library of Botany, Crane Books. 360 p.
Wilstach, Keys. 1010 p. Abstract nr P:9. Pharmacy and Materia Medica Morant J, Ruppanner H, editors.
Kennelly EJ, Baggett S, Nuntanakorn Lehmann-Willenbrock E, Riedel 9(31 vol 2):141-62. 2001-02. Arzneimittel-
P, Ososki AL, Mori S, Coleton HH. 1988. Klinische und Lloyd JU, Lloyd CG. 1931b. Kompendium der Schweiz®:
M, Duke J, Kronenberg F. 2001. endokrinologische Cimicifuga Racemosa. Bulletin Fachinformation und
Analysis of thirteen populations Untersuchungen zur Therapie of the Lloyd Library of Botany, Patienteninformation; Zeller
of black cohosh for for- ovarieller Ausfallserscheinungen Pharmacy and Materia Medica Cimifemin®; Omida
mononetin. Altern Ther Health nach Hysterektomie unter 9(30 Pt 2):224-88. Klimaktoplant®; Bioforce
Med 7(3):101-12. Belassung der Adnexe. Z Löhning A, Verspohl EJ, Winterhoff Menosan. Basel: Documed AG.
Kesselkaul O. 1957. Über die Gynäkol 110:611-8. H. 1998. Cimicifuga racemosa in 2891 p.
Behandlung klimakterischer Linde H. 1964. The composition of vitro findings using MCF-7 cells Nesselhut C, Schellhase R, Dietrich
Beschwerden mit Remifemin. Cimicifuga racemosa. 1. On the [abstract]. R, Kuhn W. 1993.
Med Monatsschr 11(2):87-8. isolation of 2 glycosides. Phytopharmakaforschung 2000 Untersuchungen zur proliferativ-
Khan IS, Ganzera M, Bedir E, Khan Arzneimittelforschung 14:1037-9. 72. Abstract nr P07. en Potenz von Phytopharmaka
IA, Walker LA. 2001. Transport Linde H. 1967a. Contents of Löhning A, Verspohl EJ, Winterhoff mit östrogenähnlicher Wirkung
of marker compounds from Cimicifuga racemosa. 2. On the H. 1999. Pharmacological stud- bei Mammakarzinomzellen.
black cohosh across CACO-2 structure of actein. Archiv ies on the dopaminergic activity Gynäkol Onkol 254(1):817-8.
cell monolayers—a model of Pharmazie 300(10):85-92. of Cimicifuga racemosa. In: 23rd Nesselhut T, Liske E. 1999.
human intestinal absorption Linde H. 1967b. Components of international LOF-symposium Pharmacological measures in
[abstract]. In: Botanical dietary Cimicifuga racemosa. 3. on “phyto-oestrogens”; 1999 Jan postmenopausal women with an
supplements: natural products at Configuration of the A, B, and 15; Belgium. Belgium: Univ isopropanolic aqueous extract of
a crossroads; 2001 Nov 8-11; C rings of actein. Archiv Gent. [Pages unavailable]. Cimicifugae racemosae rhizoma.
Asilomar CA. Washington, DC: Pharmazie 300(12):982-92. Löhning A, Winterhoff H. 2000. [abstract]. Menopause 6(4):331.
American Society of Linde H. 1968. Die Inhaltsstoffe von Neurotransmitter concentrations Abstract nr P-8.
Pharmacognosy & Council for Cimicifuga racemosa: 5. Mitt.: after three weeks treatment with Noguchi M, Nagal M, Koeda M,
Responsible Nutrition. p 50. 27-desoxyacetylacteol. Archiv Cimicifuga racemosa [abstract]. Nakayama S, Sakurai N,
Abstract nr P:44. Pharmazie 301:335-41. Phytomedicine 7(Suppl 2):13. Takahira M, Kusano G. 1998.
King J. 1855. American Eclectic Linnaeus C. 1753. Species Abstract nr SL-13. Vasoactive effects of cimicifugic
Obstetrics. Cincinnati: Moore, Plantarum. Volume 1. London: Löser B, Kruse SO, Melzig MF, acids C and D, and fukinolic
Wilstach, Keys. 741 p. Ray Society. 560 p. Nahrstedt A. 2000. Inhibition of acid in Cimicifuga rhizome. Biol
Knüvener E, Korte B, Winterhoff H. Reprinted:1959. neutrophil elastase activity by Pharmaceut Bull 21(11):1163-8.
2000. Cimicifuga and physiolog- Liske E, Boblitz N, Henneicke-von cinnamic acid derivatives from Novy RG, Kobak C, Goffreda J, Vorsa
ical estrogens [abstract]. Zepelin HH. 2000. Therapy of Cimicifuga racemosa. Planta N. 1994. RAPDs identify varietal
Phytomedicine 7(Suppl 2):12. climacteric complaints with Med 66:751-3. misclassification and regional
Abstract nr SL-11. Cimicifuga racemosa; data on Macht DI, Cook HM. 1932. A phar- divergence in cranberry
Korn WD, inventor; IBR Forsch, effect and efficacy from a ran- macological note on Cimicifuga. [Vaccinium macrocarpon (Ait.)
assignee. 1991. 6-month oral tox- domized controlled double- J Am Pharm Assoc 21(4):324-30. Pursh]. Theor Appl Genet
icity study with Remifemin-gran- blind study. In: Rietbrock N, Mahady GB, Fong HHS, Farnsworth 88:1004-10.
ulate in rats followed by an 8- Donath MF, Loew D, Roots I, NR. 2001. Botanical Dietary Oketch-Rabah HA, Mehemi I, Tsai
week recovery period. Germany; Schulz V, editors. Supplements: Quality, Safety MS, Atlas E, Kennelly E,
Patent 20-04-0913-90. Phytopharmaka VI. Darmstadt: and Efficacy. Lisse (ND): Swets Nuntanakorn P, Kronenberg F,
Kraemer H. 1920. Scientific and Steinkopff. p 247-57. & Zeitlinger. 350 p. Lupu R. 2002. Black cohosh
Applied Pharmacognosy. New Liske E, Gerhard I, Wüstenberg P. Mansfield W. 1937. Materia Medica, (Cimicifuga racemosa) does not
York: J Wiley. 741 p. 1997. Phytokombination lindert Toxicology and Pharmacognosy. contain estrogenic activity
Kruse SO, Löhning A, Pauli GF, psychovegetative Leiden. TW St. Louis: CV Mosby. 707 p. [abstract]. 2002 Apr; Boston.
Winterhoff H, Nahrstedt A. Gynäkol 10:172-5. McCoy J, Kelly W. 1996. Survey of Forthcoming.
1999. Fukiic and piscidic acid List PH, Hörhammer L. 1973. Cimicifuga racemosa for phytoe- Osol A, Farrar GE. 1947. The
esters from the rhizome of Hagers Handbuch der strogenic flavonoids. In: 212th Dispensatory of the United
Cimicifuga racemosa and the in Pharmazeutischen Praxis. ACS national meeting; 1996 States of America. 24th ed.
vitro estrogenic activity of fuki- Berlin: Springer. 1271 p. Aug 25-29; Orlando FL. [Pages Philadelphia: JB Lippincott.
nolic acid. Planta Med Liu J, Burdette JE, Xu H, Gu C, unavailable]. 1928 p.
65(8):763-4. Breemen RB van, Bhat KP, McGuffin M, Hobbs C, Upton R, Pethö A. 1987. Klimakterische

American Herbal Pharmacopoeia™ • Black Cohosh Rhizome • 2002 35


Beschwerden. Umstellung einer Cordell GA, Bowman M, markers to identify plant species development. Boulder (CO):
Hormonbehandlung auf ein Lemmo E. 2000. Triterpene gly- and variants [abstract]. Botanical Herb Research Foundation.
pflanzliches Gynäkologikum cosides from Cimicifuga race- dietary supplements: natural Available from: Yellow Creek
möglich? Arzt Praxis 38:1551-3. mosa. J Nat Prod 63:905-10. products at a crossroads; 2001 Botanical Institute, Robbinsville
Piancatelli G. 1971. Nuovi triterpeni Steinberg KK, Thacker SB, Smith SJ, Nov 8-11; Asilomar CA. (NC). 102 p.
dall’Actea racemosa. Gazz Chim Stroup DF, Zack MM, Flanders Washington, DC: American [NF] The National Formulary. 1936.
Ital 101(2):139-48. WD, Berkelman RL. 1991. A Society of Pharmacognosy & The National Formulary. 6th ed.
Radics L, Kajtar-Peredy M, Corsano meta-analysis of the effect of Council for Responsible Washington DC: Am Pharm
S, Standoli L. 1975. Carbon-13 estrogen replacement therapy on Nutrition. p 32. Abstract nr P:8. Assoc.
NMR spectra of some polycyclic the risk of breast cancer. JAMA Yamahara J, Kobayasi M, Kimura H, [USPF] United States Pharmacopeial
triterpenoids. Tetrahed Let 265(15):1985-90. Miki K, Kozuka M, Sawada T, Forum. 2001. Black cohosh.
48:4287-90. Steinegger E, Hänsel R. 1992. Fujimura H. 1985. Biologically Pharmacopeial Previews.
Ramsey GW. 1965. A biosystematic Pharmakognosie. 5th ed. Berlin: active principles of crude drugs. Volume 27(4) 2001 Jul-Aug.
study of the genus Cimicifuga Springer. 804 p. The effect of Cimicifugae rhi- Rockville (MD): United States
(Ranunculaceae) [dissertation]. Stoll W. 1987. Phytotherapeutikum zoma and its constituents in pre- Pharmacopeial Convention, Inc.
Tenn: Univ Tennessee. 265 p. beeinflusst atrophisches ventive action on the carbon
Available from: Missouri Vaginalepitel. Therapeutikon tetrachloride-induced liver disor-
Botanical Garden Library. 1:23-31. der in mice. Shoyakugaku Zasshi
Ramsey GW. 1987. Morphological Stolze H. 1982. Der andere Weg, 39(1):80-4.
considerations in the North Klimakterische Beschwerden zu Yen KY. 1992. The Illustrated
American Cimicifuga behandlen. Gyne 3:14-6. Chinese Materia Medica: Crude
(Ranunculaceae). Castanea Struck D, Tegtmeier M, and Prepared. Translator:
52(2):129-41. Harnischfeger G. 1997. Flavones Wiseman N. Taipei: SMC. 383
Ramsey GW. 1997. Cimicifuga. In: in extracts of Cimicifuga race- p. Translation of: The Illustrated
Flora of North America Editorial mosa. Planta Med 63:289. Chinese Materia Medica: Crude
Committee, editors. Flora of Vorberg G. 1984. Therapie and Prepared.
North America. Volume 3. Klimakterischer Beschwerden. Youngken H. 1930. A Text Book of
Oxford: Oxford Univ Pr. p 177- Erfolgreiche hormonfreie Pharmacognosy. Philadelphia: P
81. Therapie mit Remifemin®. Z Blakiston’s Son. 817 p.
Reynolds JEF. 1996. Martindale: The Allgem 60:626-9. Zava DT, Dollbaum CM, Blen M.
Extra Pharmacopoeia. 31st ed. Wagner H, Bladt S. 1996. Plant Drug 1998. Estrogen and progestin
London: Roy Pharm Soc. 2739 Analysis. 2nd ed. Berlin: bioactivity of foods, herbs, and
p. Springer. 384 p. spices. Proc Soc Exp Biol Med
Romm A. 2001. Treatment of incom- Warnecke G. 1985. Beeinflussung 217(3):369-78.
plete miscarriage with botanical klimakterischer Beschwerden Zhang X. 1998. Regulatory situation
therapies. J Am Herb Guild durch ein Phytotherapeutikum. of herbal medicines: a world-
2(2):16-7. Med Welt 36:871-4. wide review. Geneva (Switz):
Sayre LE. 1917. A Manual of Weirmont GT. 1985. Use of statistics Traditional Medicine
Organic Materia Medica and to develop and evaluate analyti- Programme. Available from:
Pharmacognosy. 4th ed. cal methods. Arlington (VA): Marketing and Dissemination,
Philadelphia: P Blakiston’s Son. AOAC. [Pages unavailable]. World Health Organization,
606 p. White H. 1903. Materia Medica, Geneva 27, Switzerland. 45 p.
Schairer C, Lubin J, Troisi R, Pharmacy, Pharmacology, [BHP] British Herbal
Sturgeon S, Brinton L, Hoover Therapeutics. 5th ed. Pharmacopoeia. 1996. British
R. 2000. Menopausal estrogen Philadelphia: P Blakiston’s Son. Herbal Pharmacopoeia. 4th ed.
and estrogen-progestin replace- 744 p. Exeter (UK): Brit Herb Med
ment therapy and breast cancer Whitlaw C. 1829. New Medical Assoc.
risk. JAMA 283(4):485-91. Discoveries With a Defence of [BP] British Pharmacopoeia. 1993.
Schindler H. 1992. The constituents the Linnaean Doctrine. Volume British Pharmacopoeia. Volume
of medicinal plants and testing 1. London: Whitlaw C. p 271. 2. London: Report of Health
methods for botanical tinctures. Wood GB, Bache F. 1833. The Scottish House and Health
Arzneimittelforschung 42:547-9. Dispensatory of the United Department, Welsh Office.
Schmitz U. 1993. Non–steroidal anti States of America. 1st ed. [GSL] General Sale List. 1984-94.
estrogen and alkaloids from rhi- Philadelphia: Grigg Elliot. 1073 Statutory Instrument (SI) The
zome Cimicifuga racemosa p. Medicines (Products other than
(Ranunculaceae) [dissertation]. Wood HC, LaWall C, Youngken veterinary drugs). London: Her
Witten, Germany: University of HW, Osol A, Griffith I, Majesty’s Stationery Office;
Witten/Herdecke. 221 p. Gershenfeld L. 1937. The Report nr: SI no 769, as amend-
Available from: University of Dispensatory of The United ed 1985; SI no 1540, 1990; SI
Witten/Herdecke. States of America. 22nd ed. no 1129, 1994; SI no 2410.
Scudder JM. 1903. Specific Philadelphia: JB Lippincott. [HC] Health Canada. 1995. Drugs
Medications and Specific 1894 p. directorate guidelines: traditional
Medicines. 15th ed. Cincinnati: Wuttke W, Jarry H, Heiden I, herbal medicines. Ottawa ON
Scudder Bros. 432 p. Westphalen S, Seidlová-Wuttke (Canada): Minister National
Seidlová-Wuttke D, Jarry H, Heiden D, Christoffel V. 2000. Selective Health Welfare, Canada.
I, Wuttke W. 2000. Effects of estrogen receptor modulator [HC] Health Canada. 2001. Drug
Cimicifuga racemosa on estrogen (SERM) activity of the product database product infor-
dependent tissues [abstract]. Cimicifuga racemosa extract mation: Cimicifuga homaccord
Phytomedicine 7(Suppl 2):11. BNO 1055: pharmacology and heel-044 DPS. Ottawa: Health
Abstract nr SL-9b. mechanisms of action [abstract]. Canada Therapeutic Products
Seidlová-Wuttke D, Wuttke W. 2000. Phytomedicine 7(Supp 2):12. Programme. Available from:
Selective estrogen receptor mod- Abstract nr SL-10. www.hc-sc.gc.ca/drug2/
ular activity of Cimicifuga race- Xu H, Fabricant DS, Johnson HE, product/p12531.html. 1 p.
mosa extract: clinical data Piersen CE, Bolton JL, [HRF] Herb Research Foundation,
[abstract]. Phytomedicine Farnsworth NR, Constantinou [YCBI] Yellow Creek Botanical
7(Suppl 2):11. Abstract nr SL-9a. AI. 2001. Using random ampli- Institute. 2001 Aug. Appalachian
Shao Y, Harris A, Wang M, Zhang H, fied polymorphic DNA (RAPD) botanicals: market analysis and

36 American Herbal Pharmacopoeia™ • Black Cohosh Rhizome • 2002


Black cohosh (Actaea racemosa)
Source: Weihe, Plantae officinales oder Sammlung officineller
Pflanzen (1812-1828).
Courtesy of Hunt Institute for Botanical Documentation, Carnegie Mellon University,
Pittsburgh, PA

American Herbal Pharmacopoeia™ • Black Cohosh Rhizome • 2002


TABLE OF CONTENTS
Nomenclature 1
Botanical Nomenclature
Botanical Family
Definition
Common Names

History 1
Identification 3
Botanical Identification
Macroscopic Identification
Microscopic Identification

Commercial Sources and Handling 8


Collection
Cultivation
Handling and Processing
Drying
Storage
Adulterants
Preparations

Constituents 9
Analytical 10
High Performance Thin Layer Chromatography (TLC/HPTLC)
High Performance Liquid Chromatography (HPLC)
Quantitative Standards

Therapeutics 16
Pharmacokinetics
Pharmacodynamics and Clinical Efficacy
Effects on Menopausal and Premenstrual Symptoms
Hormonal and Central Nervous System Effects
Other Effects
Conclusion
Medical Indications Supported by Clinical Trials
Medical Indications Supported by Traditional or Modern Experience
Actions
Substantiation for Structure and Function Claims
Dosages

Safety Profile 30
Side Effects
Contraindications
Precautions
Interactions
Pregnancy, Mutagenicity, and Reproductive Toxicity
Lactation
Carcinogenicity
Influence on Driving
Precautions
Overdose
Treatment of Overdose
Toxicology
Classification of the American Herbal Products Association

International Status 32
References 34

American Herbal Pharmacopoeia™


PO Box 5159
Santa Cruz, CA 95063 US
Tel: 1-831-461-6318
Fax: 1-831-475-6219
Email: ahpadmin@got.net
Website: www.herbal-ahp.org

,!7IB9C9-ecfbed!

You might also like