ArticlePDF AvailableLiterature Review

Abstract

Cimicifuga racemosa has long been advocated as an alternative to hormone-replacement therapy. However, recent experimental studies and a clinical trial have raised some questions on its utility. The aim of this systematic review is principally to summarise the trial evidence regarding the efficacy of C. racemosa in the treatment of menopausal symptoms. In addition, we also explore the evidence relating to the mechanism of action of this herbal medicine. Searches of seven computerised databases were performed to identify all randomised clinical trials (RCTs) on C. racemosa as well as animal and in vitro experiments. No language restrictions were imposed. Data were extracted by both authors according to predefined criteria and are summarised in narrative form. Four RCTs of C. racemosa as a treatment of menopausal symptoms were included. They yielded no compelling evidence for the efficacy of C. racemosa on menopausal symptoms. The small beneficial effects observed in some studies could be explained by a central activity, and an oestrogenic activity cannot be completely excluded. In spite of plausible mechanisms of action of C. racemosa, its clinical efficacy for the treatment of menopausal symptoms has not been convincingly demonstrated through rigorous clinical trials. Additional rigorous RCTs and biochemical and chemical investigations are warranted.
REVIEW ARTICLE
Francesca Borrelli ÆEdzard Ernst
Cimicifuga racemosa
: a systematic review of its clinical efficacy
Received: 15 October 2001 / Accepted in revised form: 1 March 2002 / Published online: 7 June 2002
ÓSpringer-Verlag 2002
Abstract Background:Cimicifuga racemosa has long
been advocated as an alternative to hormone-replace-
ment therapy. However, recent experimental studies and
a clinical trial have raised some questions on its utility.
The aim of this systematic review is principally to
summarise the trial evidence regarding the efficacy of
C. racemosa in the treatment of menopausal symptoms.
In addition, we also explore the evidence relating to the
mechanism of action of this herbal medicine.
Methods: Searches of seven computerised databases
were performed to identify all randomised clinical trials
(RCTs) on C. racemosa as well as animal and in vitro
experiments. No language restrictions were imposed.
Data were extracted by both authors according to
predefined criteria and are summarised in narrative
form.
Results: Four RCTs of C. racemosa as a treatment of
menopausal symptoms were included. They yielded no
compelling evidence for the efficacy of C. racemosa on
menopausal symptoms. The small beneficial effects
observed in some studies could be explained by a central
activity, and an oestrogenic activity cannot be
completely excluded.
Conclusions: In spite of plausible mechanisms of action
of C. racemosa, its clinical efficacy for the treatment of
menopausal symptoms has not been convincingly dem-
onstrated through rigorous clinical trials. Additional
rigorous RCTs and biochemical and chemical investi-
gations are warranted.
Keywords Herbal medicine ÆCimicifuga racemosa Æ
Black cohosh
Introduction
Menopause is characterised by the cessation of men-
struation and by the appearance of many vasomotor,
vaginal and psychological symptoms including hot
flashes, profuse sweating, flushing of the skin, atrophic
vaginitis, irritability and depression. After menopause
the risks of coronary heart disease and osteoporosis in-
crease. These effects are linked to hormonal changes,
including oestrogen deficiency [1]. Currently, hormone-
replacement therapy (HRT) is the most common
pharmacological treatment for relief of menopausal
symptoms. Unfortunately HRT is associated with
endometrial or breast cancer and adverse effects [2, 3, 4,
5]. Therefore, there is a buoyant market for alternative
therapies.
The herbal medicine Cimicifuga racemosa is often
promoted as such an alternative. It is a perennial herb
native to North America commonly known as ‘‘wom-
an’s herb’’. Extracts of the rhizome of C. racemosa have
long been used for the relief of pain during menses or
childbirth and for the treatment of dyspepsia, dysmen-
orrhea, epilepsy, kidney ailments, malaria, rheumatism
and rheumatic pain [6, 7, 8, 9]. Today C. racemosa is the
principal herbal remedy in for menopausal problems.
An ethanolic extract of the rhizome of this plant stan-
dardised to contain 1 mg of triterpenes calculated as 27-
deoxyacteine per 20-mg tablet (trade name Remifemin)
is widely marketed for the relief of climacteric (meno-
pausal) disorders including hot flushes and profuse
sweating. C. racemosa rhizome contains numerous
chemical components including triterpene glycosides
(actein, 27-deoxyactein, cimicifugoside), phenolic acids
(isoferulic acid, fukinolic acid), flavonoids, volatile oils
and tannins. The effects of C. racemosa are believed to
be the result of complex synergistic action of these
components. The aim of this systematic review is (i) to
Eur J Clin Pharmacol (2002) 58: 235–241
DOI 10.1007/s00228-002-0457-2
F. Borrelli
Department of Experimental Pharmacology,
University of Naples ‘‘Federico II’’,
Via D. Montesano 49, 80131 Naples, Italy
E. Ernst (&)
Department of Complementary Medicine,
School of Postgraduate Medicine and Health Studies,
University of Exeter, EX2 4NT, UK
E-mail: E.Ernst@exeter.ac.uk
Tel.: +44-392-430802
Fax: +44-392-424989
summarise the evidence for or against the efficacy of
C. racemosa in providing relief of menopausal symptoms
or other conditions and (ii) to summarise our knowledge
regarding its possible mechanism of action.
Methods
Computerised literature searches were performed to identify all
randomised controlled trials (RCTs) as well as animal and in vitro
experiments on C. racemosa. Databases included Medline, Em-
base, Amed, Phytobase, PubMed, CISCOM (Research Council for
Complementary Medicine, London) and Cochrane Library (all
from their respective inceptions to July 2001). In addition, several
(n=3) manufacturers of C. racemosa preparations were asked to
contribute published or unpublished material, and our own files
were hand searched. Bibliographies of the articles thus located
were scanned for further relevant publications. No language
restrictions were imposed. The search terms used are shown in
Table 1.
Clinical trials of C. racemosa were included in this review only if
performed randomised, blind and controlled. Clinical trials for any
indication were included. Animal and in vitro experiments on
C. racemosa were included only if performed to evaluate its
oestrogen-like activity. Studies of this plant in combination prod-
ucts were excluded.
Data were validated and extracted in a standardised, predefined
manner (Table 2). The methodological quality of clinical studies
was assessed using the scoring system developed by Jadad and
colleagues (Table 2) [10].
Results
Several clinical studies examined the effect of C. race-
mosa in menopausal women [11, 12, 13, 14, 15, 16, 17,
18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29]. Only four of
these met our inclusion/exclusion criteria [21, 22, 24, 29].
Key data of RCTs are summarised in Table 3. All trials
scored at least two of five points on the Jadad score.
Three studies reporting positive results in favour of
C. racemosa scored two, three and two points, respec-
tively [21, 22, 24]. One negative trial scored five points
[29]. The trials are described in some detail below. All
trials used a commercial product of C. racemosa, Rem-
ifemin (dosage 40–80 mg).
The first RTC on Remifemin was conducted by
Warnecke on 60 women with menopause in 1985 [21].
The women received 40 drops twice daily of C racemosa
(80 mg/day), conjugated oestrogens (0.625 mg/day) or
diazepam (2 mg/day) for 4 months. The outcome mea-
sures were: modified Kupperman menopausal index,
self-evaluation depression scale, Hamilton anxiety scale
and vaginal epithelium status. C. racemosa produced a
decrease in climacteric complaints and oestrogen-like
stimulation of the vaginal mucosa similar to conjugate
oestrogens. Cytological examinations of vaginal epi-
thelium proliferation revealed a similar percentage of
eosinophilic epithelial cells with C. racemosa and
conjugated oestrogens. The women treated with
C. racemosa extract had a greater percentage of pyknotic
nuclei than those on conjugated oestrogens. Greater
improvements in neurovegetative symptoms were noted
in the experimental group.
A double-blind trial with 80 women aged between
46 years and 58 years compared the effects of a stan-
dardised C. racemosa extract both with placebo and low-
dose oestrogen [22]. The women received two tablets of
extract (80 mg), conjugated oestrogens (0.625 mg) or
identical placebo twice daily for 3 months. The outcome
measures were: Kupperman menopausal index, mea-
suring mainly neurovegetative symptoms, Hamilton
anxiety scale for psychological complaints (both re-
corded every 4 weeks) and proliferation status of vaginal
epithelium (recorded at the beginning and the end of the
study). At the end of treatment all groups showed im-
provements compared with baseline. Since the oestrogen
dose proved to be too low, only the difference between
placebo and C. racemosa were evaluated. C. racemosa
significantly improved all parameters under investiga-
Table 1. Terms used for computer search and number of papers found. n.r. not reported, CISCOM Research Council for Comple-
mentary Medicine, London
Search terms Embase Amed PubMed
or Medline
Phytobase CISCOM Cochrane
Library
Cimicifuga racemosa (botanical name) 70 21 n.r. 8 4
Black cohosh (common English name) 9 18 n.r. 5 1
Actaea racemosa (first botanical name) 4 1 n.r. 2 0
Actein (active ingredient) 3 6 n.r. 1 0
Cimicifugoside (active ingredient) 11 8 n.r. 2 0
Cimicifugic acid (active ingredient) 6 4 n.r. 2 0
Fukinolic acid (active ingredient) 6 4 n.r. 2 0
Remifemin (trade name) 15 6 n.r. 0 0
Cimisan 2 0 n.r. 0 0
Cimicifuga (common Italian name) 120 58 n.r. 21 5
Traubensilberkerze (common German name) 4 0 n.r. 1 0
Wanzenkraut (common German name) 0 0 n.r. 0 0
Black snakeroot (common English name) 0 0 n.r. 0 0
Bugbane (common English name) 0 0 n.r. 0 0
Rattleroot (common English name) 0 0 n.r. 0 0
Rattletop (common English name) 0 0 n.r. 0 0
Rattleweed (common English name) 0 0 n.r. 0 0
236
tion, compared with placebo. The final score of the
menopausal index in women treated with the extract was
below 15, which the author considered to be a clinically
relevant result.
Lehmann-Willenbrock and Riedel [24] studied the
effect of estriol, conjugated oestrogens, oestrogen–gest-
agen sequential therapy or a C. racemosa extract on 60
hysterectomised women who all had at least one intact
ovary and complained of climacteric symptoms. The
extract (two tablets, 80 mg/day) was administered twice
daily for 6 months. The dose of estriol and conjugated
oestrogens was 1 mg/day and 1.25 mg/day, respectively,
while the dose of oestrogen–gestagen was not specified.
The outcome measures were: modified Kupperman in-
dex and serum concentrations of follicle-stimulating
hormone (FSH) and luteinising hormone (LH). These
variables were measured after 4, 8, 12 and 24 weeks. In
Table 2. Jadad scoring system to measure methodological quality
of clinical trials
Each ‘yes’ 1 point; each ‘no’ 0 points
A. Study described as randomised (includes the use of words
such as random, randomly and randomisation)?
B. Study described as double-blind?
C. Description of withdrawals and dropouts?
D. Method of generating the sequence of randomisation
described and appropriated (table of random numbers,
computer-generated etc.)?
E. Method of double-blinding described and appropriated
(identical placebo, active placebo, dummy etc.)?
Deduct 1 point if
F. Method to generate the sequence of randomisation described
and inappropriate (patients were allocated alternately,
or according to their date of birth, hospital number etc.)
G. Method of double-blinding described and inappropriate
(e.g. comparison of tablet versus injection with no double
dummy)
Table 3. Clinical studies. CE coniugated oestrogens, EG oestrogen + gestagen, FSH follicle-stimulating hormone, LH luteinising
hormone
First author,
year
(reference)
Jadad
score
Design Patients
(number)
Dosage
of extract
Control
treatments
(dosage)
Length of
treatment
(months)
Outcome
measures
Main
results
Comments
Warnecke,
1985 [21]
2 Randomised,
open trial
(3 groups)
60 Meno-
pausal
women
40 Drops
twice
daily
(80 mg)
CE (0.625
mg/day),
diazepam
(2 mg/day)
3 Modified
Kupperman
menopausal
index; self-
evaluation
depression
scale;
Hamilton
anxiety scale;
vaginal
epithelium
status
C. racemosa
improved
all parameters
and increased
vaginal
ephitelium
proliferation
as well as CE
Open study,
no placebo
control
Stoll,
1987 [22]
3 Randomised,
double-blind
trial (3 groups)
80 Women
with
climacteric
complaints
Two tablets
twice daily
(80 mg)
Placebo,
oestrogen
(0.625
mg/day)
3 Kupperman
menopausal
index;
Hamilton
anxiety scale;
vaginal
epithelium
status
C. racemosa
improved
all parameters
compared
with placebo
Lack of
oestrogen
effects
Lehmann-
Willenbrock,
1988 [24]
2 Randomised,
open trial
(4 groups)
60 Hyste-
rectomised
women with
at least one
intact ovary
Two tablets
twice daily
(80 mg)
Estriol
(1 mg/day),
CE (1.25
mg/day),
EG
6 Modified
Kupperman
menopausal
index; FSH
and LH levels
In all groups
Kupperman
index
decreased.
Differences
between
groups
were not
significant.
Only CE and
EG reduced
FSH and
LH levels
Open study,
no placebo
group
Jacobson
2001 [29]
5 Randomised,
double-blind
trial (2 groups)
26 Breast
cancer
survivors
One tablet
twice
daily
(40 mg)
Placebo 2 Menopausal
symptom
index; FSH
and LH levels
In all groups
menopausal
symptoms
improved.
No effect on
FSH and LH.
No difference
between
groups
Length of
treatment
too short
237
Animal experiments In vitro experiments
First author,
year (reference)
Test(s) used Main result First author,
year (reference)
Test(s) used Main result
Gizicky 1944 [30] Uterine weight,
ovary weight
CR increased
uterine and
ovary weight
Jarry 1985 [33] ER binding assay A fraction bound
to ER
Foldes 1959 [13] Uterine weight,
induction of oestrus,
sedative effect,
changes in ovaries
and thyroid function
CR increased
the uterine weight
and induced oestrus
Duker 1991 [25] ER binding assay Whole extract did not
bind to ER. Some
fractions bound
to ER
Siess 1960 [31] Uterine weight,
induction of oestrus
No effect
was observed
Nesselhut 1993 [42] Cell proliferation CR inhibited cell
proliferation at
concentration
>2.5 lg/ml
Jarry 1985 [32] FSH levels,
LH levels,
prolactin levels
A fraction reduced
LH levels
Jarry 1996 [43] OPS CR inhibited OPS
in porcine
Jarry 1985 [33] LH levels CHCl
3
fraction
reduced LH levels
Harnischfeger
1996 [44]
ER binding assay,
cell proliferation
CR bound to ER,
increased the cell
proliferation at 1–5
lg/ml and decreased
it at >10 lg/ml
Duker 1991 [25] LH levels CR reduced
LH levels
Eagon 1996 [45] ER binding assay CR did not bind
to ER
Einer-Jensen
1996 [34]
Vaginal
cornification,
uterine weight
CR did not show
estrogenic effects
Zava 1998 [46] ER/PR binding
assay, T47D cell
proliferation
CR did not bind to
ER/PR and did not
modify cell
proliferation
Eagon, 1997 [35] Uterine weight,
c-myc expression,
serum CP levels
and hepatic
CP mRNA levels
CR root increased
uterine weight
and CP levels
Lohning 1998 [47] MCF-7 cell
proliferation
CR increased cell
proliferation
to 0.1–10 lg/ml.
100 lg/ml were
ineffective
Eagon 1999 [36] Uterine weight,
LH levels
CR increased
uterine weight and
decreased LH levels
Lohning 1999 [38] Basal and TRH
stimulated PS
CR reduced both
basal and TRH
stimulated PS
Jarry 1999 [37] LH levels, uterine
weight and ERa,
IgF1, C3, collagen I,
osteocalcin
expression
CR either reduced
LH levels, collagen I
and osteocalcin
expression or
increased ERa
expression
Dixon Shanies
1999 [48]
T47D cells
proliferation
CR inhibited cell
proliferation to
0.1–1 % v/v
Lohning 1999 [38] Body temperature,
ketamine-induced
sleeping
CR decreased
body temperature
and prolonged
sleeping time
Eagon 1999 [36] ER binding assay CR bound ER
Freudenstein
2000 [39]
Number and size
of tumours, FSH,
LH, prolactin levels,
organ weights
CR did not show
oestrogen effects
Jarry 1999 [37] ER binding assay,
luciferase
expression in a
MCF7-a-andb
ER expressing
subclone
CR bound to ER
and activated the
transcription of
oestrogen-regulated
genes
Nisslein 2000 [40] PYR and DPD
levels, femoral
density
CR reduced
PYR/DPD levels
and bone loss
Freudenstein
1999 [49]
MCF-7 cells
proliferation
CR inhibited
cells proliferation
Liu 2001 [41] Days of oestrus,
uterine and
ovary weight
CR increased the
days of oestrus
Liu 2001 [50] ER aand b
binding assay
CR did not show
activity
Liu 2001 [41] MCF-7 cells
proliferation,
ER expression
CR increased
cells proliferation
and ER expression
Table 4. Animal and in vitro experiments to investigate the Cim-
icifuga racemosa (CR) mechanism of action. ER oestrogen recep-
tor, PR progesterone receptor, LH luteinising hormone, FSH
follicle-stimulating hormone, OPS ovarian-progesterone secretion,
PYR pyridinoline, DPD deoxypyridinoline, CP ceruloplasmin, PS
prolactin secretion, n.d. not described, n.c. no comment
238
all groups, a decrease of the modified Kupperman index
was observed within 4 weeks after the onset of therapy.
There was no significant inter-group difference regarding
the Kupperman index at the end of therapy. Only con-
jugated oestrogens and oestrogen–gestagen sequential
therapy significantly modified the serum concentration
of FSH and LH.
A recent RCT included 26 female breast cancer
survivors [29]. Each woman took one tablet of C. race-
mosa (Remifemin 40 mg) or placebo twice daily with
meals for 60 days. At the start and completion of the
study, patients completed a detailed menopausal
symptom index (heart palpitations, excessive sweating,
headaches, poor sleep, depression and irritability or
nervousness). The number and intensity of hot flashes
were recorded in a diary 3 days before baseline, on
days 7–30 and on days 57–60. FSH and LH levels were
measured in a subset of patients at both the start and
the end of the medication phase. The results showed
that both the treatment and placebo groups improved.
No statistically significant differences in blood levels of
FSH and LH were noted. Except for a reduced number
of sweating episodes, no significant beneficial clinical
effects of C. racemosa compared with placebo were
observed.
A total of 14 animal and 15 in vitro studies of
C. racemosa met our inclusion/exclusion criteria [13, 25,
30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45,
46, 47, 48, 49, 50, 51]. Key data of all studies are sum-
marised in Table 4. Experimental studies were per-
formed on the capacity of C. racemosa to modulate
either the plasma hormone levels (LH, FSH), the uterine
weight and induction of oestrus in animals or the cell
proliferation, the oestrogen-receptor expression and
competitive binding assay in vitro. The results fail to
yield a uniform picture as the mechanism of action of
C. racemosa.
Discussion
C. racemosa was approved by the German Commission
E for premenstrual discomfort, dysmenorrhea or cli-
macteric (menopausal) neurovegetative ailments. The
data summarised above do, however, not conclusively
demonstrate its efficacy. Most clinical trials were not
well designed. Some were not controlled or randomised
and included heterogeneous subject samples [11, 12, 13,
14, 15, 16, 17, 18, 19, 20, 23, 25, 26, 27, 28]. Only four
RCTs were located [21, 22, 24, 29]. Three of these trials
suggested that an extract of C. racemosa had therapeutic
benefit on menopausal symptoms improving the
Kupperman menopausal index and Hamilton anxiety
scale [21, 22, 24]. Trials with active controls suggested
that the extract had similar effects to oestrogen
replacement therapy [21, 24]. Neither of these studies
was designed as an equivalence trial in the strict sense. In
the third trial, placebo and oestrogen showed almost the
same beneficial effects on the Kupperman index and
Hamilton anxiety scale [22]; and the ineffective oestro-
gen dosage (0.625 mg/day) used was similar to the
dosage (0.625 mg/day) used by Warnecke [21]. It there-
fore seems possible that the beneficial effect observed by
Warnecke [21] and Lehmann-Willenbrock and Riedel
[24] was due to a placebo effect.
The recent RCT by Jacobson et al. [29] suggested
C. racemosa to be ineffective on menopausal symptom
index. A large placebo response in menopausal symp-
toms and no significant difference between the placebo
and C. racemosa were observed in this RCT. The only
significantly reduced number of sweating episodes was,
according to the authors, a chance finding. The lack of
effect of C. racemosa is unlikely to be due to the short
duration of therapy (2 months); in a previous RCT,
Lehmann-Willenbrock and Riedel [24] reported notice-
able benefits of C. racemosa within 4 weeks The lack of
rigorous long-term studies is, however, a clear limitation
of the evidence available to date.
Early animal studies on an extract of C. racemosa had
postulated ‘‘oestrogen-like’’ activity as evidenced by an
increase in uterine weight and an induction of oestrus
[13, 30]. Further studies [25, 32, 33, 37] suggested that
C. racemosa contained three synergistically acting com-
pounds able to reduce serum LH levels and bind to
oestrogen receptors (so as to increase the amount of
oestrogen in the blood which decreases the menopause
symptoms). Actein and cimicifugoside were believed to
be partly responsible for the reduction in LH, while the
isoflavone formononetin was thought to bind to oes-
trogen receptors. However, recent investigations imply
that C. racemosa contains compounds that act by a
mechanism that does not involve oestrogen receptors
[39, 46, 50]. The effects seem partly to be due to a central
activity mediated by dopaminergic-2 receptors rather
than to an oestrogenic activity [38]. Some investigators
have found that an extract of C. racemosa inhibited the
proliferation of human oestrogen receptor-positive
breast cancer cells (T47D and MCF-7) [42, 48, 49] and
increased oestrogen receptor levels [37, 41]. Since dop-
aminergic agonists also cause a significant decrease in
the proliferation of MCF-7 cells [51] and D1-like dop-
amine receptors mediate the in vitro transcriptional ac-
tivation of oestrogen receptors by dopamine [52, 53], the
effect of the extract could be due to the presence of
dopaminergic substances. This theory is further sup-
ported by chemical studies reporting that ethanolic and
isopropanolic extracts do not contain the isoflavone
formononetin and the flavone kaempferol [54, 55]. These
substances are concentrated in the aerial parts of the
plant. As some of the early experiments did not use
standardised extracts, the detection of formononetin and
the effect of C. racemosa on oestrogen receptors and LH
levels observed in these investigations could be due to
contaminants.
In conclusion, the notion that C. racemosa has ben-
eficial effects on menopausal symptoms is not supported
by the evidence of rigorous RCTs. If an effect on
menopausal symptoms exists, it is probably due to an
239
action discrete from oestrogen receptor activity. Further
work is needed to test the efficacy of C. racemosa for
menopausal symptoms and to assess its long-term
effects. In addition, more work is required to identify the
pharmacologically active compounds of C. racemosa
and their actions.
Acknowledgements The authors wish to thank the University of
Naples Federico II for its support.
References
1. World Health Organization Scientific Group (1981) Research
on the menopause. World Health Organization Technical
Report Series, no. 670. World Health Organization, Geneva
2. Whitehead MI, Townsend PT, Davies-Pryse J, Ryder TA,
King RJ (1981) Effects of estrogens and progestins on the
biochemistry and morphology of the postmenopausal
endometrium. N Engl J Med 305:1599–1685
3. Rubin GL, Peterson HB, Lee NC, Maes EF, Wingo PA,
Becker S (1990) Estrogen replacement therapy and the risk of
endometrial cancer. Remaining controversies. Am J Obstet
Gynecol 162:148–154
4. Bergkvist L, Persson I (1996) Hormone replacement therapy
and breast cancer. A review of current knowledge. Drug Saf
15:360–370
5. Murray MT, Pizzorno JE (1999) Menopause. In: Murray MT,
Pizzorno JE (eds) Textbook of natural medicine. Churchill
Livingstone, Kenmore, pp 1387–1396
6. Beuscher N (1995) Cimicifuga racemosa L. Black cohosh. Z
Phytother 16:301–310
7. Liske P (1998) Therapeutic efficacy and safety of Cimicifuga
racemosa for gynecologic disorders. Adv Ther 15:45–53
8. Foster S (1999) Black cohosh: Cimicifuga racemosa. A litera-
ture review. Herbal Gram Winter pp 35–50
9. Bobliz VN (2000) The efficacy, effect and safety of cimicifuga
racemosa in gynecology. Dtsch Apoth Ztg 140:107–114
10. Jadad AR, Moore RA, Carrol D, Jenkinson C, Reynolds DJM,
Gavaghan DJ, et al (1996) Assessing the quality of reports of
randomised clinical trials: is blinding necessary? Control Clin
Trials 17:1–12
11. Kesselkaul O (1957) Uber die Behandlung klimakterischer
Beschwerden mit Remifemin. Med Mschr 11:87–88
12. Kramer H (1958) Erfahrungen mit dem Cimicifuga-praparat
Remifemin. Ther d Gegenw 97:238
13. Foldes J (1959) The actions of an extract of Cimicifuga race-
mosa. Arzneimittelforschung 13:623–624
14. Stiehler K (1960) Uber die Anwendung eines standardisierten
Cimicifuga-Auszuges in der Gynakologie. Arztl Prax 11:916–
917
15. Heizer H (1960) Kritisches zur Cimicifuga. Therapie bei hor-
monalen storungen der frau. Med Klin 55:232–233
16. Brucker A (1960) Beitrag zur phytotherapie hormonaler
storungen der frau. Med Welt 44:2331–2333
17. Gorlich N (1962) Behandlung ovarieller storungen der frau in
der allgemein-praxis. Arztl Prax 14:1742–1743
18. Stolze H (1982) An alternative to treat menopausal complaints.
Gyne 3:14–16
19. Daiber W (1983) Climacteric complaints: success without using
hormones – a phytotherapeutic agent lessens hot flushes,
sweating and insomnia. Arztliche Praxis 35:1946–1947
20. Vorberg G (1984) Therapy of climateric complaints. Zeitschrift
fur Allgemeinmedizin 60:626–629
21. Warnecke G (1985) Influence of a phytopharmaceutical on
climacteric complaints. Die Meizinisch Welt 36:871–874
22. Stoll W (1987) Phytopharmacon influences atrophic vaginal
epithelium: double-blind study – Cimicifuga vs estrogenic
substances. Therapeuticum 1:23–31
23. Petho A (1987) Climacteric complaints are often helped with
black cohosh. Arztliche Praxis 47:1551–1553
24. Lehmann Willenbrock E, Riedel HH (1988) Clinical and en-
docrinologic examinations about therapy of climateric symp-
toms following hysterectomy with remaining ovaries. Zentralbl
Gynakol 110:611–618
25. Duker EM, Kopanski L, Jarry H, Wuttke W (1991) Effects of
extracts from Cimicifuga racemosa on gonadotropin release in
menopausal women and ovariectomized rats. Planta Med
57:420–424
26. Schaper and Brummer GmbH (1997) Remifemin: a plant based
gynecological agent (scientific brochure). Schaper and
Brummer GmbH., Salzgitter
27. Liske E, Wustenberg P (1998) Therapy of climacteric com-
plaints with Cimicifuga racemosa: herbal medicine with clini-
cally proven evidence. Menopause 5:250
28. Nesselhut T, Liske E (1999) Pharmacological measures in
postmenopausal women with an isopropanolic aqueous extract
of Cimicifuga racemosae rhizome (abstract). 10th annual
meeting of the North American Menopause Society, New York
29. Jacobson JS, Troxel AB, Evans J, Klaus L, Vahdat L, Kinne D,
et al (2001) Randomized trial of black cohosh for the treatment
of hot flashes among women with a history of breast cancer.
J Clin Oncol 19:2739–2745
30. Gizicky HV (1944) Arzneipflanzen in ihren Beziehungen zum
weiblichen Genitalsystem. Versuche an weissen ratten und
mausen mit cimicifuga racemosa. Z Ges Exp Med 113:635–644
31. Siess M, Seybold G (1960) Untersuchungen uber die Wirkung
von Pulsatilla pratensis, Cimicifuga racemosa und Aristolochia
clematitis auf den Ostrus infantiler und kastrierter weisser
mause. Azneimittelforschung 10:514–520
32. Jarry H, Harnischfeger G (1985) Studies on the endocrine ef-
fects of the contents of Cimicifuga racemosa. 1. Influence on
the serum concentration of pituitary hormones in ovariectom-
ized rats. Planta Med 51:46–49
33. Jarry H, Harnischfeger G, Duker E (1985) Studies on endo-
crine effects of the contents of Cimicifuga racemosa. 2. In vitro
binding of compounds to estrogen receptors. Planta Med
4:316–319
34. Einer Jensen N, Zhao J, Andersen KP, Kristoffersen K (1996)
Cimicifuga and Melbrosia lack oestrogenic effects in mice and
rats. Maturitas 25:149–153
35. Eagon CL, Elm MS, Teepe AG, Eagon PK (1997) Medicinal
botanicals: estrogenicity in rat uterus and liver. Proc Am Assoc
Cancer Res 38:293
36. Eagon PK, Tress NB, Ayer HA, Wiese JM, Henderson T,
Elm MS, et al (1999) Medicinal botanicals with hormonal
activity (abstract). Proc Am Assoc Cancer Res 40:161–162
37. Jarry H, Leonhardt S, Duls C, Popp M, Christoffel V,
Spengler B, et al (1999) Organ-specific effects of Cimicifuga
racemosa in brain and uterus (abstract). 23rd International
LOF-Symposium on Phyto-Oestrogens, University of Gent,
Belgium
38. Lohning A, Verspohl EJ, Winterhoff H (1999) Pharmacological
studies on the dopaminergic activity of cimicifuga racemosa
(abstract). 23rd International LOF-Symposium on Phyto-
Oestrogens, University of Gent, Belgium
39. Freudenstein J, Dasenbrock C, Nisslein T (2000) Lack of
promotion of estrogen dependent mammary gland tumors in
vivo by an isopropanolic black cohosh extract. Phytomedicine
7[Suppl II]:13
40. Nisslein T, Freudenstein J (2000) Effects of black cohosh on
urinary bone markers and femoral density in an ovx-rat model
(abstract). Osteoporosis Int 11[Suppl 2]:504
41. Liu Z, Yang Z, Zhu M, Huo J (2001) Estrogenicity of black
cohosh (Cimicifuga racemosa) and its effect on estrogen re-
ceptor level in human cancer MCF-7 cells. Wei Sheng Yan Jiu
30:77–80
42. Nesselhut T, Schellhase C, Dietrich R, Kuhn W (1993) Study
on the proliferative potential of phytopharmacological agents
with estrogen-like effect in breast cancer cells. Arch Gynecol
Obstet 254:817–818
240
43. Jarry H, Ludwing ML, Stephan A, Kuhn U, Christoffel CV,
Wuttke W (1996) Constituents of cimicifuga racemosa exert
direct effects on in-vitro ovarian steroid secretion (abstract).
2nd International Congress on Phytomedicine, Munich, Ger-
many
44. Harnischfeger G, Cillien N (1996) Influence of Cimicifuga
racemosa extract fractions on the proliferation of human car-
cinoma cells in vitro with regard to their estrogen receptor
sensitivity (abstract). 44th Annual Congress of Georg August
Universitat, Gottingen, Germany p 12
45. Eagon CL, Elm MS, Eagon PK (1996) Estrogenicity of tradi-
tional Chinese and Western herbal remedies (abstract). Proc
Am Assoc Cancer Res 37:284
46. Zava DT, Dollbaum CM, Blen M (1998) Estrogen and prog-
estin bioactivity of foods, herbs, and spices. Proc Soc Exp Biol
Med 217:369–378
47. Lohning A, Verspohl EJ, Winterhoff H (2000) Cimicifuga
racemosa: in vitro findings using MCF-7 cells (abstract). Phy-
topharmakaforschung, Bonn
48. Dixon Shanies D, Shaikh N (1999) Growth inhibition of hu-
man breast cancer cells by herbs and phytoestrogens. Oncol
Rep 6:1383–1387
49. Freudenstein J, Bodinet C (1999) Influence of an isopropanolic
aqueous extract of Cimicifuga racemosa rhizoma on the pro-
liferation of MCF-7 cells (abstract). 23rd Internationl
LOF-Symposium on Phyto-Oestrogens, University of Gent,
Belgium
50. Liu J, Burdette JE, Xu H, Gu C, van Breemen RB, Bhat KP,
et al (2001) Evaluation of estrogenic activity of plant extracts
for the potential treatment of menopausal symptoms. J Agric
Food Chem 49:2472–2479
51. Johnson DE, Ochieng J, Evans SL (1995) The growth inhibi-
tory properties of a dopamine agonist (SKF 38393) on MCF-7
cells. Anticancer Drugs 6:471–474
52. Hagels H, Baumert-Krauss J, Freudenstein J (2000) Compo-
sition of phenolic constituents in Cimicifuga racemosa (ab-
stract). International Congress and 48th Annual Meeting of the
Society for Medicinal Plant Research. 6th International Con-
gress on Ethnopharmacology of the International Society for
Ethnopharmacology, Zurich, Switzerland
53. Struck D, Tegtmeier M, Harnischfeger G (1997) Flavones in
extracts of Cimicifuga racemosa. Planta Med 63:289
54. Tsai MJ, O’Malley BW (1994) Molecular mechanisms of action
of steroid/thyroid receptor superfamily members. Annu Rev
Biochem 63:451–486
55. O’Malley BW, Schrader WT, Mani S, Smith C, Weigel NL,
Conneely OM, et al (1995) An alternative ligand-independent
pathway for activation of steroid receptors. Recent Prog Horm
Res 50:333–347
241
... Gatunek Cimicifuga racemosa (L.) Nutt. -pluskwica groniasta (Ranunculaceae) -oficjalnie wprowadziła do praktyki medycznej w 1828 roku Turner Physiomedical School, która zalecała stosowanie korzeni i kłączy pluskwicy groniastej jako środka obniżającego ciśnienie, przeciwzapalnego i uspokajającego (1). Badania nad surowcem zapoczątkowano w Niemczech w latach 50. ...
... Ocena skuteczności stosowania wyciągów z korzenia i kłącza pluskwicy groniastej była przedmiotem licznych badań klinicznych (1,2,(32)(33)(34)(35) , przedstawili ocenę skuteczności i bezpieczeństwa stosowania przetworów z Actea jako pojedynczych oraz w terapii łączonej z przetworami innych leczniczych surowców roślinnych, np. zielem dziurawca, nasionami lnu itd., również wskazując na rozbieżności pomiędzy publikowanymi w tym okresie wynikami. ...
... Wiele badań klinicznych produktu leczniczego Remifemin wykazało, że zmniejsza on objawy naczynioruchowe i wahania nastroju występujące w okresie menopauzy (1,2,(32)(33)(34). Monitoring po wprowadzeniu produktu do obrotu dostarczył pozytywnych danych o ograniczeniu występowania uderzeń gorąca i nadmiernej potliwości. ...
Article
Full-text available
Black cohosh (Cimicifuga racemosa L., Ranunculaceae) root and rhizome are recommended in the treatment of vasomotor symptoms related to menopausal period, constituting a plant alternative to hormone replacement therapy (HRT). Although, its mechanism of action has not been fully understood, black cohosh is a rich and complex source of secondary metabolites with varied pharmacological activity aimed at alleviating menopausal symptoms. The efficiency of its use was confirmed in clinical trials. At the same time, C. racemosa root and rhizome are characterized by the content of numerous alkaloid compounds of various structural groups, which can be responsible for toxic effects. Despite that fact, there are no regulations indicating the need of standardization of black cohosh root and rhizome extracts, assuming the repeatability of their chemical composition. The large number of dietary supplements containing black cohosh root and rhizome extracts, compared to herbal medicinal products, may pose a health risk due to the lack of standardization and risk of adulteration with other Cimicifuga species. The paper presents the C. racemosa root and rhizome in terms of its traditional use, pharmacological and pharmacognostic studies, and clinical trials. In addition, herbal products of various categories containing the herbal substance and preparations obtained from it were characterized.
... It has been used for centuries by Europeans for the treatment of menopausal symptoms. (Borrelli and Ernst, 2002;Wuttke et al., 2003). ...
Chapter
Full-text available
ZYAZICI ve Doç. Dr. Esra UÇAR'ın yapacağı Ziraat, Orman ve Su Ürünü alanlarında özgün çalışmalar içeren "DIFFERENT PERSPECTIVES IN MEDICINAL AND AROMATIC PLANTS" adlı kitapta yayınlanmaya uygun görülmüştür. Uluslararası olarak yayınlanacak olan kitap çalışmamızda bölümünüzün yer almasını düşünmekteyiz. Çalışmalarınızda başarılar dileriz.
... Actaea racemosa has a long and diverse history of medicinal usage. From very early times, Native Americans used A. racemosa for the treatment of disorders specific to women, such as amenorrhoea and menopause, and pain during menses and childbirth (Foster, 1999;Borrelli and Ernst, 2002). A standardised extract of the plant known as 'Remifemin' has been used in Germany since the mid-1950s for the treatment of menopause (Murray, 1997;LowDog et al., 2003), and is recognised by the World Health Organisation (WHO) as effective for relieving menopausal symptoms (Guo et al., 2017). ...
Article
Introduction: The genus Actaea L., comprising of 32 species, has recently emerged as a source of potential phytochemicals with promising pharmacological properties. However, there is no review available integrating the scattered scientific studies on this genus. To fill this knowledge gap, an extensive review of the genus Actaea is presented with focus on its ethnomedical uses, phytochemistry, and pharmacology. Methods: For this review, 162 publications including 152 research papers, 7 books, 2 dictionaries and one glossary published from 1896 to April, 2020 A.D. were critically reviewed and relevant data extracted. The chemical structures and formulae of compounds have been sourced and validated using PubChem database; and drawn using Chem Draw Ultra 6.0. The scientific nomenclature follows The Plant List. Results & Discussion: For over more than a century, the ethnomedical uses of 8 species of this genus in treating 29 types of human diseases under different traditional medical systems have been reported. Over the last two decades, the phytochemical studies so far conducted on 16 species of Actaea have led to the isolation of more than 400 different compounds, notably cycloartane triterpenoids, flavonoids, phenylpropanoids, and nitrogenous compounds. Based on these phytochemical studies on Actaea species, the recent resurgence on pharmacological research conducted on 16 species has led to novel leads with potential applications. Conclusions: We highlight the current knowledge gaps and discuss future research prospects on genus Actaea, which can guide its bio-prospection and sustainable use in near future.
... This plant has also been used for the treatment of kidney disorders, rheumatism and malaria, but also general malaise [23,24]. At the beginning of the twentieth century, Actaea racemosa L. was also introduced as a phytomedicine in Europe to counteract the symptoms related to premenstrual syndrome, dysmenorrhea, and menopause [25][26][27][28]. A recent clinical study also supports the safety and efficacy of Actaea racemosa L. [29,30]. ...
Article
Full-text available
Background: Parkinson’s disease (PD) is a dopaminergic neuron-related neurodegenerative illness. Treatments exist that alleviate symptoms but have a variety of negative effects. Recent research has revealed that oxidative stress, along with neuroinflammation, is a major factor in the course of this disease. Therefore, the aim of our study was to observe for the first time the effects of a natural compound such as Actaea racemosa L. rhizome in an in vivo model of PD induced by neurotoxin 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP). Methods: for the study, mice received four injections of MPTP (20 mg/kg) for the induction of PD. Starting 24 h after the first administration of MPTP we treated mice with Actaea racemosa L. rhizome (100 mg/kg) daily for seven days. Results: our findings clearly demonstrated that Actaea racemosa L. rhizome treatment decreases oxidative stress by activating redox balance enzymes such as Nrf2/HO-1. We also demonstrated that Actaea racemosa L. rhizome is capable of modulating inflammatory indicators involved in PD, such as IκB-α, NF-κB, GFAP and Iba1, thus reducing the degeneration of dopaminergic neurons and motor and non-motor alterations. Conclusions: to summarize, Actaea racemosa L. rhizome, which is subject to fewer regulations than traditional medications, could be used as a dietary supplement to improve patients’ brain health and could be a promising nutraceutical choice to slow the course and symptoms of PD.
Article
Full-text available
Endometriosis is a chronic disease characterized by pelvic inflammation. This study aimed at investigating the molecular mechanisms underlying the pathology and how they can be modulated by the administration of a natural compound, Actaea racemosa (AR). We employed an in vivo model of endometriosis in which rats were intraperitoneally injected with uterine fragments from donor animals. During the experiment, rats were monitored by abdominal high-frequency ultrasound analysis. AR was able to reduce the lesion's size and histological morphology. From a molecular point of view, AR reduced hyperproliferation, as shown by Ki-67 and PCNA expression and MAPK phosphorylation. The impaired apoptosis pathway was also restored, as shown by the TUNEL assay and RT-PCR for Bax, Bcl-2, and Caspase levels. AR also has important antioxidant (reduced Nox expression, restored SOD activity and GSH levels, and reduced MPO activity and MDA levels) and anti-inflammatory (reduced cytokine levels) properties. Moreover, AR demonstrated its ability to reduce the pain-like behaviors associated with the pathology, the neuro-sensitizing mediators (c-FOS and NGF) expression, and the related central astrogliosis (GFAP expression in the spinal cord, brain cortex, and hippocampus). Overall, our data showed that AR was able to manage several pathways involved in endometriosis suppression.
Article
Cancer is a disease in which repeated rounds of mutations cause uncontrolled growth of cells, which prospers at the expense of their neighbor cells and then eventually leads to the destruction of the whole cellular community. Chemopreventive drugs either prevent DNA damage, which results in malignancy, or they stop or reverse the division of premalignant cells with DNA damage, which inhibits the growth of cancer. There is an obvious need for an alternate strategy given the ongoing rise in cancer incidence, the ineffectiveness of traditional chemotherapies to control cancer, and the excessive toxicity of chemotherapies. From antiquity to date, the saga of the usage of plants as medicine has been the mainstay among people worldwide. In recent years, extensive studies have been conducted on medicinal plants, spices, and nutraceuticals, as these have gained much popularity in reducing the risk of several cancer types in humans. Extensive studies on cell culture systems and animal models have demonstrated that various medicinal plants and nutraceuticals from various natural resources and their products, such as major polyphenolic constituents, flavones, flavonoids, antioxidants, etc, provide considerable protection against many cancer types. As shown in the literatures, the major aim of studies conducted is to develop preventive/therapeutic agents which can induce apoptosis in cancer cells without affecting normal cells. Projects are going on worldwide to find better ways to eradicate the disease. The study of phytomedicines has shed new light on this topic as research to date has proven that they have antiproliferative and apoptotic capabilities that will aid in the development of novel cancer prevention options. Dietary substances, such as baicalein, fisetin, and biochanin A have shown that they have an inhibitory effect on cancer cells, suggesting that they may work as chemopreventive agents. This review discusses the chemopreventive and anticancer mechanisms of such reported natural compounds.
Article
Full-text available
Women’s health is an imminent concern worldwide, but it remains an ignored segment of research in most developing countries, and is yet to take the center stage in even developed nations. Some exclusive female health concerns revolve around both pathological and physiological aspects. These gender-specific maladies include breast, cervical, and ovarian cancers, and physiological concerns such as menopause and osteoporosis, which are often coexistent. Recently, women’s health issues, including postmenopausal syndrome, have attracted the attention of researchers and practitioners alike, opening newer pharmaceutical research and clinical avenues. Although not counted as a disease, postmenopausal syndrome (PMS) is a female health phenomenon underpinned by hormonal depletion. Enhanced life expectancy in women has added to their suffering, and pharmacological interventions are needed. Amongst the available treatment modalities, the use of numerous botanicals has emerged as an efficient health management tool for women. Cimicifuga racemosa (CR or Black Cohosh) is a plant/herb which has been traditionally exploited and extensively used by women. This review is an attempt to compile and provide a summary of the importance of CR in complementary and alternative therapies for the improvement of various disorders related to women, such as menopausal syndrome, mammary cancer, and osteoporosis. It aims to systematically highlight the bioactive constituents, pharmacology, pharmacokinetics, therapeutic potentials, quality control processes, chromatographic techniques, and possible mechanisms of action of clinically effective phytomedicine for women’s health. Various clinical trials and patents relating to CR and women’s health have been collated. Furthermore, the plant and its related products have been considered from a regulatory perspective to reveal its commercial feasibility. The present review summarizes the existing data on CR focusing on women’s health, which can help to introduce this traditional phytomedicine to the world and provide some reference for future drug development.
Chapter
Full-text available
Actaea racemosa (AR) also known as Cimicifuga racemosa, is a perennial plant from Ranunculaceae family which was used as traditional remedies in treatment of various condition like rheumatoid muscular pain, headache, inflammation and dysmenorrhea. Actaea racemosa was basically native to Canada and the Eastern United State. This chapter proposed the ethnopharmacological uses of Actaea racemosa, and its phytochemical properties. Specifically, in this article we focused on use of Actaea racemose for menopausal and post-menopausal symptoms management. Electronic databases including PubMed and Scopus were searched for studies on Actaea racemose and its administration in management of menopausal symptoms. Chem Office software was also used in order to find chemical structures. The key words used as search terms were Cimicifuga racemose, Actaea racemose, Ranunculaceae, Black cohosh, Menopausal symptoms. We have included all relevant animal and human studies up to the date of publication. The analysis on Actaea racemose showed various indications for different plant’s extracts. Approximately 131 chemical compounds have been isolated and identified from Actaea racemosa. According to recently studies, the most important chemicals known of the Actaea racemosa are phenolic compounds, chromones, triterpenoids, nitrogen-containing constituents. In addition, in vivo and in vitro studies reported wide range of pharmacological activities for Black cohosh like attenuating menopausal symptoms. Mechanism of action for some ethnomedicinal indications were made clear while some of its activities are not confirmed by pharmacological studies yet. Further investigations on its pharmacological properties are necessary to expand its clinical effective use. Also, additional large clinical trials are recommended for clarifying the effect of Black cohosh.
Chapter
Plants contain a diverse variety of chemical compounds that play a crucial role in maintaining good health. Different plant parts are used in Ayurvedic or traditional Indian medicine practices to treat different ailments and diseased conditions. A group of food supplements, known as nutraceuticals, are also based on the components present in these plant parts to help provide immunity and help to regulate the metabolism for a healthy living. However, it has come to light that these plant parts used in medicine and as nutrition supplements can lead to severe toxicity in our body, thus triggering severe health problems. Therefore it is essential to understand which plant species should not be used in day-to-day life and what are its toxic constituents. This chapter highlights different groups of plants and their respective parts that should be avoided to use in any form or in meager amounts to restricting the concentration of its toxic constituents to a minimal.
Article
Black cohosh, Cimicifuga racemosa, is a medicinal herb which originally belonged to the medicinal treasures of the North American Indians. The herb was introduced into modern phytotherapy via homoeopathy. The plant is distributed around the northern hemisphere, only some controlled cultivation is done in the People's Republic of China. For medicinal preparations extracts of the dried rhizome are used. Quality is controlled by determination of the total content of triterpenglycosides. As active ingredients the two triterpenglycosides actein and cimifugiosid and further the flavonoid formononetin have to be mentioned. Extracts of the rhizomes exhibit hormonal properties, especially estrogenic effects. Formononetin is a competitive ligand in the estrogen-receptor assay and it binds to receptors in the uteri of ovariectomized rats. Further pharmacological properties reported from animal experiments are hypoglycemic, hypotensive and anti-inflammatory activities. Extracts of C. racemosa do not possess toxic or mutagenic properties. The clinical efficacy could be demonstrated in open and placebo-controlled trials as well as in comparison to therapy with hormones by significant relief of neurovegetative symptoms. The herbal preparations were very well tolerated with only minor gastrointestinal side-effects.