Open menu Close menu Open Search Close search

AMERICAN DRUGS | ANATOMY | HEALTH TOPICS | HIV/AIDS GLOSSARY | DISEASES | HEALTH ARTICLES | GENOME | OCCUPATIONS

Humulus (Lupuli flos)


Spanish Simplified Chinese French German Russian Hindi Arabic Portuguese





Authorisation details
Latin name of the genus: Humulus
Latin name of herbal substance: Lupuli flos
Botanical name of plant: Humulus lupulus L.
English common name of herbal substance: Hop Strobile
Status: F: Final positive opinion adopted
Date added to the inventory: 23/11/2005
Date added to priority list: 23/11/2005
Outcome of European Assessment: Community herbal monograph
Additional Information:






Product Characteristics
COMMUNITY HERBAL MONOGRAPH ON HUMULUS LUPULUS L., FLOS
1. NAME OF THE MEDICINAL PRODUCT
To be specified for the individual finished product
2. QUALITATIVE AND QUANTITATIVE COMPOSITION 1 , 2
Well-established use
Traditional use
With regard to the registration application of
Article 16d(1) of Directive 2001/83/EC as
amended
i) Herbal substance
Humulus lupulus L., flos (hop strobile)
ii) Herbal preparations
A) Comminuted 3 herbal substance
B) Liquid extract (1:1) prepared with
ethanol/water 45% v/v
C) Liquid extract (1:10) prepared with sweet
wine
D) Tincture (1:5) prepared with
ethanol/water 60% v/v
3. PHARMACEUTICAL FORM
Well-established use
Traditional use
Herbal substance or herbal preparation in solid or
liquid dosage forms for oral use.
The pharmaceutical form should be described by
the European Pharmacopoeia full standard term.
1 The material complies with the Eur. Ph. monograph (ref. no : 01/2005:1222).
2 The declaration of the active substance(s) for an individual finished product should be in accordance with relevant herbal
quality guidance
3 Comminuted is understood to include powdered herbal substance.
© EMEA 2008
2/5
 
 
 
4. CLINICAL PARTICULARS
4.1. Therapeutic indications
Well-established use
Traditional use
Traditional herbal medicinal product for relief of
mild symptoms of mental stress and to aid sleep
The product is a traditional herbal medicinal
product for use in specified indications
exclusively based on long standing use.
4.2. Posology and method of administration
Well-established use
Traditional use
Posology
Adolescents over 12 years of age, adults, elderly
Single dose
A) 0.5-1.0 g dried inflorescences (e.g. as
comminuted herbal substance)
0.5-2.0 g dried inflorescences for preparation
of an infusion
B) 0.5-2.0 ml of liquid extract (1:1), ethanol
45% v/v
C) Liquid extract (1:10), sweet wine
corresponding to 1.25 g herbal substance.
D) 2.0-4.0 ml of tincture (1:5), ethanol/water
60% v/v
For relief of mild symptoms of mental stress, a
single dose up to 4 times daily.
To aid sleep, 1 to 2 single doses half to one hour
before bedtime with an earlier dose during the
evening, if necessary.
The use is not recommended in children under
12 years of age (see section 4.4. Special warnings
and precautions for use).
Duration of use
If the symptoms persist during the use of the
medicinal product, a doctor or a qualified health
care practitioner should be should consulted.
Method of administration
Oral use.
© EMEA 2008
3/5
4.3. Contraindications
Well-established use
Traditional use
Hypersensitivity to the active substance.
4.4. Special warnings and precautions for use
Well-established use
Traditional use
The use is not recommended in children under
12 years of age due to lack of adequate data.
For tinctures and extracts containing ethanol the
appropriate labelling for ethanol, taken from the
‘Guideline on excipients in the label and package
leaflet of medicinal products for human use’, must
be included.
4.5. Interactions with other medicinal products and other forms of interaction
Well-established use
Traditional use
None reported.
4.6. Pregnancy and lactation
Well-established use
Traditional use
Safety during pregnancy and lactation has not
been established.
In the absence of sufficient data, the use during
pregnancy and lactation is not recommended.
4.7. Effects on the ability to drive and use machines
Well-established use
Traditional use
May impair ability to drive and use machines.
Affected patients should not drive or operate
machinery.
4.8. Undesirable effects
Well-established use
Traditional use
None known.
If adverse reactions occur, a doctor or a qualified
health care practitioner should be consulted.
© EMEA 2008
4/5
4.9. Overdose
Well-established use
Traditional use
No case of overdose has been reported.
4. PHARMACOLOGICAL PROPERTIES
5.1. Pharmacodynamic properties
Well-established use
Traditional use
Not required as per Article 16(c)(1)(a)(iii) of
Directive 2001/83/EC as amended.
5.2. Pharmacokinetic properties
Well-established use
Traditional use
Not required as per Article 16c(1)(a)(iii) of
Directive 2001/83/EC as amended.
5.3. Preclinical safety data
Well-established use
Traditional use
Not required as per Article 16c(1)(a)(iii) of
Directive 2001/83/EC as amended, unless
necessary for the safe use of the product.
Adequate tests on genotoxicity were not
performed.
Tests on reproductive toxicity and carcinogenicity
have not been performed.
6. PHARMACEUTICAL PARTICULARS
Well-established use
Traditional use
Not applicable.
7. DATA OF COMPILATION/LAST REVISION
11 July 2008
© EMEA 2008
5/5


Assessment Report
ASSESSMENT REPORT
FOR HERBAL SUBSTANCE(S), HERBAL PREPARATION(S) OR COMBINATIONS
THEREOF WITH TRADITIONAL USE
HUMULUS LUPULUS L., FLOS
BASED ON ARTICLE 16D(1) AND ARTICLE 16F AND 16H OF DIRECTIVE 2001/83/EC AS
AMENDED (TRADITIONAL USE)
Herbal substance(s) (binomial scientific name
of the plant, including plant part)
Humulus lupulus L., flos
Herbal preparation(s)
Comminuted herbal substance
Liquid extracts and tincture
Pharmaceutical forms
Solid or liquid dosage forms for oral use
Rapporteur
Prof. A. J. Vlietinck
© EMEA 2008
2/30
TABLE OF CONTENTS
I.
REGULATORY STATUS OVERVIEW
2
II.
ASSESSMENT REPORT FOR HERBAL SUBSTANCE/HERBAL
PREPARATIONS WITH TRADITIONAL USE
3
II.1.
INTRODUCTION
5
II.1.1.
Description of the herbal substance(s), herbal preparations or
combination thereof
5
II.1.2.
Information on period of medicinal use in the Community regarding the
specified indication
5
II.1.2.1.
Herbal substance
6
II.1.2.2.
Herbal preparations
6
II.2.
PHARMACOLOGY
8
II.2.1.
Pharmacokinetics
8
II.2.1.1.
Phytochemical characterization
8
II.2.1.2.
Pharmacokinetic data on the herbal substance
9
II.2.1.3.
Pharmacokinetic data on the active compound, 8-PN
11
II.2.2.
Pharmacodynamics
12
II.2.2.1.
Sedative effects
12
II.2.2.1.1. In vitro studies
12
II.2.2.1.2. In vivo studies
12
II.2.2.2.
Oestrogenic effects
14
II.2.2.2.1. In vitro studies
14
II.2.2.2.2. In vivo studies
17
II.2.2.3.
Other pharmacological activities of hop strobiles and/or its constituents
20
II.2.2.3.1. Hop essential oil and hop acids
20
II.2.2.3.2. Hop prenylflavonoids
22
II.2.3.
Interactions
23
II.3.
CLINICAL EFFICACY
23
II.3.1.
Clinical studies
23
II.3.1.1.
Sedative activity
23
II.3.1.2.
Oestrogenic activity
24
II.4.
SAFETY
25
II.4.1.
Toxicity
25
II.4.1.1.
Single dose toxicity
25
II.4.1.2.
Subacute and chronic toxicity
26
II.4.1.3.
Genotoxicity
27
II.4.1.4.
Carcinogenicity
28
II.4.1.5.
Reproductive and developmental toxicity
28
II.4.1.6.
Local tolerance
28
II.4.2.
Side effects
28
II.4.3.
Contra-indications, warnings
29
II.4.4.
Interactions
29
II.4.5.
Overdoses
29
II.5.
ASSESSOR’S OVERALL CONCLUSION
30
III.
ANNEXES
III.1.
Community herbal monograph
III.2.
Literature references
© EMEA 2008
3/30
I.
REGULATORY STATUS OVERVIEW 1
MA: Marketing Authorisation;
TRAD: Traditional Use Registration;
Other TRAD: Other national Traditional systems of registration;
Other: If known, it should be specified or otherwise add ’Not Known’
Member State
Regulatory Status
Comments
Austria
MA
TRAD
Other TRAD
Other Specify:
Belgium
MA
TRAD
Other TRAD
Other Specify: Combi + foodsuppl
Bulgaria
MA
TRAD
Other TRAD
Other Specify:
Cyprus
MA
TRAD
Other TRAD
Other Specify:
Czech Republic
MA
TRAD
Other TRAD
Other Specify: Combi
Denmark
MA
TRAD
Other TRAD
Other Specify: Combi
Estonia
MA
TRAD
Other TRAD
Other Specify:
Finland
MA
TRAD
Other TRAD
Other Specify: Combi
France
MA
TRAD
Other TRAD
Other Specify:
Germany
MA
TRAD
Other TRAD
Other Specify: Combi
Greece
MA
TRAD
Other TRAD
Other Specify:
Hungary
MA
TRAD
Other TRAD
Other Specify:
Iceland
MA
TRAD
Other TRAD
Other Specify:
Ireland
MA
TRAD
Other TRAD
Other Specify:
Italy
MA
TRAD
Other TRAD
Other Specify: Food suppl.
Latvia
MA
TRAD
Other TRAD
Other Specify:
Liechtenstein
MA
TRAD
Other TRAD
Other Specify:
Lithuania
MA
TRAD
Other TRAD
Other Specify:
Luxemburg
MA
TRAD
Other TRAD
Other Specify:
Malta
MA
TRAD
Other TRAD
Other Specify: Combi + foodsuppl.
The Netherlands
MA
TRAD
Other TRAD
Other Specify:
Norway
MA
TRAD
Other TRAD
Other Specify: Foodsuppl.
Poland
MA
TRAD
Other TRAD
Other Specify:
Portugal
MA
TRAD
Other TRAD
Other Specify: None
Romania
MA
TRAD
Other TRAD
Other Specify:
Slovak Republic
MA
TRAD
Other TRAD
Other Specify:
Slovenia
MA
TRAD
Other TRAD
Other Specify:
Spain
MA
TRAD
Other TRAD
Other Specify:
Sweden
MA
TRAD
Other TRAD
Other Specify: Combi
United Kingdom
MA
TRAD
Other TRAD
Other Specify: Combi
1 This regulatory overview is not legally binding and does not necessarily reflect the legal status of the products
in the MSs concerned.
© EMEA 2008
4/30
 
II
ASSESSMENT REPORT FOR HERBAL SUBSTANCE/HERBAL
PREPARATIONS WITH TRADITIONAL USE
II.1.
INTRODUCTION
II.1.1.
Description of the herbal substance(s), herbal preparations or combinations
thereof
Humulus lupulus L., flos (Strobili), (Hops, Hop strobiles)
* Herbal substance
Botanical species
Humulus lupulus L.
Hops, Houblon (Vigne du Nord), Hopfen, Hop, Luppulo, Lupolo, Lupulo
Botanical family
Cannabaceae
Plant part
Flowers (Strobiles) : dried, generally whole, female inflorescences
(Eur. Ph. 01/2005 : 1222)
Common names
(plant part)
- Flores Humuli lupuli, Strobili humuli, Strobili lupuli, Strobuli lupuli
(Latin)
- Hops, Hop strobiles, (English)
- Cônes de Houblon (French)
- Hopfenblüten, Hopfendolden, Hopfenkätzchen, Humulus-lupulus-
Blütenstände (German)
- Hop (Dutch)
Pharmaceutical forms
Dried inflorescences (strobiles)
Glandulae lupuli (Lupulinum) : Hop grains, Lupulin glands, (English) ;
Lupulin (French), Lupulin, Hopfendrüsen, Hopfenmehl (German) ;
Hopklierharen (Dutch)
Other herbal substances
These globular grains serve as rich reservoirs of secondary metabolites
and they are contained in hop cones that are typical of female plants
(up to 40% of the dry mass).
* Herbal preparations
♦ Dried inflorescences (strobiles) : comminuted herbal substance as such or as infusions
♦ Liquid extract (1:1) : prepared with ethanol/water 45% v/v
♦ Liquid extract (1:10) prepared with sweet wine
♦ Tincture : (1:5) prepared with ethanol/water 60% v/v
II.1.2.
Information on period of medicinal use in the Community regarding the specified
In preparing this report, a number of data sources have been taken into account viz . the ESCOP
monographs published respectively in 1997 and 2003, the bibliographic references made available by
ESCOP at the end of 1997, the monograph of the European Pharmacopoeia in 2005 and the results of
several literature searches carried out in 2007.
© EMEA 2008
5/30
indication
II.1.2.1. Herbal substance
This assessment report reviews the available scientific data, particularly the pharmacological and
clinical studies available for Humulus lupulus L., strobiles (Cannabaceae) or hops.
The dried, generally whole female inflorescences of hops ( Lupuli flores ) are described in the European
Pharmacopoeia (Lupuli flos 01/2005:1222) (2005). They are also described in an ESCOP monograph
in 2003 with a therapeutic indication similar to valerian root viz . tenseness, restlessness and sleep
disorders. Hops are referred to in the herbal literature as having various forms of sedative activity
(Chadwick et al., 2006 and references cited therein namely Fluckiger and Hanbury, 1879, Maisch,
1892, Schleif and Galludet, 1907, Greenish, 1909, Wilcox, 1912, Culbreth, 1927, Washburn and
Blome, 1927, Gathercoal and Wirth, 1936, Youngken, 1950, Meyer, 1960, Millspaugh, 1974,
Blumenthal, 2000). They are often combined with more potent sedative herbs such as valerian, passion
flower and lemon balm for the treatment of sleep disturbances (Blumenthal 2000, Schultz et al., 2001).
More than 99% of the world production of hops is intended for breweries, however, since 1973 it has
been claimed that brewing sludge baths containing ca. 30% hop extracts could be used for the
treatment of a variety of gynaecological disorders (Fenselau and Talahay, 1973). References to the
traditional use of hops in the treatment of such disorders have been identified from the US (Donsback,
1977), Romania (Racz et al., 1980), France (Goetz, 1990) and Iran (Zagari, 1992). Additionally, a
number of journal articles (Bednar and Zenisek, 1961, Strenkovskaya, 1968, Fenselau and Talahay,
1973) and patents exist concerning the use of oestrogenic properties of hops, particularly for external
use in cosmetics. Koch and Heim (1953), following up on the folk legend “that women who normally
live a distance from hop gardens regularly begin to menstruate 2 days after arriving to pick hops”,
reported that hops contain the equivalent of 20-300 µg oestradiol/g. While lacking in detail, this one-
page article is apparently the first confirmation that hops may have oestrogenic activity in humans.
Koch and Heim used a version of the Doisy test, an in vivo assay with castrated female infant rodents
(Hensyl, 1990). Chury (1961) reported that the oestrogenic activity for a saponified ethanol extract of
hops was much more active than those of peas, red clover, and cabbage. Several hop-containing
cosmetic preparations have been patented since that time.
It was not until 1988 that the chemical structure of the oestrogenic principle was established (Hänsel
and Schulz, 1988). The report by Milligan et al. (1999), however, may be regarded as the beginning of
the modern, unambiguous understanding of the in vitro oestrogenic activity of hops. They reported
that hops contain a very potent phyto-oestrogen, 8-prenylnaringenin (8-PN), also called hopein, which
belongs to the class of the prenylflavonoids. This finding has been confirmed by a number of
independent research groups, both by in vivo and in vitro studies (Coldham and Sauer, 2001, Zierau et
al., 2002, Takamura-Enya et al., 2003). The high oestrogenic activity of 8-PN was reported for the
first time in 1998, when the compound was isolated from the heartwood of the tropical tree
Anaxagerea benzonensis A. Gray (Annonaceae) (Kitaoka et al., 1998). 8-PN has also been detected in
some beers, in relatively small quantities, however (levels between 20 µg – 100 µg/L) (Rong et al.,
2000, Schaefer et al., 2003). Since then several review articles on the oestrogenic properties of hops in
comparison to other phyto-oestrogens have been published (Cos et al., 2003, Ososki and Kenelly,
2003).
Hop extracts and/or compounds have also been reported to be active as antioxidants, cancer
chemopreventives, antiinflammatory agents, antimicrobials (antibacterials and antifungals), and
cytotoxics (Chadwick et al., 2006). These indications, however, are substantiated mainly by
pharmacological data and not by clinical studies.
II.1.2.2. Herbal preparations
The dried inflorescences are used in comminuted form as such or in tea mixtures or they are prepared
as infusions.
Lipophilic extracts are used for the preparation of bath oils whereas hydroalcoholic liquid extracts and
tinctures are prepared for internal use as sedatives, mostly in combinations with other sedative plant
extracts. In several cases, the hydroalcoholic liquid extracts are prepared with sweet wine.
Hop strobiles are described in the DAB10, PFX and the BHP. In the latter (1983) a liquid extract (1:1)
in 45% alcohol and a tincture (1:5) in 60% alcohol are given together with their posology. The
© EMEA 2008
6/30
German E Commission describes a monograph on hop strobiles and hop extracts as sedative in 1984
(BAnz nr. 228, dated 05.12.1984).
In Germany marketing authorisations were granted for HMPs containing:
Fluid extract (1:12.4-12.6) : ethanol 16% m/m (since 1965)
Daily dosage corresponding to 0.8 g herbal substance (HS) (in divided single dosages, 2-3 x daily)
Fluid extract (1:94-95), sweet wine (since 1978)
Daily dosage corresponding to 0.4-0.6 g HS (in divided single dosages, 2-3 x daily)
Fluid extract (1:10), sweet wine (since 1978)
Daily dosage corresponding to approximately 5 g HS (in divided single dosages, 2-3 x daily)
Dry extract (4-5:1), methanol 50% v/v (since 1993)
Daily dosage corresponding to 1-1.7 g HS (in divided single dosages, 2-3 x daily)
In Germany, the herbal substance is part of many combination products with Valerian, Passion flower
and Melissa in solid and liquid pharmaceutical forms.
Consequently, only hydroalcoholic liquid extracts among them wine and tinctures can be accepted as
traditional drugs.
Assessor’s comments
References concerning the traditional medicinal use of hops as a sedative herb date back to 1879
(Fluckiger and Hanbury). Since then many publications up to the present day have appeared in
which hop strobiles as comminuted herbal substance, alone or in tea mixtures or as infusions
continue to be recommended by proponents of herbal medicine for this indication. Unfortunately,
despite numerous attempts the constituents responsible for the sedative effects of hops have yet to
be established, although some information about the biochemical mode of action of hops as
sedative herb is now available.
Besides the dried inflorescences only liquid extracts prepared with ethanol/water (16% and 45%) and
sweet wine, and tinctures (1:5) prepared with ethanol/water 60% have been used in medicinal products
for more than 30 years. Although the fluid extracts (1:12.4-12.6) and (1:94-95), prepared with ethanol
16% m/m and sweet wine, respectively, have been marketed in Germany since 1978, their daily
dosages corresponding respectively with 0.8 g and 0.4-0.6 g herbal substance are not plausible with
the posology of the other herbal preparations. Consequently, these preparations should not be included
in the Community monographs on hops for traditional use.
Dry extracts have only been on the market since 1993 and are consequently not considered to be
traditional.
References concerning the traditional medicinal use of hops as a phyto-oestrogen date back to
Koch and Heim (1953), who confirmed the folk knowledge that the menstruation cycle of female
hop pickers is influenced by picking hops. In 1961 Chury reported the oestrogenic activity for a
saponified ethanolic extract of hops, where after several hop containing cosmetic preparations
have been patented. In 1973 Fenselau and Talahay stated that brewing baths containing hop
extracts (30%) were taken in Germany for the treatment of a variety of gynaecological disorders.
In 1988 the chemical structure of the active oestrogenic principle was elucidated and in 1999 with
Milligan the modern understanding of the in vitro and in vivo oestrogenic activity of hops began.
Since then, various dried extracts prepared with ethanolic-water mixtures have been standardised
on 8-prenyl-naringenin and sometimes other prenylflavonoids. Several pharmacological,
toxicological and clinical studies with these standardised extracts have been carried out to
investigate their usefulness as drugs to alleviate menopausal discomforts.
Although these studies are of interest, the findings are not yet sufficient to support a marketing
authorisation.
Moreover, as these extracts are enriched in 8-prenylnaringenin and prepared with modern techniques
such as supercritical CO 2 extraction, they cannot be considered to be traditional.
© EMEA 2008
7/30
Consequently, a period of at least 30 years of traditional use of hop strobiles and its preparations is
only fulfilled for their sedative effects. Since the use of dried extracts for this indication has only been
introduced since 1993, only hydroalcoholic liquid extracts including also sweet wine and dried
inflorescences as such or as infusions can be accepted as traditional herbal medicinal products.
II.2.
PHARMACOLOGY
II. 2.1.
Pharmacokinetics
II. 2.1.1. Phytochemical characterization
More than 1000 chemicals have been identified from hops including natural products and their
isomeric derivatives (Verzele and De Keukeleire, 1991; Eri et al., 2000; Farnsworth, 2003, Anonymus,
Beilstein Database, 2003).
The oleo-resin (3% - 20%) consists of various prenylated phoroglucinol derivatives called “bitter
acids”. They are classified either as “α-acids” or “β-acids”, which are distinguished by the fact that the
former ones are precipitated from a crude extract of hops by the addition of lead acetate. The β-acids,
by definition, would remain in solution. The α-acids are humulones (2-12% of dried strobile), e.g.
humulone, cohumulone, prehumulone, posthumulone and adhumulone, whereas the β-acids are called
lupulones (1-10% of dried strobile), e.g. lupulone, colupulone, adlupulone, prelupulone and
postlupulone. The α-acids are regarded as the most important constituents in determining the quality
of hops. They contribute to foam stability as well as imparting antibacterial properties. While regarded
as the principal “bitter acids” from hops, they perhaps paradoxically do not have a bitter taste, even at
concentrations of 100 µg/ml. The hop α-acids isomerize to the corresponding “iso-α-acids” under a
variety of reaction conditions e.g. more favourably at higher pH values. These bitter iso-α-acids are
artifacts and comprise more than 80% of all hop components that occur in typical beers. In the same
circumstances the β-acids are transformed into so-called hulupones (Verzele and De Keukeleire,
1991).
The essential oil (0.5-1.5%) consists mainly of simple oxidized alkanes, monoterpenes and
sesquiterpenes. The primary volatile constituents in all cultivars of hops are the monoterpene myrcene
and the sesquiterpenes β-caryophyllene and humulene (57-82% of the volatile oil) (Eri et al., 2000).
In terms of their traditional economic value, the volatile oil and bitter acids comprise the most
significant classes produced by hops. Candidate sedative compounds in hops have been put forward
viz . the bitter acids humulone and lupulone (Sikorski and Rusiecki, 1938), their degradation product
2’-methyl-3-buten-2-ol (Hänsel et al., 1982) and myrcene (Rao et al., 1990, Lanzotti et al., 1991).
The alcohol, however, is only present in trace amounts in freshly harvested hop strobiles, but it
increases of ± 0.15% of the dry weight up to 20% of the volatiles after two years due to degradation of
the bitter acids (Hänsel et al., 1982 ; Wohlfart et al., 1982a, 1983b ; Hölz, 1992).
Despite numerous attempts to characterize the sedative active constituents in hops, it is believed that
the identity of all compounds responsible for this activity of hops in humans, has yet to be determined
(Schulz et al., 2001).
A third class of compounds consists of flavonoids (0.5-1.5%) including quercetin and kaempferol
glycosides (McMurrough et al., 1981 ; De Cooman et al., 1998) and about 30 prenylated, geranylated,
oxidized and/or cyclized chalcones. The most abundant chalcones are xanthohumol (X) (up to 1% of
dried strobile and 80-90% of total flavonoids) and desmethylxanthohumol (DMX). These chalcones
are readily isomerized to the corresponding flavanones viz . respectively isoxanthohumol (IX) and a
mixture (ca. 3:2) of 6-prenylnaringenin (6-PN) and 8-prenylnaringenin (8-PN) (25-60 mg/kg) (Stevens
et al., 1997, 1999a, 1999b, 2004 ; Rong et al., 2000, Milligan et al., 2000).
The principle oestrogen from hops is the (1:1) racemate of 8-PN or hopein, formed along with
the less active 6-PN (Milligan et al., 1999).
IX is the main prenylated flavonoid in beer, since X is largely isomerized during particular stages
(at elevated temperatures) in the course of brewing (Stevens et al., 1999). These five prenylated
© EMEA 2008
8/30
flavonoids viz . X,DMX,IX, 6-PN, and 8-PN constitute a set of interesting bioactive compounds, since
they have shown their ability to inhibit proliferation of several cancer cell lines in vitro (Miranda et al.,
1999, De Keukeleire et al.,2001,Gerhauser et al., 2002).
Other constituents of hops include amino acids and proteins (15%), polysaccharides (50-60%),
minerals, phenolic acids such as chlorogenic and caffeic acids, lipids and condensed tannins (2-4%)
(Verzele and De Keukeleire et al., 1991, Hölz, 1992).
II. 2.1.2. Pharmacokinetic data on the herbal substance
So far no in vivo pharmacokinetic investigations were performed on hop extracts due not only to the
complexity of the phytochemical composition but also to the many metabolisations of the different
compounds of hops which take place after intake in humans.
Recently, however, several research teams have published their findings on the metabolism of
8-prenylnaringenin (8-PN), xanthohumol (X) and isoxanthohumol (IX). A first indication on the
ADME characteristics of these three compounds can be deduced from CaCo-2 experiments performed
by Bracke et al. (2006). They determined “the apparent permeability coefficient” (P app ) as an important
parameter for the absorption efficacy through the gut epithelium. In this in vitro system.
P app > 1x10 6 cm/s values are correlated with a good absorption, whereas values p app < 1x10 7 cm/s are
correlated with weak absorption (< 1%). For values in between, there is no real relationship between
P app and absorption, but the absorption is considered between > 1% and < 100% (Grès et al., 1998).
8-PN and IX showed P app values of respectively 1,3x10 -6 cm/s and 3,3x10 -6 cm/s, showing that these
compounds could have a good bioavailibity, whereas the P app of X (6,7x10 -7 cm/s) showed that it
might be less bioavailable. Beside the information on the absorption efficacy, the CaCo-2 system also
indicated that phase-II metabolites (glucuronidation and sulphation) are formed. A similar study has
been published by Nikolic et al. (2006). The apparent permeability coefficients were 5.2 ± 0.7 x 10 -5
and 4.9 ± 0.5 x 10 5 cm/s respectively, indicating a good absorption via passive diffusion. According to
these authors, the P app values are similar to those of drugs such as propanolol and testosterone, which
are often used as high permeability standards.
The metabolism of prenylated flavonoids is not very well documented. The biotransformation of
flavonoids is done at several places in the body, but mainly in the liver and the gut lumen, so that the
gut flora might play a significant role as well. As cytochrome P450-enzymes (CYP) are abundantly
present in the liver, they also are important elements in the metabolization process of flavonoids
(Yilmazer et al., 2001a).
Several studies showed that flavonoids are mostly excreted as glucuronides in humans and animals.
Glucuronidation is the most important reaction route for the phase II detoxification process for most of
the xenobiotics. The functionalization is catalysed by the membrane-bound
UDP-glucuronosyltransferase, mainly catalysed in the endoplasmatic reticulum of the liver. In an
in vitro study with rat-derived and human liver microsomes, the glucuronidation of X was studied.
Two important glucuronidates were characterized as respectively C-4’- and C-4-monoglucuronides of
X. It is also important to mention that the conjugated flavonoids still have a significant biological
activity (e.g. antioxidant effects) (Yilmazer et al., 2001b).
As for 8-PN, an in vitro study with human liver microsomes has revealed several metabolites. LS-MS
analysis showed that the most important breakdown products are the result of oxidation. In this
process, a hydroxyl group can appear on the prenyl chain, but also a modification of the flavanone
skeleton has been observed. The most frequent metabolites were oxidation products of the prenyl
group, in which the terminal methyl function was hydrolysed. This is in contrast to xanthohumol,
which did not show a similar metabolism (Nikolic et al., 2004 ; Zierau et al., 2004).
© EMEA 2008
9/30
Guo et al. (2006) performed a study on the identification of human hepatic cytochrome P450 enzymes
involved in the metabolism of 8-PN and IX from hops. CYP2C19 was found to catalyze the formation
of both cis- and trans-alcohols (phase I metabolisation) of the prenyl side chain of 8-PN. CYP2C8
converted 8-PN regioselectively to the trans-alcohol of the prenyl group. Finally, CYP1A2 was found
to catalyze the O-demethylation of IX to generate 8-PN. The enzymes CYP1A2, CYP2C19 and
CYP2C8 exhibit polymorphism in humans. The results obtained by Guo et al. suggest that the
oestrogenicity of hop constitutents in vivo will depend in part on metabolic conversion that may show
individual variation.
The possibility that IX would act as a pro-oestrogen was considered by Coldham et al. (2002).
The assumption was based on the extensive biogransformation capacity of the liver, which includes
demethylation. However, the exposure of IX to liver microsomes did not lead to an increase in
oestrogenic activity, from which it was concluded that no 8-PN was produced. In contrast,
Nikolic et al. (2005) describe that liver microsomes can demethylate IX, but not X. However, it was
shown that, besides demethylation, microsomes also modify the prenyl side-chain, finally resulting in
a large variation of minor degradation products. Schaefer et al. (2005) identified low levels of 8-PN in
urine after oral intake of IX by two test persons and attributed this to demethylation by the liver.
Besides the liver, the colon is also an important transformation site in the human body. The human
colon contains ∼ 10 12 micro-organisms/cm 3 (about 400 different species), with an enormous catalytic
and hydrolytic potential. The importance of this microbial community in the metabolism of phyto-
oestrogens in general has been clearly established. Wang et al. (2000) identified two bacteria
responsible for the transformation of lignans and Decroos et al. (2005) recently isolated a microbial
consortium capable of transforming the soy phyto-oestrogen daidzein into equol. Moreover, several
intestinal bacteria were shown to enhance the bioavailability of phyto-oestrogens as they possess
β-glucosidases, which are necessary for the hydrolysis of phyto-oestrogen glycosides
(Rowland et al., 2003). Thus, the gut microbiota are considered to be a factor of utmost importance for
phyto-oestrogen bioavailability (Turner et al., 2003). In view of the importance of the microbial
microflora observed with other well-known phyto-oestrogens, the group of Verstraete (University of
Ghent, Belgium) has launched a study to investigate the most important substances in the hops extract
(IsoX, 8-PN and X) in a SHIME reactor (SHIME or ‘ S imulator of the H uman I ntestinal Mi crobial
E cosystem’). Parts of the results have already been published by Possemiers et al. (2005). The results
obtained showed that certain bacteria in the gut are able to O-demethylate IX, resulting in 8-PN
formation. In an experiment with faecal cultures, this conversion was observed in one-third of the
samples, indicating the importance of interindividual variability in the intestinal microbial community.
These results stress the importance of the gut microbial community toward the final biological activity
of phyto-oestrogens, a factor that should not be neglected when determining the final active dose
inside the body (Possemiers et al., 2006).
© EMEA 2008
10/30
II. 2.1.3. Pharmacokinetic data on the active compound, 8-PN
The pharmacokinetic data which can be found in literature, are mostly on xanthohumol (X). They
show that the bioavailability of X is rather low (Nookandeh et al., 2004 ; Avula et al., 2004). X and its
metabolites are excreted mainly in faeces within 24 h of administration.
Schaefer investigated the pharmacokinetics of 8-PN in rats and dogs in his Ph.D. work (2004). The
full ADME profile and bioavailability profile (using synthetic 8-PN) that he proposes can be found in
the thesis. Briefly, they found that the ingested 8-PN is almost completely absorbed after ingestion.
Still, systemically circulating 8-PN concentrations were only 5% of the ingested amount, indicating a
strong action (presystemic elimination) before the 8-PN could go into the systemic circulation.
The 8-PN in the systemic circulation is also bound to serum proteins (sex hormone binding globulins
in dogs). This binding would be less likely for X, in view of its non-oestrogenic characteristics. Also
Nikolic et al. (2004, 2006) concluded that the bioavailability of 8-PN was reduced significantly by
intestinal and hepatic metabolism.
The comparatively high metabolic stability of 8-PN and its pronounced presystemic elimination via
the bile resulted in enterohepatic recirculation in both rats and dogs. The concentration profile in the
serum after oral uptake showed two maxima: one after 1 h and another (higher concentration) after
2 to 4 h, which is an indication of the enterohepatic concentration. About 50% of the ingested amount
was recovered unchanged in the faeces. The excretion mainly goes via the biliary route, as renal
excretion (unchanged and conjugated 8-PN) is limited, especially after oral administration.
The pharmacokinetic results obtained in rats and dogs were more or less confirmed in a human study
performed by Rad et al. (2006). The study was performed using a randomized, double-blind, placebo-
controlled, dose-escalation design with three groups of eight healthy postmenopausal women. In each
group six subjects received 8-PN and two subjects placebo. 8-PN was given orally in doses of 50, 250
or 750 mg. Drug concentrations in serum, urine and faeces were measured up to 48 h. Serum
concentrations of free 8-PN showed rapid drug absorption and secondary peaks suggestive of marked
enterohepatic recirculation. Independent of the treatment group, approximately 30% of the dose was
recovered in excreta as free compound or conjugates over the 48 h observation period. The first C max
and AUCO-48 h showed dose linearity with ratios of 1:4,5 : 13.6 (C max ) and 1:5,2 : 17.1 (AUC).
Assessor’s comments
The in vitro studies indicated extensive liver biotransformation of X, IX and 8-PN upon absorption.
However, the extent of dietary polyphenol absorption in the small intestine is rather limited (10-20%),
thereby implying that a large proportion reaches the colon. Naringenin, the non-prenylated analogue of
8-PN, showed intensive biotransformation in the intestine, including ring cleavage and
dehydroxylation, followed by absorption and urinary excretion. The extent of degradation strongly
depended on compound concentration and individual composition of the gut microflora of the
different human subjects. On the other hand, when X was fed to rats, it was mainly recovered in
unchanged form from the faeces (89%).
The finding that IX may be 0-demethylated by human hepatic cytochromes P450 to form the potent
phyto-oestrogen 8-PN is of considerable importance. In addition, under the acidic conditions of the
stomach X can be cyclized to form IX and provide yet another route for the formation of 8-PN. Based
on these results, IX and X should be included among the compounds used for standardisation of hop
extracts to be evaluated for oestrogenicity in vitro and in vivo.
The pharmacokinetic profile of 8-PN was studied in rats and dogs and later in human females. It was
shown that 8-PN was rapidly and almost completely absorbed after oral administration. The free
substance is then taken up in the jejunum, enters the portal circulation and goes to the liver. Thereafter,
besides unchanged 8-PN which goes into the systemic circulation, 8-PN undergoes mainly phase II
metabolisation and the hydrophilic conjugates formed are mainly eliminated in the bile.
Bacteria in the gut can deconjugate the phase II metabolites, and lipophilic substances can be taken up
again and go to the enterohepatic circulation. Free substance is mainly excreted via the faeces, but also
conjugates are excreted from the blood with urine.
© EMEA 2008
11/30
In conclusion, the pharmacokinetic profile of 8-PN is characterized by rapid and probably complete
enteral absorption, a pronounced presystemic elimination with subsequent enterohepatic recirculation,
dose linear pharmacokinetics and competively little phase I metabolism. Conjugation to
β-glucuronides or sulphates seems to constitute the main metabolic pathway. Intestinal and hepatic
phase I metabolism plays a minor role in the inactivation of 8-PN. This is in complete contrast to other
natural or synthetic oestrogens, whose decreased oral bioavailability is due to intestinal conjugation
and hepatic metabolic inactivation.
The studies also indicate that 8-PN is transformed into a wide array of metabolites, and some of these
products may have different pharmacological activity than their precursors. It would, therefore, be of
interest to determine the biological activities and potential toxic effects of these metabolites,
particularly since it is known that hydroxylated metabolites of 17-β-oestradiol (mainly the 16-OH
metabolite) are much more active, thereby increasing the risks for oestrogen-dependent cancers.
II. 2.2.
Pharmacodynamics
II. 2.2.1. Sedative effects
II.2.2.1.1. In vitro studies
Over the past decade considerable pharmacological research has been carried out on hop strobiles and
its constituents particularly with respect to oestrogenic activity. However, the publications on in vitro
studies relating to the sedative effects of hops are very scarce.
On the contrary, some spasmolytic effects have been found for hops.
An alcoholic extract of hop strobile (1 g of dried drug in 10 ml of 70% ethanol) produced a strong
spasmolytic effect on isolated smooth muscle from guinea pig intestine with ED 50 values equivalent to
37x10 -6 g of hop strobile per ml for acetylcholine-induced contractions compared to 60x10 -9 g/ml with
atropine, and 39x10 -6 of hop strobile per ml for bariumchloride-induced contractions compared to
57x10 -7 g/ml with papaverine. The extract also inhibited contractions of rat uterus with an ED 50
equivalent to 31x10 -6 g of hop strobile per ml (Caujolle et al., 1969).
Also an effect on the calcium flux has been detected.
A methanolic extract from hop strobile showed strong inhibitory activity on calcium fluxes, inhibiting
depolarization-induced 45 Ca 2+ uptake in clonal rat pituitary cells by 94,7% at 20 µg/ml (p<0.001).
The activity was attributed to prenylated flavonoids, although individual compounds from hop strobile
have not so far been tested in this way (Rauha et al., 1999).
Recently, Meissner and Häberlein (2006) investigated the influence of xanthohumol (X) on the
binding of muscimol-Alexa-Fluor 532 (Mu-Alexa), a fluorescently labeled GABA A receptor agonist
by fluorescence correlation spectroscopy. An incubation of hippocampal neurons with 75 nM of X
increased the specific Mu-Alexa binding with ca. 17%, which was selectivly found in GABA A
receptor Mu-Alexa complexes with hindered lateral motility [D bound2 = (0,11 ± 0,03) µm 2/ s ) ]
as described with midazolam, a benzodiazepine agonist. It was further shown that the modulatory
activity of X on the GABA A receptor was not mediated via an interaction with benzodiazepine
receptors. The authors concluded that X may play an important role for the sedative effects of hop
preparations.
II.2.2.1.2. In vivo studies
Old reports have indicated that preparations of hops have sedative-like activity in frogs (Staven-
Groenberg, 1928 ; Munch et al., 1933 ; Steidle, 1932), pigeons (Sikorski and Rusiecki, 1938), gold
fish (Bouchardy, 1953) and golden carp (Grumback and Mirimanoff, 1955). According to Wohlfart
(1982, 1993), however, these animal tests show many methodological shortcomings.
Only a few animal experiments have been carried out to study the supposed sedating effects of hop
extracts in recent times. Following gavage of different extracts from hops or lupulone, Hänsel and
Wagener (1967) observed no indication of sedation in mice or rats. They used three hop extracts, two
produced with ethanol and another one with methylisobutylketone. Both ethanolic extracts were dried
and administered in oil. Locomotor activity was unaffected up to doses of 500 mg/kg bw and no
antagonistic effect against metamphetamine-induced stimulation was observed. Hexobarbital-induced
© EMEA 2008
12/30
sleeping time remained unchanged. In the Rotarod test up to 200 mg/kg bw caused no impairment of
coordination, and no muscle relaxation was observed.
On the contrary, Bravo et al. (1974) described a reduction in locomotor behaviour of mice following
the intraperitoneal administration of three different extracts of hops (aqueous, ethereous and
alcoholic). The ethereous one was the most active, since it completely inhibited mice motor activity,
20 minutes after injection. Since, however, this effect was only seen in very high doses
(200 mg extract/20 g mouse) it cannot be responsible for the claimed sedative effect in patients. On the
other hand, this study clearly showed that the sedative effect of hops is strongly dependent on the type
of solvent used for the extraction procedure.
Lee et al. (1993a) observed a dose-dependent suppression of spontaneous locomotor activity in mice
after the intraperitoneal administration of 100 mg/kg (p<0.05), 250 mg/kg and 500 mg/kg (p<0.001) of
hop extract (96% ethanolic dry extract). The same authors further investigated the CNS effects of hop
extract, using other behavioural tests such as potentiation of pentobarbital-induced sleep, hypothermic
analysis and anticonvulsant tests (Lee et al., 1993b). Pentobarbital-induced sleeping time increased
dose-dependently ; not significant at 100 mg/kg, by 1,9-fold at 250 mg/kg (p<0,05) and 2,6-fold at
500 mg/kg (p<0,01). In the hot plate test, latency time for licking the forepaws increased with doses of
100 and 250 mg/kg (p<0,01). Rotarod performance decreased by 59% and 65% respectively at
250 mg/kg, and 500 mg/kg (p < 0,001 after 120 minutes). The time to onset of convulsion and survival
time after administration of pentylenetetrazole were significantly lengthened by 500 mg/kg (p<0,001),
but not by 250 mg/kg. A significant and time dependent fall in rectal temperature was observed after a
dose of 500 mg/kg (p<0,001 after 120 minutes). Thus hop strobile extract showed sedative and
hypnotic properties at lower doses (100-250 mg/kg), and at a higher dose of 500 mg/kg it also
produced anticonvulsive and hypothermic effects.
The relevance of these findings remains unclear, since all experiments were carried out after
intraperitoneal injection and no experiments were done following oral application, so that the
bioavailability of the preparation used remains questionable.
In a recent publication (Zanoli et al., 2005), the central effects of a hop CO 2 extract and a fraction
containing the α-acids were studied in animal experiments following oral application of the hop
extract dissolved in Tween 80 (10%) or the α-acid fraction dissolved in peanut oil. Acute effects on
locomotor activity and pentobarbital sleeping time were studied, as well as behavioural parameters in
the elevated maze and the forced swimming test. The authors observed a significant increase of the
pentobarbital-induced sleeping time in rats without affecting the latency to the loss of the righting
reflex. This effect was dose-dependent starting from a minimal dose of 10 mg/kg. Neither the extract
nor its α-acid fraction effected the locomotor activity in the open-field test or excerted an anxiolytic
effect in rats submitted to the elevated plus- maze test.
Interestingly, both hop CO 2 extract and the α-acids containing fraction significantly reduced the period
of immobility in the forced swimming test, when administered three times (24 h, 5 h and 1 h), before
the test indicating an antidepressant activity.
It was concluded that hop CO 2 extract and a hops containing α-acids fraction exert a pentobarbital-
enhancing property without influencing the motor behaviour of rats and an antidepressant activity.
These results seem to show that the α-acids present in hop CO 2 extract can explain the use of hops in
sleep disturbances and that the α-acids could be a new class of compounds for the development of
natural antidepressant agents.
More recently, Schiller et al. (2006) confirmed the sedating effects of the liphophilic extract reported
by Zanoli et al. (2005). They investigated several ethanolic extracts (40% v/v and 90% m/m) and
CO 2 extracts from diverse hop varieties as well as α-acids and β-acids fractions and pure hop oil.
All hop extracts increased ketamine-induced sleeping time in mice. The increase in duration of
ketamine narcosis proved to be a specific central effect and not caused by a pharmacokinetic
interaction, as could be confirmed by a comparable increase in ether-induced sleeping time. In contrast
to the findings of Zanoli et al. they also observed a reduction of locomotor activity. The low doses
Zanoli et al. applied (20 mg/kg/bw as maximum) may be the reason for this discrepancy.
Like Zanoli et al. no anxiolytic effects of hop preparations were found. A decrease in body
temperature induced by all hop extracts as additional parameter was observed, which confirms the
sedating activity of the hop preparations. The results of Schiller et al. are similar to those of Lee et al.
© EMEA 2008
13/30
(2003), who found a marked sedation viz . a reduction of locomotor activity, increase of pentobarbital-
induced sleeping time and antagonistic effects against pentylenetetrazol-induced convulsions
following intraperitoneal injection of a hop extract. On the contrary, the results of Hänsel and
Wagener (1967) are in clear contrast to the findings of Schiller et al. (2006). The latter authors explain
this discrepancy by the use of different raw materials, different conditions of storage by which the
content of α- and β-acids might be reduced significantly and different extraction solvents used.
Finally, Schiller et al. showed that not only the α-acids, but also the β-acids and the hop oil, although
to a lesser extent, exert distinct sedating effects and contribute to the activity of the plants. They also
do not rule out the presence of further sedating components in hops.
A degradation product of the bitter acids, humulones and lupulones, the five carbon olefinic alcohol,
2’-methyl-3-buten-2-ol, given intraperitoneally to mice at high dosage, 800 mg/kg showed central
nervous depressant activity (Hänsel et al., 1980 ; Wohlfart et al., 1983a). Although present only in
small amounts in fresh hops, higher levels of this compound may be generated in vivo by metabolism
of the bitter acids, reaching its maximum concentration after 2 years of storage at room temperature
(Hänsel et al., 1982 ; Wohlfart et al., 1982, Wohlfart, 1983b, Hänsel and Schultz, 1986)). The sedative
effect of this alcohol is comparable, in the same dosage range, to that of the structurally related drug
methylpentynol (Wohlfart et al., 1983a).
Thus, if this compound can fully explain the sedative activity attributed to hops, than it must be
formed in vivo from hop constituents such as bitter acids, that would then be considered as
“pro-drugs” analogous to the case of the oestrogenic activity (Chadwick et al., 2006).
The hypothesis, however that this alcohol can be formed in vivo by metabolisation of α-acids has not
been demonstrated to date (Zanoli et al., 2005). Similarly the proposal that the sedative effect of hops
was due to its content of myrcene, which has shown to have analgesic activity in mice (Hänsel and
Wohlfart, 1980) has not been established.
Recently, Grundmann et al. (2006) showed that the hypothermic effects of hops could be antagonized
with the competitive melatonin receptor antagonist luzindole. Based upon a study in which it was
found that a combination of valerian and hops interacts with serotoninergic 5-HT 4e , 5-HT 6 , 5-HT 7 and
melatoninergic ML 1 and ML 2 receptors (Abourashed et al., 2004 ; Butterweck et al., 2007, Brattström,
2007), these authors evaluated the hypothermic activity of hop extract in mice. In a dosage of
250 mg/kg hops extract significantly decreased body temperature in male BL6/C57 mice (ΔT-1,1°C)
2 h after oral administration. The effects of the plant extract were comparable to melatonin (50 mg/kg,
ΔT-0,8°C, 2 h after i.p. injection). The hypothermic effects of both, melatonin and hop extract could
be antagonised with the competitive melatonin receptor antagonist luzindole.
The authors concluded that this data suggest that the hypothermic effects of hop extract are mediated
through activation of melatonin receptors. Since it is known that melatonin has both hypnotic and
hypothermic effects at physiological levels and that the hypnotic effect may be mediated via the
hypothermic action of melatonin (Zemlan et al., 2005), a similar effect may be suggested for hop
extract. The authors also concluded that neither the α- and β-acids, nor the essential oil were
responsible for these effects (Personal communication by Butterweck).
II. 2.2.2. Oestrogenic effects
II. 2.2.2.1. In vitro studies
II.2.2.2.1.1. Oestrogenic activity of hop strobiles
Circumstantial evidence over many years, including menstrual disturbances reported to be common
among female hop pickers, linked hop strobiles with potential oestrogenic activity (Verzele, 1986 ;
De Keukeleire et al., 1999). In Germany, hop baths were used to treat gynaecologic disorders and hop
extracts have been reported to reduce hot flushes in menopausal women (Goetz, 1990). However,
early studies to confirm this activity experimentally were unconclusive or contradictory due to
methodology of inadequate sensitivity (De Keukeleire et al., 1997, 1999).
In a recent screening of plant drugs for oestrogenic activity, a 50%-ethanolic extract (2 g of hop
strobile to 10 ml) exhibited binding to oestrogen receptors in intact, oestrogen-dependent [ER(+)],
© EMEA 2008
14/30
human breast cancer MCF-7 cells with a potency equivalent to 0,5 µg of oestradiol per 2 g of dried
strobile (for comparison, the potencies of 2 g of thyme or red clover were equivalent to 0,5 or 3 µg of
oestradiol, respectively).
The extract also showed significant ability to stimulate cell proliferation in ER(+)T47D, but not in
ER(-)MDA 468, breast cancer cells (Zava et al., 1998). In contrast, in a different series of experiments,
a similarly-prepared extract of hop strobile at concentrations of 0,01-1,0% V/V was found to
significantly inhibit serum-stimulated growth of ER(+)T47D breast cancer cells (p<0,001)
(Dixon-Shanies and Shaikh, 1999).
Ovarian cells isolated from immature female rats, which 48 hours previously had been injected
(primed) with pregnant mare’s serum gonadotrophin, were incubated with follicle-stimulating
hormone to induce oestradiol secretion. Addition to the culture medium of purified water-soluble
fractions F 1 or F 2 from defatted hop strobile extract reduced the amounts of oestrogen E 2 released from
the ovarian cells (p<0,01) with a probably related decrease in cAMP release (p<0,05)
(Okamato and Kumai, 1992).
In 2005 Overk et al. compared the oestrogenicities of the extracts of hops and red clover ( Trifolium
pratense ) and those of their individual constituents, including respectively prenylated flavanones and
isoflavonoids, using a variety of in vitro oestrogenic assays. The hop extract consisted of a chloroform
partition of a methanolic extract from a previously SF-C0 2 -extracted Nugget hops cultivar and the red
clover extract was an ethanolic extract containing 30% isoflavonoids prepared for a phase II clinical
trial.
The IC 50 values for the oestrogen receptor α and β binding assays (according to Obourn et al. (1993)
and Liu et al. (2001)), were 15 and 27 µg/ml, respectively for hops and 18,0 and 2,0 µg/ml,
respectively for the red clover extract.
Both of the extracts demonstrated also significant activities of transiently transfected ERE-luciferase,
quantitative RT-PCR of an oestrogen-inducible gene, and AP-enzyme induction assays (EC 50 values of
1,1 for hop extracts and 1,9 µg/ml for red clover extracts).
II.2.2.2.1.2. Oestrogenic activity of 8-prenylnaringenin (8-PN)
8-PN, a flavanone occurring in hop strobiles at levels of 25-60 mg/kg (Rong et al., 2000), has been
shown to be a potent phyto-oestrogen with activity greater than that of other established plant
oestrogens (Milligan et al., 1999).
Oestrogenic activity of a much lower order (less than one-hundredth of that of 8-PN) has also been
detected in three other hop flavanones, 6-prenylnaringenin (6-PN), 8-geranylnaringenin (8-GN) and
6,8-diprenylnaringenin (6,8-PN), while xanthohumul (X) and isoxanthohumol (IX) were found to be
inactive (Milligan et al., 2000). EC 50 values for 17β-oestradiol, 8-PN, 6-PN, coumestrol, genistein and
daidzein were 0,3, 40, > 4000, 70, 1200 and 2200 nM, respectively in a screen using oestrogen-
inducible yeast ( Saccharomyces cerevisiae ) expressing the human oestrogen receptor, and
0,8, 4, 500, 30, 200 and 1500 nM respectively, for stimulation of alkaline phosphatase activity in a
human endometrial cell line (Ishikawa Var I). The relative binding affinities of 17β-oestradiol,
8-PN, coumestrol and genistein with rat uterine cytosol containing soluble oestrogen receptor
were 1, 0023, 0008 and 0,003 respectively (Milligan et al., 1999).
The high oestrogenic activity of 8-PN was confirmed by Zierau et al. (2002) using two different
bioassays viz . a yeast based oestrogen receptor assay, containing a stably transfected oestrogen
receptor α (ERα) construct and an expression plasmid carrying oestrogen-responsive sequence
controlling the reporter gene lac-Z encoding the enzyme β-galactosidase and a transactivation assay
using MVLN cells, a bioluminescense MCF-7 derived cell-line.
The same research group showed using a yeast-based androgen receptor assay strong anti-androgen
activities for 8-PN and 6-(1,1-dimethylallyl)naringenin (6-DMA-N), while the parent compound
naringenin was inactive. In an androgen receptor activity-assay based on the analysis of putative
specific antigen (RSA) concentrations in the supernatants of treated PC3(AP)2 cells only
6-DMA-N showed antiandrogenic activity at concentrations of 10 -5 . 8-PN and naringenin had no
detectable antiandrogenic effects.
© EMEA 2008
15/30
6-DMA-N is a structurally related compound, isolated from the African tree Monotes engleri
(Zierau et al., 2003).
The oestrogenic activity of 8-PN was further confirmed in competitive binding assays using purified
human recombinant oestrogen receptors α and β (ER α and ER β ). 8-PN competed strongly with
17β-oestradiol for binding to both receptors with a relative binding affinity of about 0,1, compared to
1,0 for 17β-oestradiol and 0,001 for 8-GN (Milligan et al., 2000).
In another study, involving displacement of [ 3 H ]-17β-oestradiol, 8-PN showed competitive binding
affinity for the oestrogen receptor in bovine uterine cytosol with an IC 50 of 140 nM, compared to
1,0 nM for 17β-oestradiol and 320 nM for genistein. 8-PN also dose-dependently stimulated the
proliferation of cultured, oestrogen-dependent, human breast cancer MCF-7 cells with an EC 50 of
1,9 nM, compared to 0,0032 nM for 17β-oestradiol and 47 nM for genistein, suggesting that it was an
oestrogen receptor agonist (Kitaoka et al., 1998).
The same authors synthetized racemic 8-PN, separated and assayed both enantiomers, and reported
that there was no significant difference in ER binding potency between the 2R and the 2S forms
(Kitaoka et al., 1998).
In an in vitro receptor binding assay, using recombinant human ER α and ER β from cytosolic SF 9 -cell
extracts, Schaefer et al., (2003), showed that 8-PN exhibited > 2-fold higher affinity for ER α than ER β .
Using a mammalian cell-based transactivation assay consisting of U 2 -osteosarcoma cells transient
transfected with either ER α or ER β and a luciferase reporter gene construct these authors demonstrated
that 8-PN is the strongest plant-derived ER α agonist identified so far, being 10-fold more potent than
coumestrol and 100-fold stronger than genistein, but only 70 times weaker than 17β-oestradiol.
The transactivational analysis revealed also a >3,6-fold higher oestrogenic activity of 8-PN at ER α
than ER β , a strong contrast to primarily ERβ-activating coumestrol and genistein.
At the same time they found that the in vivo oestrogenic activity of 8-PN in reproductive tissue was
about 20,000-fold weaker compared to 17β-oestradiol.
As such, 8-PN mimics the effects of the endogenous hormone and could be a natural SERM (selective
oestrogen receptor modulator) with promising potential for the treatment of various oestrogen
deficiency-related conditions (Schaefer et al., 2003).
In 2004 Bovee et al. developed a rapid yeast oestrogen bioassay stably expressing oestrogen receptors
α and β and green fluorescent proteins and tested various phyto-oestrogens. They confirmed the
earlier results of Schaefer et al. that 8-PN is relatively more potent with ER α and less potent than
coumestrol and genistein with ER β .
In 2005 Overk et al. compared the oestrogenicities of the extracts of hops and red clover and those of
their individual constituents viz . 8-PN, 6-PN, IX and X from hops and daidzein, formononetin,
biochanin A and genistein from red clover.
The results of the extracts have been discussed under point II.2.2.2.1.1. Here we give the results of the
individual constituents.
The competitive ER α and ER β binding assays using tritiated 17β-oestradiol are based on the method of
Obourn et al. (1993) with minor modifications (Liu et al., 2001). As already mentioned before the red
clover extract preferentially bound to the ER β receptor nine times greater than to ER α . The hop extract
had nearly a 2-fold preference for the ER α as compared with ER β . For the isolated compounds, the
relative binding affinities for ER α were as follows :
genistein ≈ 8-PN > daidzein > biochanin A > formononetin > IX, while the relative binding affinities
for ER β were genistein > 8-PN ≈ daidzein > biochanin A > formononetin ≈ IX.
Genistein and 8-PN had similar affinities for ER α with IC 50 values of 0,51 and 0,3 µM respectively,
whereas genistein was more selective for ER β as indicated by the IC 50 value of 0,020 and 1,7 µM,
respectively. Both of the extracts, genistein and 8-PN activated the oestrogen response element (ERE)
in Ishikawa cells, while the extracts, biochanin A, genistein, and 8-PN significantly induced
ERE-luciferase expression in MCF-7 cells.
© EMEA 2008
16/30
Hop and red clover extracts as well as 8-PN up-regulated progesterone receptor (PR) m-RNA in the
Ishikawa cell line. In the MCF-7 cell-line, PR m-RNA was significantly up-regulated by the extracts,
biochanin A, genistein, 8-PN and IX. The two extracts had EC 50 values of 1,1 and 1,9 µg/ml
respectively in the alkaline phosphatase induction assay. The EC 50 values of the individual compounds
are consistent with values reported by Milligan et al. (1999) viz . 8 PN >> genistein ≈ daidzein > XI >
biochanin A. None of the compounds had anti-oestrogenic activity.
II.2.2.2.2. In vivo studies
II.2.2.2.2.1. Antigonadotrophic effects of hop strobiles
Purified water-soluble fractions from defatted hop strobile extract were administered subcutaneously
twice daily for 3 days to immature female rats primed with 25 IU of pregnant mare’s serum
gonadotrophin (PMSG). None of the fractions induced a change in uterine weights. However, fractions
F 1 (20 mg/rat) and F 2 (50 mg/rat) significantly suppressed PMSG-induced gain in ovarian weights by
about 25% (p<0,05) compared to controls. Under the same conditions, two further fractions (4 mg/rat)
purified from F 1 suppressed gain in ovarian weights by 42% and 33% (p<0,01) compared to controls
(Kumai and Okamoto (1984). In further experiments on PMSG-primed immature rats, by comparison
with saline-treated control animals, subcutaneously administered fractions F 1 and F 2 reduced the
number of ovulations (p<0,05) ; suppressed levels of serum luteinizing hormone (p<0,001) ;
suppressed thymidine kinase activity in uterine tissue (p<0,01) ; reduced 17β-oestradiol E 2 secretion in
cultures of ovarian cells from the rats (p<0,001) ; and reduced progesterone production in cultures of
luteal cells from the rats (p<0,05 to p<0,001) (Okamato and Kumai, 1992).
To date no other in vivo studies on the oestrogenic effects of hop strobiles have been published.
II.2.2.2.2.2. Oestrogenic effects of 8-prenylnaringenin (8-PN)
In vivo studies with 8-PN are scarce. Only one report highlighted favourable effects of hops on hot
flushes in humans (Goetz, 1990), but preclinical studies aimed at evaluating the efficacy of 8-PN for
alleviating menopausal symptoms and discomforts are still ongoing. Most investigators until now have
focused on animal models. It should be noticed that the relative potency of oestrogenic compounds
in vivo largely depends on a number of factors, including the route of administration and the nature of
the response monitored. Moreover, estimates of oestrogenic activity are markedly affected depending
on the nature of the in vivo bioassay used. Milligan et al. (2002) tested 8-PN in two in vivo assays viz.
an assay based on the rapid response of the uterine vasculature to oestrogenic stimulation and an assay
based on the uterotrophic reponse and mitotic responses of the uterine and vaginal epithelium. In both
assays ovariectomized female Swiss albino mice, about 2-3 months of age, were used.
In the first assay, a quantitative index of the vascular permeability was obtained from the leakage of
radiolabelled albumin from the circulation. Several test substances were subcutaneously administered
to the ovariectomized mice and after 4 hours a significant increase in the vasular permeability was
obtained for oestriol and 17β-oestradiol, but both coumestrol and 8-PN were considerably less potent
(< 1% relative to 17β-oestradiol). The dose-response relationship for 8-PN was similar to that of
coumestrol, and a large stimulatory effect was produced by 100 mmoles 8-PN. The amount of
genistein required to produce the same effect was at least ten-fold greater. The administration of
daidzein produced no detectable uterine vascular response at the doses used. The responses of 8-PN
and 17β-oestradiol were blocked completely by prior treatment of the animals with anti-oestrogen
(ICI 182, 780).
In the second assay the oestrogenic potency of the test substances was tested after continuous
administration in the drinking water by monitoring the uterotrophic response and cell mitosis in the
vaginal and uterine epithelia.
Both 8-PN (100 µg/ml) and 17β-oestradiol (100 ng/ml) produced significant increase in vaginal
mitosis after 72 h compared with the negative control (p<0,05 and p<0,005, respectively). However,
although 17β-oestradiol also produced significant increases in uterine mass and in epithelial mitosis,
there were no significant differences in the mice exposed to any of the 8-PN treatments.
© EMEA 2008
17/30
They concluded that in addition to its in vitro activity 8-PN shows oestrogenic effects in the two
in vivo test systems. It was noted that 100 µg 8-PN/ml in the drinking water (equivalent to an intake of
about 15 mg per kg per day) was able to induce the characteristic oestrogenic mitotic response in the
vaginal epithelium of ovariectomized mice. The apparent lack of effect of 8-PN on uterine epithelial
mitosis may reflect with the limited amounts given or the temporal differences in mitotic responses
induced by oestrogens in the uterine luminal, uterine glandular and vaginal luminal epithelia of
ovariectomized mice (Finn and Publicover, 1981).
Coldham and Sauer (2001) showed that a dietary supplement for breast enhancement containing hop
extracts was only weakly active in mouse uterotrophic assays following administration in feed or after
subcutaneous injection of the extract at doses of 8-PN up to 250 times higher than that recommended
for women. They concluded that the dietary supplement is unlikely to produce oestrogenic effects
in vivo at the level of the uterus.
In other experiments ovariectomized rats, as a model for oestrogen deficiency induced osteoporosis,
were treated subcutaneously with racemic 8-PN (OVX + 8-PN) at 30 mg/kg/day or with
17β-oestradiol (OVX + OE) at 0,01 mg/kg/day, or with vehicle only (OVX). Another group of rats
was sham-operated, i.e. subjected to ovariectomy surgery without removing the ovaries, and treated
with the vehicle only (sham).
After 2 weeks of treatment, 24-hour urine samples were collected and body weight gain, uterine
weight and bone mineral density were determined. The uterine weights of sham and OVX + 8-PN rats
were found to be 165% higher (p<0,001), and of OVX + OE rats 235% higher (p<0,001), than those of
OVX rats.
Body weight gains of sham, OVX + OE and OVX + 8-PN were significantly lower (p<0,05) than
those of OVX rats. Urinary excretion of hydroxyproline (a conventional marker of bone resorption)
was 1.62 µg/g/day from OVX rats compared to 1,18 and 1,16 µg/g/day for sham and OVX + OE rats
respectively (p<0,01), and 1,01 µg/g/day from OVX + 8-PN rats (p<0,001). The levels of urinary
hydroxypyridinium cross-links (assayed as pyridinoline and deoxypyridoline), which are recognized to
be directly related to bone matrix degradation, were significantly lower in sham rats (p<0,01), and in
OVX + OE and OVX + 8-PN rats (p<0,001), than in OVX rats.
Bone mineral densities of 139,1, 141,9 and 141,9 mg/cm 2 in sham, OVX + OE and OVX + 8-PN rats,
respectively, were significantly higher (p<0,001) than that in OVX rats (132,1 mg/cm 2 ). It was
concluded that 8-PN functions as an oestrogen receptor agonist in reproductive tissues and that the
dosage used had completely prevented ovariectomy-induced bone loss (Miyamoto et al., 1998).
Recently, the influence of 8-PN on oestrogen-related genexpression in liver and uterus tissues was
investigated in rats (Diel et al., 2004). At a 100-fold dose with respect to 17β-oestradiol, 8-PN induced
a qualitatively similar, but less pronounced expression profile. On the other hand, 8-PN was found to
be more potent than 17β-oestradiol in inducing expression of IGFBP-1 (Insulin growth factor binding
protein-1). This factor has been correlated to an improvement of the vascular endothelial function and
blood pressure homeostasis (Laughlin et al., 2004), while higher concentrations of IGFBP-1 are also
associated to a decreased risk of prostate cancer (Ngo et al., 2003).
Schaefer et al. (2003) determined the oestrogenic potency in vivo and in vitro of 8-PN using the classic
uterine growth assay and vagina growth assay in juvenile rats. Their findings confirm the conclusions
of Diel et al. (2004) that 8-PN is a pure oestrogen agonist in vitro , exhibiting an oestrogenic activity
profile comparable to oestron. The in vivo oestrogenic activity of 8-PN in representative tissue,
however, is 20,000-fold weaker compared to 17β-oestradiol. They also suggested that 2S(-)-8-PN has
moderately higher ER affinity and oestrogenic activity, in vitro and in vivo than 2R(+)-8-PN.
In 2005 Hümpel et al. studied 8-PN in adult ovariectomized rats, an established animal model to
mimic hormone dependent osteoporosis in menopausal women. The study demonstrated that 8-PN can
completely protect from ovariectomy induced bone-loss while exhibiting minimal (dose independent)
trophic effects on uterus and endometrium. It is estimated that at equivalent bone protective doses of
17β-oestradiol and 8-PN, the phyto-oestrogen has a 10-fold lower stimulatory effect on uterus and
endometrium. The tissue specific effect of 8-PN was confirmed in a transgenic receptor mouse model
(ERE-LUC mice). Here they also found pronounced oestrogenic activity in prostate.
© EMEA 2008
18/30
In a recent in vivo study using the medaka sex reversal/vtg gene expression assay no oestrogenic
effects of all the naringenin-type flavonoids including also 8-PN were observed. Natural sex steroid
hormones and isoflavonoids such as genistein, on the contrary, can functionally reverse the phenotype
sex of fish (Zierau et al., 2005).
Assessor’s comments
Sedative effects
Although no in vitro studies, directly related to the sedative activity of hop strobiles have been
published, nevertheless several studies in animals have been carried out to investigate the
neuropharmacological properties of Humulus lupulus , traditionally used in the treatment of
different CNS disorders such as insomnia, excitability and restlessness.
One of the studies showed that hop strobile extracts exert sedative and hypnotic properties in mice
at lower doses (100-250 mg/kg) and produce other activities such as anticonvulsive and
hypothermic effects at higher doses of 500 mg/kg.
Hop extracts were able to prolong the pentobarbital sleeping time without affecting the latency to
the loss of the righting reflex in rats, confirming the first study concerning the hypnotic properties
and the traditional observation of sleepiness in hops-pickers. In the same study it was shown that
the α-acids were responsible for the pentobarbital sleep-enhancing properties as well as for the
antidepressant activity found for the hop strobile extract.
In a recent study, carried out with several ethanolic and CO 2 extracts of hops, administered by
gavage to mice, it was clearly shown that all extracts reduced the spontaneous locomotor activity,
increased the ketamine-induced sleeping time and reduced body temperature, confirming a central
sedating effect. No indications of anxiolytic activity were found for any of the test preparations.
The sedating activity could be attributed to three categories of constituents of lipophilic hop
extracts. Though the α-bitter acids proved to be the most active constituents, the β-acids and the
hop oil clearly contributed to the sedating effect of lipophilic hop extracts as well.
The authors also suggested that the contradictory results obtained by different research teams in
the determination of different pharmacological effects was due to the use of different raw
materials, different storage conditions leading to various amounts of active components and the
use of different extraction solvents for the production of the preparations.
Furthermore, candidate molecules for the sedative effects of hop strobiles such as myrcene and
2’-methyl-3-buten-2-ol have been suggested, but their role as sedative agents has not been
substantiated.
Recently, however, it has been shown that one of the chalcones viz . xanthohumol influences the
GABA A receptors and their lateral mobility at hippocampal neurons in a similar way as the
benzodiazepine agonist midazolam without interfering with the benzodiazepine receptors.
Thus, xanthohumol may play an important role for the sedative effect of hop preparations, but this
should be confirmed by in vivo studies.
More recently the in vitro study in which it was suggested that a combination of hops and valerian
interacts with melatoninergic ML 1 receptors, was confirmed by an in vivo investigation on mice.
It was shown that hop extracts (250 mg/kg) had comparable hypothermic effects as melatonin
(50 mg/kg), which could be blocked by the competitive melatonin receptor antagonist luzindole.
Since it is known that the hypnotic effect of melatonin may be mediated via its hypothermic
action, a similar effect for hops may be suggested.
The authors also concluded that this effect was not due to the acids or the essential oil of hops,
since both were absent in the hop extract used in this study.
Oestrogenic effects
In contrast to the studies on the sedative effects, the phyto-oestrogen, responsible for the
oestrogenic activity of hop strobiles has been isolated and identified as the prenylated flavanone,
8-prenylnaringenin or hopein (8-PN). The oestrogenic activity proved to be considerably greater
than that of established phyto-oestrogens such as coumestrol (present in red clover) and genistein
and daidzein (present in soy). The oestrogenicity has been examined in great detail by a number
© EMEA 2008
19/30
of independent research groups worldwide. The other prenylated flavonoids were extremely
weakly oestrogenic or devoid of any oestrogenicity. 8-PN exerts its activity through oestrogen
receptor-mediated mechanisms. It binds very strongly to both oestrogen receptor isoforms
(ER α and ER β ), but in contrast to most known phyto-oestrogens, 8-PN is selective for the
oestrogen receptor-α. As such it mimics the effects of the endogenous 17β-oestradiol. Both have
similar profiles, however, the activity of 8-PN is 5- to 100-fold weaker depending on the test
system and the particular reaction conditions.
8-PN is orally active in ovariectomized mice as a significant increase in vascular permeability was
observed within 4 hours after subcutaneous injection of both 17β-oestradiol and a 100-fold dose of
8-PN. Administration of 17β-oestradiol (100 ng/ml) or 8-PN (100 µg/ml) to the drinking water
showed, after 71 h, a considerable increase in vaginal mitosis. Although 17β-oestradiol caused
also a substantial increase in uterus weight and in epithelial mitosis, significant differences with
8-PN were not observed.
In other experiments, ovariectomized rats were injected with 8-PN over a period of 14 days.
A favourable effect on bone metabolism was observed. Removal of the ovaria normally results in a
drastic increase in levels of bone resorption markers in urine, a decrease in mineral bone density
and a reduction in uterus weight. Treatment with 17β-oestradiol (0,01 mg/kg/day) or 8-PN
(30 mg/kg/day) gave quantitatively comparable effects on bone and uterus showing that 8-PN
functions as an oestrogenic agonist.
The influence of 8-PN on oestrogen-related gen expression in liver and uterus tissues was
investigated in rats. At a 100-fold dose with respect to 17β-oestradiol, 8-PN induced a
qualitatively similar, but less pronounced expression profile. On the other hand a stronger
upregulation of the expression of IGFBP-1 was observed. This growth factor has been correlated
to an improvement of the vascular endothelial function and blood pressure homeostasis, while
higher concentrations of IGFBP-1 are also associated with a decreased risk of prostate cancer.
In conclusion, 8-PN, present in hop strobiles at levels of 25-60 mg/kg, should be considered to be the
active oestrogenic compound of hops. It should be noticed that desmethyl-xanthohumol serves as
pro-oestrogen, since it can be metabolised into a mixture of 8-PN and 6-PN.
II. 2.2.3. Other pharmacological activities of hop strobiles and/or its constituents
In a very comprehensive review article, De Keukeleire and Heyerick (2005) discussed the intriguing
biological activities of hops and its constituents.
II.2.2.3.1. Hop essential oil and hop acids
Antimicrobial activity
Some antimicrobial activity of hop essential oil has been reported (Racz et al., 1980 ; Grange and
Davey, 1990 ; Langezaal et al., 1992 ; Simpson and Smith, 1992 ; Ohsugi et al., 1997 ; Tagashira et
al., 1997 ; Matos et al., 2001) and hops are included in various cosmetic preparations. The lupulones
(β-acids) have a longstanding reputation as antibacterials (against gram-positive bacteria), which had
led to some applications of hop products rich in β-acids, particularly in the sugar industry. It appears
that the three prenyl groups present in lupulone interfere with the building up and the functioning of
the bacterial cell walls leading eventually to leakage of the cell contents.
Resistance of gram-negative bacteria to the resin acids is attributed to the presence of a phospholipid-
containing outer membrane, as lupulones and humulones are inactivated by serum phospholipids
(Teuber and Schmalreck 1973). Structure-activity relationship studies have indicated the requirement
of a hydrophobic molecule and a six-membered central ring for such activity (Schmalreck et al.,
1975).
The acids are thought to possess little activity towards fungi or yeasts. However, antifungal activity
has been documented for the bitter acids towards Trichophyton, Candida, Fusarium and Mucor species
(Mizobuchi and Sato, 1984).
© EMEA 2008
20/30
Antidiabetic activity
Isohumulones are the main bittering principles in beer. Recently, interesting antidiabetic properties of
isohumulones have become apparent (Kondo, 2003). Via a reporter system, it was found that agonistic
effects were exerted on the activity of the nuclear receptors PPARalpha (‘Peroxisome Proliferator-
Activated Receptor-alpha’) and PPARgamma. The PPAR’s are important regulators of the glucose
and fat metabolisms and agonists are applied to treat non-insulin-dependent diabetes (diabetes type II)
and hyperlipidemia. Administration of isohumulones to a mouse model for diabetes type II (KK-A y )
led to a decrease of plasma triglycerides and free fatty acids in a dose-dependent manner. Moreover,
the glucose levels were significantly diminished in KK-A y -mice. The results indicate that intake of
isohumulones may favourably influence conditions of hyperglycemia and hypertriglyceridemia.
Treatment of 20 subjects suffering from mild diabetes with isohumulones (twice a day during
12 weeks), in dosages that were equivalent to only a few glasses of strongly hopped beers, showed a
decline in glucose levels and other parameters, indicating that oral intake of isohumulones ameliorates
insulin sensitivity In patients with diabetes type II.
Activity on osteoporosis
Humulone, the major constituent of the mixture of α-acids, has been shown to inhibit bone resorption
using an in vitro “pit formation assay” (formation of pits on dentine slices incubated with mouse bone
cells). Xanthohumol and humulone have been identified as inhibitors of bone resorption at
concentrations at or above 10 -6 and 10 -11 , respectively (p<0,01).
Humulone showed high inhibitory activity with an IC 50 of 5,9x10 -9 M (Tobe et al., 1997a). These
findings indicate that hops may be active against osteoporosis.
It should be noted, however, that xanthohumol and especially humulone, have no oestrogenic
activities, which may indicate that the inhibition of bone resorption of hops is not associated with
oestrogenicity (De Keukeleire et al., 1999).
Antinflammatory activity
Humulone also proved to be a potent inhibitor of the expression of cyclooxygenase-2 (COX-2) via
interaction with NFκB, which translates into pronounced anti-iflammatory activity (Yamamoto et al.,
2000). Also in vivo anti-inflammatory effects have been seen for humulone, which seemed to be the
active anti-inflammatory agent of hop strobiles.
A dry methanolic extract of hop strobile, applied topically at 2 mg/ear, inhibited
12- 0 -tetradecanoylphorbol-13-acetate (TPA)-induced ear oedema in mice by 90% (p<0,01) six hours
after TPA treatment. Humulone, isolated from hop strobile by bioassay-guided fractionation and
identified as an anti-inflammatory constituent, inhibited the oedema with a ID 50 of 0,2 mg/ear
(ID = inhibitory dose).
Topically-applied humulone also inhibited arachidonic acid-induced inflammatory ear oedema in mice
with an ID 50 of 2,2 mg/ear (p<0,01 against controls) compared to 0,4 mg/ear (p<0,01) for indometacin
(Yasukawa et al., 1993).
Antiproliferative activity
Humulone has been found to inhibit angiogenesis (formation of new blood vessels, which is essential
for tumour growth), as well as proliferation (uncontrolled growth) of endothelial cells (Shimamura
et al., 2001). The latter effect has been confirmed by an in vivo test, in which tumour promotion could
be inhibited.
Humulone applied topically at 1 mg/mouse to the backs of mice markedly inhibited the tumour-
promoting effect of TPA on 7,12-dimethylbenz[ a ]-anthracene-initiated skin tumour formation. In the
control group 100% of mice developed tumours (first tumour appeared in week 6), compared to only
7% in the humulone-treated group (first appearance in week 16). Humulone treatment resulted in a
99% reduction in the average number of tumours per mouse at week 18 (p<0,01) (Yasukawa et al.,
1995).
© EMEA 2008
21/30
II.2.2.3.2. Hopprenylflavonoids
Cancer chemopreventive activity
8-Prenylnaringenin (8-PN) is effective in the aggregation of MCF-7/6 breast cancer cells, which
indicates that the compound may inhibit metastasis (Rong et al., 2001). Moreover, it was shown that
8-PN inhibits angiogenesis in vitro and in vivo (Pepper et al., 2004). Various tests using different
cancer cell lines showed no toxicity of 8-PN at levels below 50 µg, while antiproliferative activities
were also observed (Tokalov et al., 2004).
Xanthohumol (X) and to a lesser extent dehydrocycloxanthohumol (DHX) and isoxanthohumol (IX)
show a broad spectrum of inhibitory mechanisms at the initiation, promotion and progression stage of
carcinogenesis (Gerhauser et al., 2002).
On examining proliferation of human breast cancer cells (MCF-7 and MDAMB 231), colon cancer
cells (HT-29), prostate cancer cells (LNCaP and PC-3) and cancer cells of the ovaries (A-2780), a
dose-dependent inhibition of the growth of all cancer cells was noted on addition of low
concentrations of X (low micromolar range) (Miranda et al., 1999 ; Gerhauser et al., 2001a ; 2002).
The activity is correlated with inhibition of DNA polymerase-alpha, the only eukaryotic enzyme
capable of initiating DNA synthesis de novo . In the presence of X , many cancer cells undergo
apoptosis and programmed cell death. An additional antiproliferative mechanism was found in HL-60
leukemia cells (Gerhauser et al., 2002). Terminal cell differentiation is induced whereby cancer cells
start to behave normally again. In these tests, X was 10-fold more active than genistein present in soy.
Inhibition of carcinogenesis ex vivo was also demonstrated for X using so-called mouse mammary
gland organ cultures. In this model system, breast tissue is removed from mice and small cancer
lesions are caused by addition of a potent carcinogen. Extremely low concentrations of X (low
nanomolar range) completely inhibited formation of these lesions (Gerhauser et al., 2002).
Interestingly, resveratrol, a stilbenoid-type polyphenol held responsible for a number of health-
beneficial effects of red wine, was 210-fold less active in this assay.
X showed an inhibitory activity on specific cytochrome P450 enzymes that mediate the conversion of
procarcinogens to carcinogens (Miranda et al. 2000a ; Henderson et al., 2000). Also, it was observed
that X induces the activity of the enzyme quinone reductase in vitro , which is indicative of
detoxification of a variety of carcinogens (Miranda et al., 2000b ; Dietz et al., 2005). The compound
was able to scavenge reactive oxygen species and to interfere with formation of radicals. On trapping
of hydroxyl and peroxy radicals, X was 9- and 3-fold, respectively, more active than a well-known
reference, Trolox (Gerhauser et al., 2001b), while inhibition of oxidation of low-density lipoproteins
(LDL) was more effective than that caused by alpha-tocopherol (Miranda et al., 2000c).
Interestingly, studies have shown that X has anti-inflammatory properties by inhibition of the activities
of cyclooxygenases (COX-1 and COX-2) that account for the production of prostaglandins
(Gerhauser et al., 2002).
Effects on the synthesis of triglycerides
Xanthohumol (X) considerably interferes in the synthesis of triglycerides. The activity of
diacylglycerol transferase, a liver enzyme which is involved in the formation of glycerides, was
substantially inhibited (Tabata et al., 1997). This interaction is associated with positive effects on
hypertriglyceridemia including a decreased risk for associated diseases such as atherosclerosis and
diabetes. It was very recently found, using HepG2 cells as model system, that X decreased
apolipoprotein B secretion in a dose-dependent manner under both basal and lipid-rich conditions
(Casaschi et al., 2004). Furthermore, X inhibited the synthesis of triglycerides in the microsomal
membrane and the transfer of newly synthesized triglycerides to the microsomal lumen indicating that
triglycerides availability is a determining factor in the regulation of apolipoprotein B secretion. The
inhibition of triglycerides synthesis is caused by a reduction in diacylglycerol transferase activity
suggesting that X is a potent inhibitor of apolipoprotein B secretion.
© EMEA 2008
22/30
Assessor’s comments
Besides the most potent phyto-oestrogen currently known with possible health-beneficial properties
(8-PN), hops also contains xanthohumol, the quantitatively most important prenylchalcone (up to
1,3% m/m), which displays potential therapeutic properties (Gerhauser and Frank, 2005)
This compound shows a wide spectrum of inhibition mechanisms at all stages of carcinogenesis. Low
concentrations of X dose-dependently inhibit the growth of cancer cells among them breast cancer
cells (MCF-7 and MDAMB-231) and cancer cells of the ovaries (A-2780).
Consequently, if hop-based phytotherapeuticals would be developed as potential alternatives to
hormone replacement therapy for menopausal women, not only the high oestrogenic potential of 8-PN
should be taken into account, but also the cancer chemopreventive activity of xanthohumol.
Standardisation of both active components in hop-derived extracts is therefore recommended.
The studies described in this assessment report may be considered as a rationale for further studies and
trials to establish efficacy.
II. 2.3.
Interactions
Limited evidence from one animal study suggests that hops may potentiate the effects of sedative
drugs (Lee et al., 1993b).
Interactions with other phyto-oestrogens such as alfalfa, black cohosh, bloodroot, burdock, kudzu,
licorice, red clover, soy and others, are theoretically possible. It should be noted, however, that like
17β-oestradiol, 8-PN is an agonist, whereas phyto-oestrogens containing isoflavonoids as active
oestrogenics are known to be mixed agonists/antagonists. Soy isoflavonoids act as agonists when no
endogenous oestrogens are available, but become antagonists when high endogenous levels of
oestrogens are available.
Laboratory research shows that oestrogen-like substances in hops may have effects on oestrogen-
sensitive parts of the body. It is not clear what interactions may occur when hop extracts are used
together with other hormonal therapies such as birth control pills, hormone replacement therapy,
tamoxifen, or aromatase inhibitors such as letrozole, because no studies have been concluded to
confirm this. Hops may interfere with the way the human body processes certain drugs using the
liver’s ‘cytochrome P450’ enzyme system, so that the levels of certain drugs may be decreased in the
blood, but they have not been confirmed by studies. Clinical cases of drug interactions with hops have
not been reported to date.
II.3.
CLINICAL EFFICACY
II. 3.1.
Clinical studies
II. 3.1.1. Sedative activity
No clinical studies have been conducted to date with hops or hop preparations as single component
products for the treatment of restlessness or insomnia.
Four non-controlled and three placebo or reference-controlled, double-blind clinical studies in patients
suffering from non-organic sleep disorders have been conducted with a fixed extract combination
(Ze 91019) of valerian root and hop strobile extracts. Both extracts were prepared with 45% methanol
m/m with a dry extract ratio of 5-3:1 (valerian) and 6.6:1 (hops), respectively. (Brattström, 1996,
Lataster and Brattström, 1996, Flesch, 1997, Füssel et al., 2000, Notter et al., 2003, Rodenbeck and
Hajak, 1998, Schellenberg et al., 2004 and Koetter et al., 2007).
In addition, one non-controlled and four placebo or reference-controlled double-blind clinical studies
are reported using a related fixed combination product consisting of dry extracts of valerian root and
hops (Wegener et al., 2003, Schmitz and Jäckel, 1998, Morin et al., 2005, Leathwood et al., 1982,
Müller-Limmroth and Ehrenstein, 1977).
© EMEA 2008
23/30
Assessor’s comments
Preclinical and clinical studies are available for combinations of hops with other sedative plants such
as valerian, passion flower and lemon balm. Consequently, separate monographs on combinations will
be elaborated.
II. 3.1.2. Oestrogenic activity
As described in previous sections, preclinical in vitro and in vivo tests suggested that hop extracts
exert an oestrogenic activity.
Before the publication in 1999 by Milligan et al., only one small clinical trial specifically related to the
oestrogenic properties of hops had been identified. In this placebo controlled study, 20 patients
experiencing hot flushes due to ovarian insufficiency (15 in menopausal phase and 5 following
ovariectomy), were treated with a dry aqueous extract of hop strobile (5:1), initially at 1,6-2,6 g/day,
later in some cases reduced to 1,2-1,6 g/day. Five other patients received placebo. Assessment was
based on scores calculated by multiplying the intensity of hot flushes (scale 1 to 3) by their frequency
(scale 1 to 9). In verum patients, the initial average score of 22,7 decreased to 8,2 after 30 days of
treatment, whereas in placebo group, the initial score of 20 decreased only to 18. Compared to the
placebo group, 76% of the verum patients achieved a statistically significant improvement in scores
and 7 out of 20 patients achieved a reduction of at least 15 points (Goetz, 1990). While the author
reported that the formulation was effective in treating hot flushes, it was neither chemically nor
biologically standardized by any modern standards (Chadwick et al., 2006).
Recently, a first prospective, randomized, double-blind placebo-controlled study on the use of a hydro-
alcoholic standardized hop extract to alleviate menopausal discomforts was published (Heyerick et al.,
2006). The hop extract was standardized on 8-PN (100 or 250 µg), which may seem a small amount,
but comparison of 8-PN with established phyto-oestrogens indicated that 8-PN had several 100-fold
more potent oestrogenic activity than e.g. soy isoflavones such as genistein and dadzein, which have
been administered in daily doses of 50 to 100 mg (Milligan et al., 1999). It was therefore deduced that
amounts of 100 µg of 8-PN might have considerable efficacy. The hop-based capsules contained hop
extract that was obtained by an aqueous ethanolic extraction of spent hops following extraction of
hops using SC-C0 2 (food supplement, Menohop R ).
In this three-armed clinical trial 67 patients experiencing mild to severe menopausal discomforts
(more specifically at least 2-5 hot flushes per day corresponding to a score of at least 2 for the item
“hot flushes” on the modified Kupferman-index (KI) during several weeks), were treated with the hop
extract standardized on 100 µg 8-PN (n:20), the hop extract standardized on 250 µg 8-PN (n=20) or
placebo (n=26) over 12 weeks. Assessment was based on the responses obtained using a modified KI
and a patients questionnaire.
All groups, including placebo, showed a significant reduction of the KI both after 6 and after
12 weeks. The hop extract at 100 µg 8-PN was significantly superior to placebo after 6 weeks
(p=0,023) but not after 12 weeks (p=0,086). No dose-response relationship could be established, as the
higher dose (250 µg) was less active than the lower dose both after 6 weeks and after 12 weeks. Still, a
trend for a more rapid decrease of KI was noticed for both active groups as compared to placebo.
In particular, the decrease in hot flushes score (isolated from KI) was found significant for both
treatment groups after 6 weeks (p<0,01) with respect to placebo. Results of the patient’s questionnaire
were consistent with those of the KI, with the most pronounced effects being observed for the 100 µg
treatment. The authors concluded that a daily intake of a hop extract, standardized on 8-PN, exerted
favourable effects on vasomotor symptoms and other menopausal discomforts. Since, however, no
dose-response relationship could be established, further clinical studies are needed to confirm the
present observation.
A randomized, double-blind cross-over study is in preparation (Erkolla et al.).
More recently a study has been published in which a medical device in form of gel was tested intra-
vaginally in women with genital atrophy. This study was designed as an open non-controlled clinical
© EMEA 2008
24/30
study to assess the efficacy and safety of the gel, containing 1% of a mother tincture hop extract
(according to the French Pharmacopoeia X), hyaluronic acid, liposomes formed by cholesterol and
lecithin, all considered to be active ingredients.
The authors conclude that the results showed a marked effect of the tested product on the vaginal
dryness and other menopausal symptoms and that no treatment related adverse events were
complained by the patients (Morali et al., 2006).
Although the results of this study are interesting, it is difficult to draw conclusions, since the hop
extract was not standardized on 8-PN and the relative contribution of different other ingredients cannot
be deduced from the results obtained.
Assessor’s comments
To date only two small placebo-controlled clinical studies have been published using hop extracts, on
20 and 67 patients, respectively. A standardized hop extract (on 8-PN) was used in the latter study
only. The fact that in the three-armed clinical trial no clear dose-response relationship was found is of
concern. The relatively small numbers of patients may have contributed to the absence of a clear
dose–response relationship; a larger follow-up study is required to demonstrate clinical efficacy. The
data available are not sufficient to support a well-established use indication for hops in the treatment
of menopausal symptoms.
II.4.
SAFETY
II. 4.1.
Toxicity
Hops have been used in the brewing industry for centuries, without any known adverse effect to the
health of consumers. Thus, given the history of long-term and present use in humans with no
significant adverse effects, it is considered that hops are safe (Chadwick et al., 2006).
II. 4.1.1. Single dose toxicity
The following toxicological data are given in Hagers Handbuch (1993).
Ethanolic hop extract : LD 50 3,500 mg/kg/bw p.o., 1,200 mg/kg/bw s.c. (mice) :
LD 50 2,700 mg/kg/bw p.o. (rats).
Methylisobutylketone-hop extract : LD 50 2,700 mg/kg/bw p.o. (mice) ; LD 50 415 mg/kg/bw p.o. (rats).
Lupulone : LD 50 525 mg/kg/bw p.o. ; LD 50 1,200 mg/kg/bw s.c. : 600 mg/kg/bw i.m. (mice) :
LD 50 1,800 mg/kg/bw p.o. ; LD 50 330 mg/kg/bw i.m. (rats).
Humulone : LD 50 1,500 mg/kg/bw p.o. ; 600 mg/kg/bw i.m. (rats).
A standardized hydro-alcoholic hop extract enriched in 8-PN, and consisting of 0,22% 8-PN,
0,2% 6-PN, 5,5% xanthohumol and 1,7% isoxanthohumol (Menohop ® ) was tested for acute toxicity
and mutagenicity in rats (Publication in preparation).
The starting dose used in the acute toxicity studies was based upon the dose, which was used in the
clinical trial viz . 2,6 mg/kg/day and calculated via a multiplication factor of 6,66 from man to rats
(Notox ® , 2005). Three female Wistar rats in four groups each were treated with escalating doses of 25,
250, 1000 and 2500 mg hop extract/kg body weight (bw) per day by oral gavage during
2 consecutive days in order to determine the maximum tolerated dose (MTD). Clinical signs, body
weight and food consumption were determined, as well as clinical pathology prior to microscopy and
macroscopy at termination.
Based on the data from this dose escalation phase the dose level for MTD was selected to be
2500 mg/kg/day. Although a real MTD was not reached, this dose level represents a 100-fold expected
human dose, so that the safety margin is expected to be sufficiently high. During the MTD-phase
10 rats (5 males and 5 females) were given one dose of 2500 mg/kg daily for 5 consecutive days by
oral gavage. No mortality or clinical signs were noted during the whole MTD-phase and finally no
treatment related findings in haematology and clinical biochemistry parameters were observed and no
findings were noted after macroscopic and microscopic examination. It was concluded that repeated
© EMEA 2008
25/30
oral administration of 2500 mg/kg/bw in male and female Wistar rats on 5 consecutive days were well
tolerated and that no clear signs indicative of test substance induced toxicity were found.
These results obtained in rats were confirmed recently in humans by Rad et al. (2006), who
investigated the kinetics and systematic endocrine effects of 8-PN after single oral doses.
It was found that single oral doses of up 50-75,0 mg 8-PN (ca. 3500-fold the intended amount of
Menohop ® in clinical studies) were well tolerated by postmenopausal women and that no drug-related
adverse events were recorded.
Recently, toxicity and cell cycle effects of synthetic racemic 8-PN in human cells was studied in
comparison to quercetin and related flavonoids. These investigations were done on different cell lines
viz . the promyeloid leukemia cell line HL 60 and the adherent breast cancer cell line MCF 7. A low
toxicity and weak cytostatic properties of 8-PN and related naringenin derivatives were found
(Tokalov et al., 2004).
Besides the oestrogenic effects, Effenberger et al. (2005) also studied the cytostatic effects of the most
prominent prenylflavonoids present in hops. It was shown that only at high concentration (> 10 -4 M)
prenylflavonoids such as 8-PN, 6-PN, X and IX displayed cytostatic effects on V79 cells (Chinese
hamster fibroblasts).
II. 4.1.2. Sub-acute and chronic toxicity
In their investigations on the oestrogenic effects of 8-PN on bone metabolism, Miyamoto et al. (1998)
found no overt signs of toxicity after administration of 8-PN at a dose of 30 mg/kg/drug
subcutaneously for 2 weeks.
Recently, Christoffel et al. (2006) have published a study on the effects of 8-PN on the
hypothalamo-pituitary-uterine axis in rats after a 3-month treatment. Therefore, a number of
oestrogen-related parameters, in the hypothalamus, pituitary and uterus were chosen to compare the
putative oestrogenic effects of 8-PN with those of 17β-oestradiol. Two doses of 8-PN and
17β-oestradiol-3-benzoate (E 2 B) respectively 6,8 and 68,4 mg/kg/bw of 8-PN and 0,17 and
0,7 mg/kg/bw of E 2 B and their effects were compared on uterine weight, pituitary hormones (LH, FSH
and prolactin) and the expression of oestogen-regulated genes and of oestrogen receptor (ER) α and
ER β in the hypothalamus, pituitary and uterus.
Both doses of E 2 B and the high dose of 8-PN suppressed serum LH and FSH, and stimulated serum
prolactin levels, uterine weight, and progesterone receptor, insulin-like growth factor I and
complement protein C 3 mRNA transcripts.
In the preoptic and the mediobased areas of the hypothalamus, all treatments had negligible effects on
ER α and ER β and gonadotrophin-releasing hormone (GnRH) receptor gene-expression, while ERβ and
GnRH receptor transcripts in the anterior pituitary were reduced under both E 2 B doses and the high
8-PN dose. The mRNA concentrations of the LH α and LH β subunits in the pituitary were suppressed
by E 2 B and 8-PN.
In summary, 8-PN had very similar though milder effects than E 2 B on all tested parameters. The
authors concluded that inhibition of climacteric complaints by E 2 B takes place in the hypothalamus,
where it inhibits the overactive GnRH pulse generator. 8-PN may be used to inhibit climacteric
symptoms effectively. They proposed to carry out human pharmacological studies in order to show
whether the stimulatory effect of 8-PN on the uterus would require the concomitant administration of
progesterone to prevent endometrial over stimulation.
Up till now, no data are available on the long-term toxicology of hop extracts when used by
menopausal women. It should, however, not been neglected that hop extracts are rich in xanthohumol,
which is considered to possess antiproliferative effects on both cancer cell lines of human breast and
ovaries by inhibitory mechanisms at the initiation, promotion and progression stages (Gerhauser et al.,
2002; Gerhauser and Frank, 2005 ; Van Hoecke et al., 2005).
It is being questioned whether dietary and/or environmental exposure to phyto-oestrogens could
impose health risks such as endocrine disruption. In case of hop prenylflavonoids, beer is the main
dietary source. The average beer consumption in the United States was calculated at about 225 mL of
beer per capita per day in 2001 (USDA, 2003). When assumed that this amount was consumed as US
major brand lager/pilsner beers (500-1000 µg prenylflavnoids/l beer), the daily intake of
© EMEA 2008
26/30
prenylflavonoids would be about 0,14 mg (Stevens et al., 1999b). However, the concentrations
detected in beer (and therefore average intake) strongly depend on the brewing process, as strong ales
contain up to 4 mg prenylflavonoids/l. Although xanthohumol (X) is the predominant prenylflavonoid
present in hops (0,1-1% of dry weight), most of it is transformed into isoxanthohumol (IX) by thermal
isomerisation during wort boiling. Therefore IX is the major prenylflavonoid found in beer and is
present in concentrations from 500 µg/l (lager/pilsner) up to 4 mg/l (strong ale) (Stevens et al., 1999b ;
Rong et al., 2000). Similarly, desmethylxanthohumol (DMX) is converted into 8-PN resulting in final
concentrations in beer of up to 100 µg 8-PN/l. But despite the high activity of 8-PN, the total
oestrogenic activity in beer is still 500 to 1000 times lower than the concentration needed for harmful
in vivo activity (~ 100 mg/l) (Milligan et al., 2002). Moreover, many beers are now made using hop
extracts instead of whole hops, giving lower concentrations of 8-PN or no 8-PN at all. Therefore, it is
generally agreed that based on the current knowledge, no detrimental health effects can be attributed to
phyto-oestrogens upon moderate beer consumption (Milligan et al., 1999, 2002 ; Stevens et al., 2004).
Hop extracts have also been used for several years in food supplements without any known problems,
also in combination with other plants such as valerian. The Belgian Health Authorities have set upper-
limits for phyto-oestrogens in dietary supplements. For hop extract, the threshold was set at 400 µg
8-PN/day (for soy isoflavones at 40 mg/day)(Belgian legislation on herbals: MB, 25/02/2005).
The potential side-effects of soy preparations such as the thyroid effect have not been mentioned for
hops, so that every phyto-oestrogen should be considered differently.
II. 4.1.3. Genotoxicity
An overview on the genotoxicity of 8-PN was found in the “Comparative toxicogenomics database”
8-PN has been mentioned therein for interactions with alkaline phosphatase, CYP1A2, ESR1 and
2, GSTA1, IL6, PGR, TFF1 and VWF.
In the Ames mutagenicity test, a hydroethanolic extract of hop strobile showed weakly mutagenic
potential in Salmonella typhimurium strains TA98 and TA100 with or without activation (Göggelmann
et al., 1986). These data were confirmed for TA98 strains only for the hop extract enriched in 8-PN
(Menohop ® ) (Publication in preparation).
The hop extract enriched in 8-PN (Menohop ® ) has been tested in the Salmonella typhimurium reverse
mutation assay with four histidine-requiring strains (TA1535, TA1537, TA100 and TA98) and in the
Escherichia coli reverse mutation asay with a tryptophan requiring strain of Escherichia coli
WP2vvrA. The tests were performed in two independent experiments in the presence and absence of
S9-mix (Aroclor-1254 induced rat liver S9-mix). The concentration range varied from 100 to
5000 µg/plate. The hop extract showed up to 3,6 and 2,8-fold, dose-related increases in the number of
revertant colonies in tester strain TA98 compared to the solvent control, both in the absence and
presence of S9-mix (2 experiments). All other bacterial strains showed negative responses over the
entire dose range. Since hop extracts contain quercetin glycosides (McMurrough 1981; De Cooman et
al., 1998), the slight mutagenic effect in the Salmonella typhimurium TA98 strain which was not
observed in the other strains, might be a false positive result, as it is well-known that quercetin gives
positive results in the Ames test but negative results in other genotoxicity tests. Therefore, it was
decided to carry out a mouse lymphoma test in order to show that the hop extracts are not mutagenic.
Hops extract were tested up to concentrations of 160 and 100 µg/ml in the absence and presence of
8% and 12% (v/v), S9 mix respectively in an in vitro mammalian cell gene mutation test with L5178Y
mouse lymphoma cells. In this test the effects of hop extracts on the induction of forward mutation at
the thymidine-kinase locus (TK-locus) in L5178Y mouse lymphoma cells were determined. It was
shown that hop extract was not mutagenic in the TK mutation test system under the experimental
conditions and consequently does not cause a point mutation or chromosomal aberrations. Since the
suppressed mutagenic effect in the Ames test was not confirmed in the same mutation test in
mammalian cells, the overall conclusion can be drawn that hop extract is not mutagenic and that
therefore carcinogenicity studies are not needed (Guideline EMEA/HMPC/32116/2005).
© EMEA 2008
27/30
Assessor’s comments
To date, adequate genotoxity studies have not been carried out for the aqueous or hydroalcoholic
extracts of hops included in the monograph on traditional use.
II. 4.1.4. Carcinogenicity
To date, no data on carcinogenicity studies on hop strobiles or its preparations are available.
II. 4.1.5. Reproduction and developmental toxicity
No preclinical data on reproductive and developmental toxicity of hop strobiles or its preparations are
available.
Kumai and Okamoto. (1984) investigated purified fractions of hop strobile extract for reproductive
toxicity in female rats primed with 25 IU of pregnant mare’s serum gonadotrophin.
Fractions F 1 (20 mg/rat) and F 2 (50 mg/rat), administered subcutaneously twice daily for 3 days, did
not induce any change in uterine weights but ovarian weights decreased significantly (p<0,05) by
25,7% and 24,9% respectively. Under the same conditions, two further fractions (4 mg/rat) purified
from F 1 produced significant decreases (p<0,01) of 42,0% and 33,1% in ovarian weight.
Caujolle et al. (1969) showed that an alcoholic extract of hop strobile (1 g of dried drug in 10 ml of
70% ethanol) inhibited concentrations of rat uterus with an ED 50 equivalent to 31x10 -6 of hop strobile
per ml.
In view of this in vitro antispasmodic effect on the uterus and lack of toxicity data, the use of hops and
its preparations should be avoided during pregnancy and lactation. It is clear that the extracts enriched
in 8-PN are intended for menopausal complaints and consequently should not be used by fertile
women. This is also the case for the indications of hop preparations as sedative.
II. 4.1.6. Local tolerance
Data on local tolerance for hop strobiles and its preparations are not available. Nevertheless, allergic
reactions have been reported for hops, although only following external contact with the herb and the
oil.
II. 4.2.
Side effects
Based on traditional use and available studies, there have been no serious side effects reported with
hops. Drowsiness or sedation may occur. Use caution if you are driving or operating heavy machinery.
In animal studies, hops have increased stomach acid, but there is no available research in humans in
that area. Based upon animal studies, hops may increase blood sugar levels in diabetic patients, but
may lower blood sugar in nondiabetic patients. Consequently, the effects of hops on blood sugar levels
are unclear (Anonymous, 2003a). It should be noted that oral intake of isohumulones, present in hops
and its preparations, is reported to improve insulin sensitivity in patients suffering from diabetes type
2. Consequently, it might be expected that the blood sugar level in such patients would decrease after
oral intake of hops.
Allergic reactions from hops have been reported, particularly in hop harvesters. The contact dermatitis
that has occurred in hop-pickers is attributed to myrcene, present in fresh oil but readily oxidized
(Mitchell and Rook, 1979). Additionally, a mechanical dermatosis has been attributed to the rough
hairs on the stem and secretions of the yellow glandular hairs on hops (Estrada et al., 2002).
Respiratory allergy caused by handling of hop cones have been documented (Newark, 1978), a
subsequent patch test using dried, crushed flowerheads proved negative. Positive patch test reactions
have been documented for fresh hop oil, humulone and lupulone.
Both of these allergic reactions are not likely to occur when using the hop extract, since allergens are
supposed to be removed (Estrada et al., 2002).
Moreover, no clinical cases of allergy or anaphylaxis resulting from the therapeutic use of hops have
been published (Anonymous, 2003a).
© EMEA 2008
28/30
Since hop extracts might contain small amounts of oestrogens, it is not known as yet what the effects
would be of use for viz . more than three months, on conditions such as breast, uterine, cervical or
prostate cancer or endometriosis. However, it should be noted that oestrogens such as 8-PN are only
present in hop preparations in significant amounts when old hops are used as starting material and
special enrichment procedures are employed for the hop preparations.
It should also be noted that hop preparations may contain antiproliferative compounds such as
humulones, xanthohumol and isoxanthohumol.
Finally, some hop preparations contain high levels of alcohol and should be avoided during pregnancy.
II. 4.3.
Contra-indications, warnings
It has been suggested that hops should not be taken by individuals suffering from depressive illness, as
the sedative effect may accentuate symptoms (Anonymous, Harvard Medical School, 2003b). The
sedative action may potentiate the effects of existing sedative therapy and alcohol.
Allergic reactions have been reported for hops, although only following external contact with the herb
and oil.
Concerning pregnancy and lactation, in vitro antispasmodic effect on the uterus has been documented.
In view of this and lack of toxicity data, the use of hops during pregnancy and lactation should be
avoided.
II. 4.4.
Interactions (see also under II.2.3)
Interactions with drugs, dietary supplements and other herbs have not been thoroughly studied. From
the many publications on hops and its constituents, it is theoretically possible that hops may increase
the drowsiness caused by sedative drugs and alcohol.
Hops may also affect blood sugar levels but no clinical data are available to support this effect.
II. 4.5.
Overdoses
Not known.
Assessor’s comments
In view of its long term use and present use in humans hops is considered to be non-toxic and safe
with no significant adverse effects.
The experimental toxicological data on hop preparations are rather limited and incomplete but as a
whole show low toxicity.
Substantial toxicological experiments have been carried out on a standardized hydro-alcoholic extract,
enriched in 8-PN, and consisting of 0,22% 8-PN, 0,2% 6-PN, 5,5% xanthohumol and
1,7% isoxanthohumol (Menohop ® ). The single-dose toxicity studies as well as the genotoxicity studies
showed the extract to be safe in all concentrations tested i.e. no adverse effects have been seen in
doses up 1000-fold the intended dose for clinical studies.
Investigations on the toxicity of the oestrogenic active ingredient of hop extracts viz . 8-PN were
carried out during 2 weeks and also three months, indicating that 8-PN had very similar but milder
effects than 17β-oestradiol on all tested oestrogenic parameters. Although, today no data are available
on the long-term toxicology of hop extract when used by menopausal women, it should nevertheless,
be stressed that hop extracts also contain xanthohumol, which is considered to possess antiproliferative
effects in several cancer cell lines.
Hop extracts such as Menohop ® have been used for several years in food supplements without any
known adverse effects. The Belgian authorities have set upper-limits for phyto-oestrogens in dietary
supplements. For hop extracts and soy-isoflavones the threshold was set at 400 µg 8-PN/day and
40 mg/day isoflavonoids, expressed as the glycoside of the main isoflavonoid respectively. It can also
be concluded from the many publications that no detrimental effects can be attributed to phyto-
oestrogens upon moderate beer consumption.
In conclusion, the safety assessment of hops and hop preparations is mainly based upon the many
years of experience from the extensive medicinal use in man, which indicate hop preparations to be
safe pharmaceutical agents.
© EMEA 2008
29/30
II.5.
ASSESSOR’S OVERALL CONCLUSION
The sedative effect of hops and preparations thereof has long been recognised empirically, so that a
period of at least 30 years of traditional medicinal use is easily fulfilled for this indication.
Its use has been made plausible by many in vitro and in vivo pharmacological studies. Recently, it has
been suggested that besides the acids and the essential oil other constituents such as xanthohumol may
play an important role in the sedative effect of hop preparations.
No clinical studies have been conducted to date with hop extracts alone as active drug for insomnia,
but at least seven placebo- or reference-controlled clinical studies have been carried out with fixed
combinations of hop extracts and valerian root extracts. Whether hop extracts act as mild sedative
independently, as a synergist, or not at all remains to be determined.
Although several hop-containing cosmetic preparations have been patented since 1961 for the external
treatment of various gynaecological disorders, almost no traditional oestrogenic formulations of hops
have been found in literature until the discovery of the oestrogenic principle in hops in 1988.
Consequently, the traditional medicinal use of hops as a phyto-oestrogenic preparation does not fulfil
the requirement for at least 30 years on the market. Since 8-PN was recognized as the phyto-
oestrogenic principle in hops, many modern hop extracts enriched and standardised upon the active
constituent have been prepared and pharmacologically investigated. It has been shown that 8-PN
behaves like 17β-oestradiol, but has much milder effects on all oestrogenic parameters tested than the
parent compound.
To date, only two small placebo-controlled clinical studies have been conducted with such extracts to
investigate menopausal discomforts. These studies, however, should be considered only as pilot
studies with results, encouraging enough to justify a full-scale trial. The experimental toxicological
data, however, obtained with these extracts, have shown the extracts to be devoid of genotoxicity and
adverse effects in concentrations up 1000-fold the dose used for the clinical studies.
Consequently, the well-established use as phyto-estrogenic drug for these hop extracts can not be
granted as yet.
In conclusion, single hop preparations can be accepted as traditional herbal medicinal products for
their sedative effects. Fixed combination products consisting of dry extracts of hop strobiles with other
sedative plants will be considered in separate monographs.
The data available on the oestrogenic properties of hop preparations are not sufficient to support a
marketing authorization.
III
ANNEXES
III.1
Community herbal monograph
III.2
Literature references
© EMEA 2008
30/30


Source: European Medicines Agency



- Please bookmark this page (add it to your favorites).
- If you wish to link to this page, you can do so by referring to the URL address below this line.



https://theodora.com/drugs/eu/lupuli_flos_herbal.html

Copyright © 1995-2021 ITA all rights reserved.