Regulatory status in selected countries

Bilberry is official in the European Pharmacopoeia (2006).

Bilberry fruit is covered by a positive Commission E monograph and has the following applications:

• Non-specific, acute diarrhoea

• Topical treatment of mild inflammation of the mucous membranes of the mouth and throat.

Bilberry does not have GRAS status. However, it is freely available as a ‘dietary supplement’ in the USA under DSHEA legislation (1994 Dietary Supplement Health and Education Act).

Bilberry is not included in Part 4 of Schedule 4 of the Therapeutic Goods Act Regulations of Australia and is freely available for sale.

References

1. Grieve M. A Modern Herbal. New York: Dover Publications, 1971. pp. 99–100

2. German Federal Minister of Justice (ed). German Commission E for Human Medicine Monograph, Bundes-Anzeiger (German Federal Gazette) no. 76, dated 23.04.1987, no. 50, dated 13.03.1990.

3. Smeh NJ. Creating Your Own Cosmetics –Naturally. Garrisonville: Alliance, 1995. pp. 81, 142

4. Martinelli EM, Scilingo A, Pifferi G. Anal Chim Acta. 1992;259(1):109–113.

5. Yue X, Xu Z. J Food Sci. 2008;73(6):C494–4499.

6. Nalliah RE, Phillips JS, Gaier AJ, et al. Int J Food Sci Nutr. 2009;60(suppl 1):209–219.

7. Chiej R. The Macdonald Encyclopedia of Medicinal Plants. London: Macdonald, 1984. Entry No. 321

8. Penman KG, Halstead CW, Matthias A, et al. J Agric Food Chem. 2006;54(19):7378–7382.

9. Wagner H, Bladt S. Plant Drug Analysis: A Thin Layer Chromatography Atlas, 2nd ed. Berlin: Springer-Verlag, 1996. p. 282

10. Bisset NG, Wichtl M, eds. Herbal Drugs and Phytopharmaceuticals, German edition, Stuttgart: Medpharm Scientific Publishers, 1994.

11. Brenneisen R, Steinegger E. Pharm Acta Helv. 1981;56(7):180–185.

12. Rimando AM, Kalt W, Magee JB, et al. J Agric Food Chem. 2004;52(15):4713–4719.

13. Lyons MM, Yu C, Toma RB, et al. J Agric Food Chem. 2003;51(20):5867–5870.

14. Erlund I, Marniemi J, Hakala P, et al. Eur J Clin Nutr. 2003;57(1):37–42.

15. Akerstrom A, Jaakola L, Bang U, et al. J Agric Food Chem. 2010;58(22):11939–11945.

16. Terrasse J, Moinade S. Presse Med. 1964;72:397–400.

17. Demure G. PhD Thesis in Medicine: Etude expèrimentale et clinique d’un nouveau facteur vitaminique P: les Anthocyanosides. France 1964, Clermont.

18. Bombardelli E. Therapia Angiol. 1976;5:177.

19. Bell DR, Gochenaur K. J Appl Physiol. 2006;100(4):1164–1170.

20. Bertuglia S, Malandrino S, Colantuoni A. Pharmacol Res. 1995;31(3–4):183–187.

21. Colantuoni A, Bertuglia S, Magistretti MJ, et al. Arzneimittelforschung. 1991;41(9):905–909.

22. Detre Z, Jellinek H, Miskulin M, et al. Clin Physiol Biochem. 1986;4:143–149.

23. Lietti A, Cristoni A, Picci M. Arzneimittelforschung. 1976;26(1):829–835.

24. Wegmann R, Maeda P, Tronche P, et al. Ann Histochim. 1969;14:237–256.

25. Cluzel C, Bastide P, Wegman R, et al. Biochem Pharmacol. 1970;19:2295–2302.

26. Fursova A, Gesarevich OG, Gonchar AM, et al. Adv Gerontol. 2005;16:76–79.

27. Song J, Li Y, Ge J, et al. Phytother Res. 2010;24(4):520–524.

28. Meunier MT, Duroux E, Bastide P. Plantes Méd Phytothér. 1989;23(4):267–274.

29. Milbury PE, Graf B, Curran-Celentano J, et al. Invest Ophthalmol Vis Sci. 2007;48(5):2343–2349.

30. Rahman MM, Ichiyanagi T, Komiyama T, et al. Free Radic Res. 2006;40(9):993–1002.

31. Maatta-Riihinen KR, Kahkonen MP, Torronen AR, et al. J Agric Food Chem. 2005;53(22):8485–8491.

32. Maridonneau I, Braquet P, Garay RP, Farkas L, Gabor M, Kallay F, et al, eds. Flavonoids and Bioflavonoids, 1981, Amsterdam, Elsevier, 1982:427–436.

33. Mavelli I, Rossi L, Autuori F, et al, Cohen G, Greenwald RA, eds. Oxy Radicals Their Scavenger Systems. Proceedings International Conference Superoxide Dismutase, 3rd ed., New York, Elsevier, 1982:326–329.

34. Francesca Rasetti M, Caruso D, Galli G, et al. Phytomedicine. 1996/1997;3(4):335–338.

35. Laplaud PM, Lelubre A, Chapman MJ. Fundam Clin Pharmacol. 1997;11(1):35–40.

36. Jakesevic M, Aaby K, Borge GI, et al. BMC Complement Altern Med. 2011;11:8.

37. Joseph JA, Shukitt-Hale B, Denisova NA, et al. J Neurosci. 1999;19(18):8114–8121.

38. Galli RL, Shukitt-Hale B, Youdim KA, et al. Ann N Y Acad Sci. 2002;959:128–132.

39. Andres-Lacueva C, Shukitt-Hale B, Galli RL, et al. Nutr Neurosci. 2005;8(2):111–120.

40. de Rivera C, Shukitt-Hale B, Joseph JA, et al. Neurobiol Aging. 2006;27(7):1035–1044.

41. Ronziere MC, Herbage D, Garrone R, et al. Biochem Pharmacol. 1981;30:1771–1776.

42. Monboisse JC, Braquet P, Randoux A, et al. Biochem Pharmacol. 1983;32:53–58.

43. Mian E, Curri SB, Leitti A, et al. Minerva Med. 1977;68(52):3565–3581.

44. Piovella C, Curri BS, Piovella M, et al. Therapia Angiol. 1979;35:119.

45. Piovella F, Ricetti MM, Almasio P, et al. Minerva Angiol. 1981;6:135.

46. Curri SB, Leitti A, Bombardelli E. Giorn Minerva Derm. 1976;111:509.

47. Cristoni A, Magistretti MJ. Il Farmaco Ed Pr. 1987;42(2):29–43.

48. Persson IA, Persson K, Andersson RG. J Agric Food Chem. 2009;57(11):4624–4629.

49. Mauray A, Milenkovic D, Besson C, et al. J Agric Food Chem. 2009;57(23):11106–11111.

50. Mauray A, Felgines C, Morand C, et al. Nutr Metab Cardiovasc Dis, 2010. [Epub ahead of print]

51. Ziberna L, Lunder M, Moze S, et al. Cardiovasc Toxicol. 2010;10(4):283–294.

52. Morazzoni P, Magistretti MJ. Fitoterapia. 1990;61(1):13–21.

53. Pulliero G, Montin S, Bettini V, et al. Fitoterapia. 1989;60(1):69–75.

54. Lala G, Malik M, Zhao C, et al. Nutr Cancer. 2006;54(1):84–93.

55. Mutanen M, Pajari AM, Paivarinta E, et al. Asia Pac J Clin Nutr. 2008;17(suppl 1):123–125.

56. Magistretti MJ, Conti M, Cristoni A. Arzneimittelforschung. 1988;38(5):686–690.

57. Cristoni A, Malandrino S, Magistretti MJ. Arzneimittelforschung. 1989;39(5):590–592.

58. Domitrovic R, Jakovac H. Food Chem Toxicol. 2011;49(4):848–854.

59. Bao L, Abe K, Tsang P, et al. Fitoterapia. 2010;81(8):1094–1101.

60. Bao L, Yao XS, Yau CC, et al. J Agric Food Chem. 2008;56(17):7803–7807.

61. Puupponen-Pimia R, Nohynek L, Alakomi HL, et al. Biofactors. 2005;23(4):243–251.

62. Choi EH, Park JH, Kim MK, et al. Biofactors. 2010;36(4):319–327.

63. Bao L, Yao XS, Tsi D, et al. J Agric Food Chem. 2008;56(2):420–425.

64. Takikawa M, Inoue S, Horia F, et al. J Nutr. 2010;140(3):527–533.

65. Ichiyanagi T, Shida Y, Rahman MM, et al. J Agric Food Chem. 2006;54(18):6578–6587.

66. He J, Magnuson BA, Lala G, et al. Nutr Cancer. 2006;54(1):3–12.

67. Morazzoni P, Livio S, Scilingo A, et al. Arzneimittelforschung. 1991;41(1):128–131.

68. Sakakibara H, Ogawa T, Koyanagi A, et al. J Agric Food Chem. 2009;57(17):7681–7686.

69. Bitsch I, Janssen M, Netzel M, et al. Int J Clin Pharmacol Ther. 2004;42(5):293–300.

70. Kay CD, Mazza G, Holub BJ. J Nutr. 2005;135(11):2582–2588.

71. Nurmi T, Mursu J, Neinonen M, et al. J Agric Food Chem. 2009;57(6):2274–2281.

72. Cai H, Thomasset SC, Berry DP, et al. Biomed Chromatogr. 2011;25(6):660–663.

73. Ghiringhelli C, Gregoratti F, Marastoni F. Minerva Cardioang. 1977;26:255–276.

74. Corsi C, Pollastri M, Tesi C, et al. Fitoterapia. 1985;56(suppl 1):23.

75. Grismondi GL. Minerva Gin. 1981;33(2–3):221–230.

76. Teglio L, Tronconi R, Mazzanti C, et al. Quad Clin Ostet Ginecol. 1987;42(3):221–231.

77. Berta V, Zucchi C. Fitoterapia. 1988;59(suppl 1):27.

78. Allegra C, Pollari G, Criscuolo A. Minerva Angiol. 1982;7:39–44.

79. Gatta L. Fitoterapia. 1988;59(suppl 1):19.

80. Carmignani G. Lotta Contro La Tuberce Malattie Polm Soc. 1983;53:732.

81. Orsucci PL, Rossi M, Sabbatini G, et al. Clin Ocul. 1983;4:377.

82. Mattioli L, Dallari S, Galetti R. Fitoterapia. 1988;59(suppl 1):41.

83. Repossi P, Malagola R, de Cadihac C. Ann Ottal Clin Ocul. 1987;113(4):357–361.

84. Perossini M, Guidi G, Chiellini S, et al. Ann Ottal Clin Ocul. 1987;113(12):1173–1190.

85. Morazzoni P, Bombardelli E. Fitoterapia. 1996;67(1):3–29.

86. Gandolfo E. Boll Ocul. 1990;69(1):57–72.

87. Fiorini G, Biancacci A, Graziano FM. Ann Ottal Clin Ocul. 1965;91(6):371–386.

88. Zavarise G. Ann Ottalmol Clin Ocul. 1968;94(2):209–214.

89. Virno M, Recori Giraldi J, Auriemma L. Boll Ocul. 1986;65(4):789–796.

90. Caselli L. Arch Med Intern. 1985;37:29–35.

91. Jayle GE, Aubert L. Therapie. 1964;19:171–185.

92. Jayle GE, Aubury M, Gavini G, et al. Ann Ocul. 1965;198:556–562.

93. Vannini L, Samuelly R, Coffano M, et al. Boll Ocul. 1986;65(suppl 6):569.

94. Contestabile MT, Appolloni R, Suppressa F, et al. Boll Ocul. 1991;70(6):1157–1169.

95. Muth ER, Laurent JM, Jasper P. Altern Med Rev. 2000;5(2):164–173.

96. Canter PH, Ernst E. Surv Ophthalmol. 2004;49(1):38–50.

97. Pezzangora V, Barina R, De Stefani R, et al. Gaz Med Ital. 1984;143(6):405–409.

98. Oliva E, Nicastro A, Sorcini A, et al. Aggior Med Chir. 1990;8:1.

99. Massenzo D, Gentile A, Mosciaro O. Riv Ital Otl Aud Fon. 1992;12(1):65–68.

100. Colombo D, Vescovini R. Giorn Ital Ost Gin. 1985;7(12):1033–1038.

101. Karlsen A, Paur I, Bohn SK, et al. Eur J Nutr. 2010;49(6):345–355.

102. Thomasset S, Berry DP, Cai H, et al. Cancer Prev Res. 2009;2(7):625–633.

103. Mills, S. Bone, K., St Louis, Elsevier Churchill Livingstone, 2005.

104. Eandi M. Cited in Morazzoni P, Bombardelli E. Fitoterapia 1996;67(1):3–29.

105. Pourrat H, Bastide P, Dorier P, et al. Chim Ther. 1967;2:33–38.

106. Schimmer O, Kruger A, Paulini H, et al. Pharmazie. 1994;49(6):448–451.

107. American Herbal Pharmacopoeia. Bilberry Fruit – Vaccinium myrtillus L.: Standards of Analysis, Quality Control and Therapeutics. Santa Cruz: American Herbal Pharmacopoeia, 2001.

108. Baisi F. Report on clinical trial of bilberry anthocyanosides in the treatment of venous insufficiency in pregnancy and of post-partum hemorrhoids [in Italian]. Livorno Hospital, 1987, Obstetrics and Gynecology Operating Unit.

109. Baudon J, Bruhat M, Plane C, et al. Lyon Mediterr Med. 1969:46.

110. Blumenthal M, ed. The Complete German Commission E Monographs: Therapeutic Guide to Herbal Medicines. Austin: American Botanical Council, 1998.

111. Eandi M. Cited in Morazzoni P, Bombardelli E. Fitoterapia 1996;67(1): 3–29.

Black cohosh

(Actaea racemosa var. racemosa L.)

Synonyms

Cimicifuga racemosa (L.) Nutt. (botanical synonym), bugbane, black snakeroot (Engl), Cimicifugae rhizoma (Lat), schwarzes Wanzenkraut, Cimicifugawurzelstock (Ger), cimicaire, actée à grappes (Fr), sølvlys (Dan).

What is it?

Black cohosh is still widely referred to by its former botanical name, Cimicifuga racemosa. However, a taxonomic revision published in 1998 reclassified the genus Actaea to include Cimicifuga.1 The common names of black snakeroot and rattle snakeroot refer to its former use in North America, where it is native, to treat snakebite including that of rattlesnake. The old generic name Cimicifuga comes from the Latin ‘to chase insects away’ and reflects on a reputed use of the European species. Black cohosh rhizome is a popular treatment for menopausal symptoms, and proprietary medicines based on black cohosh are registered in Germany. The part used therapeutically consists of the fresh or dried rhizome with attached roots.2,3

Effects

Suppresses luteinising hormone (LH), allays inflammation, promotes fertility, improves bone density.

Traditional view

Black cohosh, a favourite of the Eclectic physicians, was used for myalgia, neuralgia (not of spinal origin), chorea, female reproductive tract disorders (amenorrhoea, dysmenorrhoea, ovarian pain and menorrhagia) and rheumatic conditions (arthralgia, muscular rheumatism). Other conditions treated included whooping cough, tinnitus and mastitis.4,5 Black cohosh is also used to treat premenstrual syndrome and secondary amenorrhoea in Germany.3,6

Summary actions

Hormone modulating, antirheumatic, spasmolytic.

Can be used for

Indications supported by clinical trials

Treatment of menopausal symptoms; to promote fertility.

Traditional therapeutic uses

Used particularly for arthritis and rheumatism, neuralgia, sciatica, menstruation disorders (amenorrhoea, dysmenorrhoea, menorrhagia, ovarian pain), respiratory tract disorders, (whooping cough, asthma), tinnitus.

May also be used for

Extrapolations from pharmacological studies

Adjunct in the treatment of conditions requiring reduction in LH levels (e.g. infertility, miscarriage, cyst formation, ovarian tumorigenesis and polycystic ovary syndrome). Potentially as an aid in the treatment of osteoporosis.

Preparations

Dried or fresh rhizome for decoction, liquid extract or solid dose forms for internal use.

Dosage

Typical adult dosage ranges used by Western herbalists are:

• 0.9 to 6 g/day of dried root and rhizome or by decoction

• 0.9 to 6 mL/day of a 1:1 liquid extract

• 1.5 to 3 mL/day of a 1:2 liquid extract, 3.0 to 7.5 mL/day of the 1:5 tincture, or equivalent doses in tablet or capsule form.

However, herbalists typically now recommend doses at the lower end of these ranges. In addition doses at the higher end have been linked to adverse reactions such as headaches (see Overdosage section).

Typical adult dosage ranges used in most clinical trials of German products:2

• Standardised extract equivalent to 40 to 140 mg/day of dried root and rhizome and containing approximately 3% triterpene glycosides.

Duration of use

May be taken long term within the recommended dosage, although the Commission E recommends not for more than 6 months.

Summary assessment of safety

Generally no adverse effects are expected from ingestion of black cohosh when used at the lower end of the recommended dosage. Dosage at the higher end may cause headaches. Black cohosh has been associated with rare cases of liver damage, although this link is disputed. It is not recommended during pregnancy except for assisting childbirth, although there is no strong indication of harm.

Technical data

Botany

Black cohosh is a member of the Ranunculaceae (buttercup) family and grows to an average height of 150 cm.7 It produces white blossoms in slender, feathery drooping racemes and a dry fruit containing numerous seeds. The leaves have three-pointed, trilobate leaflets. The fleshy, dark brown/black rhizome is a creeping underground stem, from which follow dark brown roots.2,8

Adulteration

Other Actaea/Cimicifuga species, particularly C. americana, have been unintentionally mixed with black cohosh due to the similarity in aboveground appearance. Occasionally black cohosh is adulterated with the underground portions of baneberry (Actaea pachypoda and A. rubra).2 In August 1998, Australian manufacturers were alerted to the possibility of substitution of black cohosh by other Cimicifuga spp. In May 2001 the Therapeutic Goods Administration Laboratories advised that 35% of the Australian products they tested indicated the presence of species other than Cimicifuga racemosa. Manufacturers were advised to verify their raw materials to ensure the correct Cimicifuga species is used. Other medicinal Cimicifuga species were implicated such as C. foetida, C. dahurica, C. heracleifolia and C. simplex.9 See also the findings of Health Canada regarding adulteration (cited under Side effects in the safety section later in this monograph).

Key constituents

Constituents of black cohosh include at least 20 triterpene glycosides (saponins) of the cycloartane type, including actein, 23-epi-26-deoxyactein, cimiracemoside A and cimicifugoside.

Other constituents include aromatic acids (including ferulic, isoferulic and acyl caffeic acids), resins (cimicifugin), tannins and fatty acids.10,11 Despite earlier reports, the herb does not contain significant quantities of isoflavones (see below).

image

Pharmacodynamics

The pharmacology of black cohosh is poorly understood. The question of whether the herb has oestrogenic activity in particular remains unresolved and conflicting results from many studies on this topic are available. While the majority of more recent studies indicate that black cohosh extracts do not contain compounds that are potent agonists at either of the two known oestrogen receptors, black cohosh appears to possess several pharmacological effects that are consistent with oestrogen-like activity, including lowering of LH levels (by injection) and protection against menopausal bone loss. Meanwhile, other potential mechanisms of action have emerged such as serotonergic, dopaminergic and opioid receptor activity, all of which could potentially play a part in the ameliorating effects of black cohosh on menopausal symptoms. In addition, anti-inflammatory activity has been demonstrated in vitro, supporting some traditional uses of the herb. Any oestrogenic effects from the herb are likely to be complex and mild.

Hormonal activity

In early research, prolonged injections of black cohosh extract in rats and mice increased the weight of the uterus and established menstrual cycles in juvenile and climacteric animals.12 Black cohosh extract demonstrated a selective reduction of serum LH in ovariectomised rats. FSH (follicle-stimulating hormone) and prolactin levels were unchanged. Intraperitoneal administration of black cohosh extract (24 mg dried extract per day) resulted in significant inhibition of LH secretion after the third day. Fractionated extraction of black cohosh and subsequent testing with three fractions indicated that the LH-suppressive substances resided in the dichloromethane extract. The intact glycosidal components of this extract were more active with regard to LH suppression than the aglycone form. Oral administration of the non-hydrolysed extract demonstrated a significant inhibition of LH, although much lower than that achieved by injection.13 (This does not necessarily mean the aglycone is inactive, as the glycoside may provide enhanced bioavailability with cleavage of the sugars yielding an active aglycone.)14

A subsequent study by the same group found serum levels of LH in ovariectomised rats were reduced after intraperitoneal administration of a trichloromethane fraction of a black cohosh methanolic extract, whereas the ethanolic fraction of the extract did not affect LH levels. The trichloromethane fraction also demonstrated an ability to bind to oestrogen receptors in vitro. This fraction was further separated into three subfractions, two of which suppressed LH secretion in vivo and displaced oestrogen in vitro. This indicates that at least two groups of compounds may be responsible for the endocrine activity of black cohosh.15 One active compound identified was the isoflavone formononetin. However, five more recent studies have failed to detect formononetin in black cohosh,2 a fact that casts some doubt on the extract used in the study.

In other early research, water and chloroform fractions prepared from a black cohosh methanol extract were tested in ovariectomised rats by intraperitoneal injection over several days (chronic administration). The chloroform fraction demonstrated a strong LH-suppressing effect after 3 days; the water fraction was inactive. Further fractionation of the chloroform extract led to the conclusion that the LH-suppressive effect of black cohosh extract was caused by at least three different synergistically acting compounds.16

Another later study in ovariectomised rats showed that a dichloromethane (lipophilic) extract (60 mg subcutaneously for 7 days) produced effects similar to those of oestradiol on LH levels and other biomarkers influenced by oestrogen receptors.17

Two commercial black cohosh extracts were tested for their ability to compete with oestradiol for antigen binding sites on an antibody (IgG) directed against oestradiol (radioimmunoassay). Both extracts ran parallel with the displacement curve obtained with oestradiol, which supports the presence of oestrogenic compounds in black cohosh.18 However, no oestrogenic activity was found after oral or subcutaneous administration of black cohosh extract (6, 60, 600 mg/kg) to groups of immature mice and ovariectomised rats respectively.19

A 50% aqueous ethanolic extract of black cohosh did not bind to oestrogen or progesterone receptors of human breast cancer cell lines (MCF7 and T47D) in vitro (100 μg/mL) and did not affect cell proliferation.20 Similarly, a methanolic extract (200 μg/mL) did not bind to human recombinant alpha and beta oestrogen receptors in vitro.21 Gene expression studies in MCF7 cells found that a lipophilic black cohosh extract had no oestrogenic activity, but instead exerted anti-proliferative and pro-apoptotic effects at the transcriptional level.22 (See also below under Effects on human breast cancer cells.)

Only the lipophilic subfraction of a dry, hydroethanolic extract of black cohosh was able to activate the human alpha oestrogen receptor, but neither the total extract nor the lipophilic sub-fraction showed any in vivo uterotrophic effects in 21-day-old rats.23

One hundred and ten women experiencing menopausal symptoms, who had received no hormone replacement therapy (HRT) for at least 6 months, received either a standardised black cohosh extract or placebo. After 8 weeks of treatment, LH levels were significantly reduced (p<0.05) in the black cohosh group, but FSH was unchanged.16

Effects on menopausal bone loss

Several studies have indicated that black cohosh might protect against menopausal bone loss (see also under Clinical trials).

An in vitro study investigated the effects of an ethanolic black cohosh extract on bone nodule formation in mouse pre-osteoblast cells.24 The extract did not stimulate osteoblast proliferation, but significantly increased bone nodule formation (500 ng/mL), an effect that was shown to result from enhanced gene expression of osteocalcin and Runx2 (a transcription factor involved in osteoblast differentiation). Interestingly, co-treatment with a selective oestrogen receptor antagonist abolished the effects on gene expression, demonstrating the involvement of an oestrogen receptor-dependent mechanism.

An experimental model was designed to understand the mechanism of action of black cohosh on bone tissue and to compare its effects with oestrogen and testosterone.25 RANK (receptor activator of nuclear factor-kappaB, NF-kappaB) and its ligand RANKL largely regulate osteoclast activity and hence bone breakdown. Crosslaps (or Ratlaps in rats), specifically the metabolic products of bone-specific collagen-1 alpha1, are markers of such bone degradation. When black cohosh, oestradiol and testosterone were given to castrated rats of both sexes, oestradiol and black cohosh reduced levels of RANKL and Ratlaps, the latter parameter only in female rats. The authors suggested that the bone sparing effect of black cohosh is therefore partly mediated by inhibition of RANKL. However, the receptors involved in mediating this effect are not thought to be oestrogen receptors.

In a 35-day study of metaphyseal fracture healing in ovariectomised rats with early stage osteoporosis, the effects of a black cohosh supplemented diet (dried aqueous ethanolic extract, average 24.9 mg/day) was compared with oestrogen treatment (average 0.03 mg/kg/day).26 A high rate of metaphyseal callus formation was observed in the animals receiving black cohosh, but oestrogen improved fracture healing more, with the authors commenting that the 5-week treatment period was possibly too short for black cohosh to demonstrate its full potential.

The triterpenoid glycoside 25-acetylcimigenol xylopyranoside isolated from black cohosh was shown in vitro to potently block osteoclastogenesis induced by tumour necrosis factor-alpha (TNF-alpha) and a related cytokine. The compound was also found to reduce bone loss induced by TNF-alpha in vivo.27

Effects on human breast cancer cells

Because of the potential oestrogen-like action of black cohosh, the issue of its safety in oestrogen-dependent tumours is of significant clinical interest, and a number of studies have been undertaken in an attempt to clarify this issue.

Unlike oestradiol, black cohosh extract did not stimulate growth of mammary tumour cells in vitro. In fact, a dosage of 2.5 mg/mL led to a strong inhibition of proliferation.28 The simultaneous incubation of tumour cells with tamoxifen (anticarcinogenic agent, oestrogen antagonist) and black cohosh displayed a much stronger inhibition of growth than for either substance alone.29 (Oestrogen is contraindicated in patients with oestrogen receptor-positive breast carcinoma, since it promotes the growth of the tumour cells.)

Several other studies have demonstrated that black cohosh does not promote the growth of human breast cancer cells in vitro. A 50% aqueous ethanolic extract (100 μg/mL) did not promote cell proliferation of T47D cells.20 In another study, a methanol-water extract was fractionated. A fraction rich in triterpenoid glycosides inhibited the growth of two human breast cancer cell lines (IC50 values of 10 to 20 μg/mL).30 The same fraction also caused cell cycle arrest at G1 (30 μg/m and G2 (60 μg/mL), suggesting that different compounds are involved in causing cell cycle arrest. The triterpene glycosides actein, 23-epi-26-deoxyactein and cimiracemoside A were shown to be involved in these effects.

In a subsequent gene expression study conducted by the same group, actein was shown to activate transcription factors that enhance apoptosis.31 It also repressed cell cycle genes and acted synergistically with two chemotherapy agents, doxorubicin and 5-fluorouracil, in causing growth inhibition of the breast cancer cell line MDA-mB 453.32 The same study also showed that actein enhanced the induction of apoptosis by paclitaxel, 5-fluorouracil and doxorubicin.

A review of the safety and efficacy of black cohosh for cancer patients assessed data from clinical (n=5) and pre-clinical (n=21) studies as well as case reports.33 The five clinical studies all involved women with breast cancer or a history of the disease. None of the adverse effects reported from these trials (including one case of breast cancer recurrence) was linked to the black cohosh treatment. The authors of the review concluded that black cohosh does not have phyto-oestrogenic activity, appears to be safe for breast cancer patients and may potentially be protective against breast cancer, as it appears to inhibit the growth of tumour cells in vitro (See also under Clinical trials and Toxicology.)

Effects on prostate cancer cells

In vitro studies have demonstrated that black cohosh can inhibit proliferation of prostate cancer cells. In one study, the cell growth inhibitory effects of an isopropanolic extract of black cohosh on androgen-sensitive LNCaP and androgen-insensitive PC-3 and DU 145 prostate cancer cells were investigated. Results showed the extract caused a significant dose- and time-dependent downregulation of all prostate cancer cell lines after 72 h (IC50 values between 37.1 and 62.7 μg/mL). Further, the study demonstrated that the extract killed prostate cancer cells by induction of apoptosis and activation of caspases, regardless of the hormone responsive status of the cells.34

A phenolic compound isolated from black cohosh, petasiphenone, was studied in vitro with regard to its effects on the proliferation of LNCaP cells (incubated with 10 nM oestradiol, 1 nM dihydrotestosterone, or without either) and their secretion of prostate-specific antigen (PSA). A dose-dependent inhibition of the LNCaP cells was observed regardless of hormone treatment, while PSA release was not altered.35

In an in vivo study, a black cohosh extract was tested in immunodeficient mice inoculated sc with LNCaP cells. Inoculation with the prostate cancer cells resulted in formation of solid tumours in 12 of 18 control animals, compared with only five of 18 animals treated with dietary black cohosh extract (2 to 2.8 mg extract/day). After 10 weeks, the amount of tumour tissue in the black cohosh-treated animals was significantly less than in controls. Serum testosterone levels were not significantly affected by the treatment.36

Other activity possibly contributing to menopausal effects

While the nature and degree of any oestrogenic activity of black cohosh remains unresolved, other potential mechanisms of action have emerged. That several mechanisms might be at play in a chemically complex herbal medicine is of course unsurprising.

A commercial black cohosh extract demonstrated in vitro dopaminergic activity in the dopamine receptor D2 assay,37 and there are suggestions that serotonergic effects might play a role in the therapeutic effects of black cohosh. Researchers at the University of Illinois at Chicago working on black cohosh extract did not find evidence of oestrogen receptor binding or oestrogenic effects in animals, but did observe strong binding to several serotonin receptor subtypes in vitro, with partial agonist activity at the 5-HT7 receptor.38 The same group has also shown that black cohosh acts as a mixed competitive ligand and partial agonist at the human mu opioid receptor.39

Central opioid activity has also been shown in a mechanistic study involving 11 postmenopausal women.40 The study employed two methods to examine the effects on central opioid function: naloxone challenge (n=6) and positron emission tomography with a selective micro-opioid receptor radioligand (n=5), before and after 12 weeks of standard treatment with a black cohosh isopropanolic extract (40 mg/day). Treatment did not affect oestrogen levels or spontaneous LH pulsatility, but administration of naloxone (a competitive mu opioid receptor antagonist) caused suppression of mean LH pulse frequency. This was most marked during sleep, when the mean interpulse interval was prolonged by 90 minutes. Positron emission tomography showed significant increases in mu opioid receptor binding potential in parts of the brain involved with emotional and cognitive function.

Because of the close relationship between the central nervous and hormone systems, it is plausible that mechanisms involving serotonergic, dopaminergic and opioid receptors are at least partly responsible for the effects of black cohosh on menopausal symptoms.

Anti-inflammatory and antioxidant activities

Several recent in vitro studies have lent some support to the traditional use of black cohosh in the treatment of inflammatory conditions. An aqueous extract (up to 6 mg/mL) was found to inhibit the generation of nitric oxide in lipopolysaccharide (LPS)-stimulated macrophages in a concentration-dependent manner. The mechanism for this was shown to be the reduced expression of inducible nitric oxide synthase (iNOS) rather than inhibition of iNOS activity. The triterpenoid glycoside 23-epi-26-deoxyactein was identified as the active compound.41

Another black cohosh compound, cimiracemate A (140 µM), has been shown to suppress the in vitro production of the inflammatory cytokine TNF-alpha induced by LPS in macrophages.42

Aqueous black cohosh extracts (3 and 6 mg/mL) have also been shown to reduce the release of TNF-alpha and the interleukin IL-6 (another inflammatory cytokine) and almost completely block the release of interferon-gamma in LPS-stimulated whole human blood. In contrast, IL-8 (also an inflammatory chemokine) was stimulated. Among five prevalent compounds isolated from the extract, isoferulic acid was found to be responsible for the inhibition of TNF-alpha and IL-6, but not for the stimulation of IL-8.43

Black cohosh showed potent antioxidant activity in the oxygen radical absorption capacity (ORAC) assay in a study that applied a sequential three solvent (ethyl acetate, methanol and 50% aqueous methanol) extraction process and combined the antioxidant activity of the three extracts. Black cohosh was the second most potent of 53 medicinal plants tested, being second only to olive leaf.44

Another in vitro study found black cohosh to be an effective scavenger of 1,1-diphenyl-2-picrylhydrazyl free radicals. Nine compounds with activity in this assay were isolated and six of these (methyl caffeate>caffeic acid>ferulic acid>cimiracemate A>cimiracemate B>fukinolic acid) were found to also reduce menadione-induced DNA damage in cultured S30 breast cancer cells.45

Other activity

Pretreatment with cimicifugoside inhibited blastogenesis in mouse splenic lymphocytes and brought about a decrease in the number of plaque-forming colonies using sheep erythrocytes (SRBC). The anti-SRBC response in the plaque-forming assay was suppressed in mice after pretreatment by intraperitoneal administration, and delayed hypersensitivity was suppressed after intravenous administration. The immunosuppressive activity of cimicifugoside is directed toward B-cell function, with larger doses being required for suppression of T-cell function.46

Black cohosh extract, and several fractions obtained from it, demonstrated hypotensive activity after intravenous administration at 1 mg/kg to rabbits. The hypotensive activity was observed in particular with the resinous part and may be due to actein. A hypotensive effect was not observed in human volunteers (by intravenous administration), although a peripheral vasodilatory effect was evident, even in subjects with peripheral arterial disease.47 Oral administration of black cohosh extract (2 g/kg) inhibited 5-hydroxytryptophan-induced diarrhoea in mice.48

Pharmacokinetics

The triterpene glycoside actein has been shown to be bioavailable in rats when administered by gastric intubation (37.5 mg/kg). Serum levels of actein peaked after 6 h at 2.4 μg/mL, dropping to 0.1 μg/mL 24 h post administration. The urine level of actein at 24 h was 0.8 μg/mL.49

The pharmacokinetics of one of the most abundant triterpenoid glycosides in black cohosh, 23-epi-26-deoxyactein, was studied in 15 healthy, postmenopausal women. They received single oral doses of a 75% ethanolic extract containing 1.4, 2.8 or 5.6 mg of 23-epi-26-deoxyactein. Serial blood and 24 h urine samples were collected and analysed. Peak plasma levels (2.2 to 12.4 ng/mL) and area under the time-concentration curve (a measure of bioavailability) of 23-epi-26-deoxyactein increased proportionally with dosage, and the half-life was 2 to 3 h. Less than 0.01% of the compound was recovered in the urine 24 h after administration, but metabolites were not detected.50

Clinical trials

Menopause

In phytotherapy, black cohosh is predominantly used in the treatment and management of menopausal symptoms. Menopause is the permanent cessation of menstruation and fertility defined as the absence of ovarian follicular activity for at least 12 months. The symptom picture associated with the transition from peri- to postmenopause can range from mild to debilitating, and the duration of symptoms can range from a few months up to 10 years.

Experts on menopause refer to a wide variety of physical symptoms including hot flushes, cardiac complaints, fatigue, vertigo, sweating, muscle pain, muscle spasm, joint pain, urinary incontinence, vaginal dryness and vaginal epithelium atrophy, and psychological symptoms: depression, anxiety, nervousness, irritability, forgetfulness, sleep disturbances and decreased libido. Vasomotor symptoms of hot flashes and night sweats are the most common physical manifestation, occurring in up to 80% of menopausal women.51 Women who experience vasomotor symptoms are also more likely to suffer sleep disturbances and depressive symptoms and experience a negative impact on their quality of life.

A broad array of treatments including hormone therapy, antidepressant medications, herbal and nutritional therapies, and diet and lifestyle modifications are utilised by women to ameliorate unwanted menopausal symptoms.

Black cohosh is the herbal medicine most commonly recommended for managing psychological and physical symptoms of menopause. Standardised black cohosh extract was used with success in the 1950s and 1960s for the treatment of menopausal symptoms, menstrual disturbances in young women (secondary but not primary amenorrhoea) and symptoms arising from ovarian dysfunction or insufficiency.

Research interest in black cohosh has gained momentum since the publication of the Women’s Health Initiative study in 2002. This study found an association between the long-term use of HRT and increased breast cancer and cardiovascular risk.52 As a result of these findings, prescribers and users of HRT began to explore non-hormonal alternatives, the most common being black cohosh. Black cohosh is prescribed alone in a number of proprietary formulas, but is also used in combination with other herbs and nutrients in formulations for menopausal symptoms.

Black cohosh for menopausal symptoms has been investigated extensively in clinical trials, but not all trials have been convincing. This may in part be due to poor trial design, including small sample size and variability in the dose used. Many trials (including several with positive outcomes) have used an extract dose equivalent to 40 mg/day of dried herb, a dose which is low in comparison to the more traditional one recommended in the British Herbal Pharmacopoeia 1983 (0.9 to 6 g/day dried root and rhizome).4

A number of recent comprehensive monographs,53 systematic reviews54,55 and a meta-analysis56 have examined the efficacy and safety of black cohosh for menopausal symptoms; this body of literature represents the most rigorous and global assessment of black cohosh currently available.

A systematic review published in 2008 included six double blind, randomised, controlled trials (n=1112) of black cohosh for the relief of menopausal symptoms.55 Trials that included women suffering medically induced menopause were excluded. Each of the included trials focused on menopausal symptoms, and used a placebo or a standard drug treatment as control. Three of the six trials used an isopropanolic extract in a daily dose equivalent to 40 mg herb, one trial used 160 mg of a 70% ethanolic extract (equivalent to 5 mg of triterpene glycosides), another used a 58% ethanolic extract equivalent to 40 mg of herb, and the sixth used 6.5 mg of a 60% ethanolic extract. Despite the authors’ conclusion that the evidence for the efficacy of black cohosh was still inconclusive, five of the six trials produced some positive outcomes. Details of these six studies are provided below.

In an early trial, Stoll used a randomised, double blind design in 75 women with symptoms of menopause. The 3-month study compared treatment with black cohosh with conjugated oestrogens and placebo. The Kupperman Menopausal Index (KMI), Hamilton anxiety scale, and proliferation status of vaginal epithelium were the outcome measures, and results demonstrated that black cohosh improved all parameters compared with placebo.57

Another 3-month randomised, double blind, multicentre trial involving 62 postmenopausal women also compared black cohosh (equivalent to 40 mg/day), conjugated oestrogens and placebo. Outcome measures included the Menopause Rating Scale (MRS), markers of bone formation and degradation, and endometrial thickness. Results of this trial showed that both active treatments were effective in reducing menopausal symptoms and both had beneficial effects on bone metabolism. Unlike treatment with conjugated oestrogens, black cohosh did not affect endometrial thickness.58

Osmers and co-workers conducted a 12-week randomised, double blind, placebo-controlled multicentre trial involving 304 women with menopausal symptoms. Women in the black cohosh group received extract equivalent to 40 mg/day, and the MRS was again the primary outcome measure. The women receiving black cohosh experienced significantly greater improvement in menopausal symptoms, especially hot flushes, compared with the placebo group. Women in early menopause benefited the most.59

A multicentre, randomised, placebo-controlled, double blind study was conducted by Frei-Kleiner and co-workers in 122 menopausal women experiencing at least three hot flushes daily. They were treated over 12 weeks with a black cohosh extract (equivalent to 42 mg herb) or placebo. Outcome measures were a weekly score for hot flushes, the Kupperman Index and the MRS. Overall the results showed no significant difference between the two groups. However, significant benefits of black cohosh were evident in women with more severe symptoms (Kupperman Index score of at least 20), and the effects of black cohosh over placebo almost reached significance (p=0.052) for the subgroup comprising perimenopausal women.60

In the HALT study, a 12-month randomised, double blind, trial was undertaken with 351 peri or postmenopausal women. They received either black cohosh (160 mg of a 70% ethanolic extract equivalent to 5 mg triterpene glycosides daily), a multi-botanical preparation (alfalfa, chaste tree, dong quai, false unicorn, licorice, oats, pomegranate, Siberian ginseng, boron; 200 mg daily), the multi-botanical plus telephone counselling to increase dietary soy intake, conjugated oestrogens with or without medroxyprogesterone acetate, or placebo. The main outcome measures were frequency and intensity of vasomotor symptoms. The study found no difference between the herbal interventions and placebo at 3, 6 or 12 months, whereas the HRT resulted in a significant decrease in vasomotor symptoms compared with placebo (p<0.001).61

Bai and colleagues conducted a 3-month double blind, multicentre, non-inferiority trial in China in 244 women with menopausal symptoms. They were randomised to receive either black cohosh extract (equivalent to 40 mg/day) or the synthetic steroid drug tibolone (2.5 mg/day), which has been shown in numerous placebo-controlled trials to be effective in the treatment of menopausal symptoms. The main outcome measures were the KI and the frequency of adverse events. The effects of the two treatments were similar and clinically relevant: in the black cohosh group mean KI decreased from 24.7 at baseline to 11.2 and 7.7 after 4 and 12 weeks, respectively, while the corresponding scores for the tibolone group were 11.2 and 7.5. These results demonstrated that the black cohosh treatment was as effective as tibolone treatment (non-inferiority was statistically significant, p=0.002), with the benefit-risk balance for black cohosh being significantly superior to tibolone (p=0.01).62

A 2010 meta-analysis by researchers at McGill University in Montreal included nine randomised, placebo-controlled trials of black cohosh-containing preparations for menopausal symptoms. Four of the included trials have been reviewed above; the additional five trials used black cohosh in combination with other active ingredients such as St John’s wort, isoflavones, lignans or other herbs. Overall, the authors of this meta-analysis found that six of the nine studies demonstrated a significant effect for the black cohosh-containing intervention over placebo. Combining the data from seven trials, they calculated an estimated improvement in menopausal vasomotor symptoms of 26% (95% CI 11–40%) with black cohosh treatment.56 However, the value of comparing different treatments in a meta-analysis is questionable. The five trials of black cohosh in combination with other active ingredients are reviewed below.

The efficacy of black cohosh in combination with St John’s wort was assessed in 89 symptomatic peri- and postmenopausal women in a 12-week double blind, randomised, placebo-controlled, multicentre study. The main outcome measure was the Kupperman Index (KI); biochemical parameters (hormone levels and lipid profiles) and pathology (vaginal atrophy) were also measured. The active treatment consisted of a 264 mg tablet containing 0.0364 mL of extract from black cohosh rhizome (equivalent to 1 mg terpene glycosides) and 84 mg of dried extract from Hypericum perforatum, equivalent to 0.25 mg hypericin. Treatment with the herbal preparation resulted in significantly greater reduction in KI scores after 4 (p=0.002) and 12 (p<0.001) weeks, compared with placebo. There were no other clinically significant differences between the two groups (although HDL levels decreased marginally in the placebo group). The results demonstrated this combination of black cohosh and St John’s wort to be effective in alleviating menopausal symptoms.63

A second study also examined the efficacy of the fixed combination of black cohosh and St John’s wort in women with menopausal symptoms. The double blind, randomised, placebo-controlled study included 301 women experiencing menopausal symptoms with a pronounced psychological component. They were treated with ethanolic St John’s wort extract and isopropanolic black cohosh extract or placebo for 16 weeks. The MRS and the Hamilton Depression Rating Scale were used to measure outcomes. The mean MRS score decreased 50% (0.46 to 0.23) in the treatment group and 19.6% (0.46 to 0.37) in the placebo group, whereas the Hamilton Depression Rating Scale total score decreased 41.8% (18.9 to 11.0) in the treatment group compared with 12.7% (18.9 to 16.5) in the placebo group. The herbal treatment was significantly (p<0.001) better than placebo for both measures.64

Another randomised, double blind, placebo-controlled trial in 50 healthy peri- and postmenopausal women assessed a formula containing standardised extracts of black cohosh, dong quai, milk thistle, red clover, American ginseng and chaste tree berry for 3 months. Frequency and intensity of menopausal symptoms were monitored by way of a structured questionnaire, administered weekly. Biochemical tests, breast checks and transvaginal ultrasonography were also performed. Women receiving the herbal formula reported a significant and progressive reduction in menopausal symptoms over the placebo group. After 3 months there was a 73% decrease in hot flushes and a 69% reduction of night sweats, with a decrease in symptom intensity and improved sleep quality in the herbal group. Complete cessation of hot flushes was reported by 47% of women in the herbal group compared with 19% in the placebo group. Vaginal ultrasonography, hormone levels (oestradiol, FSH), liver enzymes or thyroid-stimulating hormone showed no change in either group.65

A study by an Italian group evaluated the short-term effects of a combination of black cohosh with isoflavones and lignans on acute menopause-related symptoms in postmenopausal women using a double blind, randomised, placebo-controlled design. Eighty healthy, postmenopausal women were randomly assigned to receive either the combination formula or a placebo (calcium supplement). The groups were similar at baseline, but after 3 months of treatment the KI was significantly lower (p<0.05) in the black cohosh-phyto-oestrogen group compared with the placebo group.66

A supplement containing black cohosh and soy isoflavones was studied in a randomised, placebo-controlled, double blind multicentre 12-week trial involving 124 women who experienced at least five vasomotor symptoms every 24 h. They were randomised to receive daily either the black cohosh-isoflavone supplement or placebo. The modified KI and the Greene Climacteric Scale were used to measure outcomes. After 6 and 12 weeks of treatment, all scores had improved in both groups compared with baseline, and there was no statistically significant difference between the supplement and placebo groups.67

A large scale, observational study comparing the effect of black cohosh alone and black cohosh in combination with St John’s wort also deserves mention. The study was conducted in Germany between March 2002 and March 2004, and included 6141 women with any menopausal symptoms and who had not taken HRT in the 4 weeks preceding the study. The participants had mostly mild to moderate symptoms and were treated with recommended doses of black cohosh monotherapy (isopropanolic extract equivalent to 20 mg herb per tablet) or combination therapy (isopropanolic extract of black cohosh equivalent to 30 mg herb plus St John’s wort extract equivalent to 245 to 350 mg herb per tablet). The treating physician determined the choice and dose of treatment. Patients were evaluated at baseline, 3 and 6 months, with some patients electing to continue for a further 6 months. The primary outcome measures were the MRS and the PSYCHE sub-score of the MRS. In women receiving black cohosh monotherapy, total MRS scores declined from baseline by 0.10 and 0.14 after 3 and 6 months, respectively, whereas the corresponding decreases were 0.12 and 0.18 for the combination therapy, suggesting that the addition of St John’s wort provided an additional effect. Similarly, the changes on the PSYCHE sub-score were significantly greater with the combination therapy than with black cohosh alone (p<0.001). However, the data are potentially confounded by the fact that baseline scores for the two groups were not equal and doses were not fixed. Improvements were sustained by both treatments at 6 and 12 months.68

In a small double blind, placebo-controlled trial, black cohosh had no significant anxiolytic activity in women with anxiety disorder due to menopause.69

Breast cancer patients with menopausal symptoms

Black cohosh used in the management of hot flushes in patients with breast cancer has also been studied in clinical trials. Two such studies, with conflicting results, are summarised below. In an open label randomised trial, 136 breast cancer survivors (35 to 52 years) were treated over 12 months with a black cohosh extract for hot flushes caused by tamoxifen therapy. The patients had all been treated with segmental or total mastectomy, chemotherapy and radiation therapy and were receiving tamoxifen 20 mg/day (n=46) or tamoxifen 20 mg/day plus black cohosh (equivalent to 20 mg herb per day) (n=90). Results revealed those in the intervention (black cohosh) group had fewer and less severe hot flushes compared with controls. After 12 months’ treatment nearly half of those receiving black cohosh were completely free of hot flushes, while 24.4% of the black cohosh group versus 73.9% of the tamoxifen group were still reporting severe hot flushes (p<0.01).70

An earlier randomised, placebo-controlled trial involving 85 breast cancer survivors stratified for tamoxifen use (59 on tamoxifen and 26 not on tamoxifen) did not find any effect of black cohosh over placebo in terms of number and severity of hot flushes. The dose of black cohosh used in this trial was not provided in the report. Subjects completed a 4-day hot flush diary at baseline and at 30 and 60 days, as well as a menopause symptom questionnaire at baseline and at the final visit. FSH and LH levels were measured in a subset of patients at the first and final visits, but no significant differences were detected between the two groups.71

Bone loss prevention

Bone mineral loss (osteoporosis) is a major consideration for postmenopausal women, as it is a major age-related source of morbidity and mortality. A 12-week double blind study in 62 postmenopausal women compared the effects of a black cohosh preparation (equivalent to 40 mg herb per day), conjugated oestrogens or placebo on bone metabolism and other parameters. Black cohosh and conjugated oestrogens were found to have beneficial effects of similar magnitude on bone metabolism. Black cohosh treatment significantly increased serum levels of bone-specific alkaline phosphatase, indicative of increased osteoblast activity. Treatment with conjugated oestrogens, however, did not produce this effect, but appeared to decrease the activity of osteoclasts. Hence, while the net effects on bone of the two treatments were comparable, it appeared that the mechanism of action differed.58

This effect of black cohosh on bone density parameters appears to be mild and, on current evidence, it could not be credibly proposed as a treatment for osteoporosis. However, it probably should play a role as part of an appropriate diet, supplementation and lifestyle regime for the prevention of osteoporosis in both men and women, especially those with borderline osteopenia.

A study in 128 women found that, while an exercise programme favourably affected bone health, adjuvant supplementation with black cohosh (40 mg/day) did not enhance this effect.72 Perhaps a higher dose might have been necessary for this experimental design.

Arthritis

In a randomised, double blind, placebo-controlled trial, 82 male and female patients with osteoarthritis and rheumatoid arthritis received a licensed over-the-counter (OTC) herbal medicine (two tablets/day) or placebo for a period of 2 months. The formula contained black cohosh, willow bark, guaiacum resin, sarsaparilla and poplar bark. Although there was no significant difference between the two groups for most symptoms, a significant decrease (p<0.05) in pain scores occurred for those taking the herbal formula. Many patients reported a decline in health related to the cold, damp, windy weather experienced near the end of the trial, which may have altered the findings. A relative improvement in mood scores was also noted for those taking the herbal tablets.73 The authors of this study advised that the results may not be relevant to the activity of black cohosh, as the formulation predominantly comprised herbs containing salicylate derivatives.

Infertility

Unexplained infertility is typically defined as the failure to conceive over 1 year for couples exhibiting no apparent abnormalities. It is believed to occur in 15% to 30% of couples trying to conceive. Medically it is often treated with the fertility drug clomiphene citrate, which can be used in conjunction with gonadotropins to help trigger ovulation. In a controlled clinical trial, patients with unexplained infertility who had not responded to clomiphene therapy alone were randomly divided into two groups. The first group was given black cohosh dry extract at 120 mg/day (500 mg of dried root) from days 1 to 12 of the cycle. Both groups received clomiphene from days 3 to 7 and human chorionic gonadotropin (HCG) injection close to ovulation. All were recommended to have timed intercourse every day for the corresponding week. Endometrial thickness was measured on the day of the HCG injection and was found to be significantly thicker for the group receiving black cohosh (8.9±1.4 mm versus 7.5±1.3 mm, p<0.001). Serum progesterone concentrations measured in the luteal phase (days 21 to 23) were significantly higher in the black cohosh group (13.3±3.1 ng/mL versus 9.3±2.0 ng/mL, p<0.01). The pregnancy rate was also significantly higher for the group given black cohosh (36.7% versus 13.6%, p<0.01), even after one treatment cycle. These results suggest a benefit to including black cohosh in the medical management of infertility.74

A follow-up trial by the same group using a similar design found that follicular-phase black cohosh exerted a better activity than ethinyloestradiol in terms of improving cycle characteristics in 134 infertile women treated with clomiphene citrate. No significant difference was found regarding clinical pregnancy rates.75

Toxicology and other safety data

Toxicology

No toxic effects were observed from oral administration of standardised black cohosh extract (up to 5 g/kg/day) for 26 weeks in rats.76 A constituent isolated from the chloroform fraction of black cohosh extract, likely to be actein, did not provoke acute toxicity when administered by intragastric and hypodermic routes to rabbits. The minimum lethal dose of this constituent was greater than 500 mg/kg (ip) in mice, 1000 mg/kg (oral) in rats and 70 mg/kg (iv) in rabbits. In subchronic toxicity studies over 30 days, the minimum lethal dose was greater than 10 mg/kg (ip) in mice and 6 mg/kg (oral) in rabbits.77

Standardised black cohosh extract did not show mutagenic activity in the Ames test.78 Scientists from Duquesne University observed that the incidence of metastasis increased in sexually mature female transgenic (MMTV-neu, genetically engineered) mice fed black cohosh (at amounts said to reflect the normal human dose, about 0.3 mg/mouse) for 12 months. The incidence of mammary tumours was not increased.79 This experimental model, in which female mice spontaneously develop mammary tumours through the activation of an oncogene common in human breast cancer, is still highly controversial in terms of providing reproducible and relevant results. The experimental conditions were highly artificial (for example, feeding black cohosh to mice for 12 months is the equivalent of a woman taking it continuously for at least 30 years).

The relevance of using a mouse model to assess the safety of a treatment that is already widely used in the community can be queried; it could be that this in vivo study is nothing more than a scientific curiosity. The best way to assess any risks associated with black cohosh consumption is to study the health of women already taking it.

Such studies have now been published. The association between a range of ‘hormone-related supplements’ (including black cohosh) containing ‘phyto-oestrogens’ and breast cancer incidence was reassessed in a retrospective case-control epidemiological study.80 The US study examined 949 cases of women with breast cancer and 1524 controls and specifically targeted use of black cohosh, American ginseng (Panax quinquefolius), red clover (Trifolium pratense), dong quai (Angelica sinensis) and yam products (Dioscorea species). After adjusting for variables such as age, education, age at full-term pregnancy, menopause status, family history of breast cancer and use of HRT, intake of the above herbal products (as a group) was associated with a reduced incidence of breast cancer (adjusted odds ratio (OR) 0.65, 95% confidence interval (CI): 0.49 to 0.87). However, it was only black cohosh that demonstrated a highly significant breast cancer protective effect (adjusted OR 0.39, 95% CI: 0.22 to 0.70). The authors concluded that additional confirmatory studies are required to determine whether black cohosh could be used as a treatment to prevent breast cancer.

Within a German case-control study, associations between patterns of herbal product use and incidence of breast cancer were investigated in 10 121 postmenopausal women.81 Use of herbal products was inversely associated with invasive breast cancer (OR 0.74) in a dose-dependent manner. The two black cohosh subgroups (isopropanolic and other types of extracts) demonstrated moderate protection, but it was most marked for chaste tree (Vitex agnus-castus, OR 0.4). As part of the VITAL epidemiological study, 35 016 postmenopausal women were queried on their use of dietary supplements and followed for up to 7 years.82 Black cohosh use was not found to be associated with an increased risk of invasive breast cancer.

However, questions remain as to whether black cohosh can be safely used by women with diagnosed breast cancer. While more information is required, findings from one clinical study strongly imply that black cohosh lacks any oestrogenic activity in breast or endometrial tissue. This was a prospective, open, uncontrolled safety study in which baseline status was compared by blinded observers with status after 6 months of treatment.83 A total of 74 women were treated with black cohosh extract daily (40 mg/day), and 65 women completed the study. Mammograms were performed and breast cells were collected by percutaneous fine needle aspiration biopsies at baseline and after 6 months. Breast cell proliferation was assessed using the Ki-67/MIB-1 monoclonal antibody (cells positive for this marker are in a state of active proliferation). Safety was monitored by adverse event reporting, laboratory assessments and measurement of the endometrium by vaginal ultrasound.

None of the women showed any increase in mammographic breast density. Furthermore, there was no increase in breast cell proliferation. The mean change in the proportion of Ki-67-positive cells was 0.5%±2.4% for paired samples. The mean change in endometrial thickness was 0.0±0.9 mm. A modest number of adverse events were possibly related to treatment, but none of these was serious. Laboratory findings and vital signs were normal. The findings suggest that the isopropanolic extract of black cohosh tested did not cause adverse effects on breast tissue. Furthermore, the data did not indicate any endometrial or general safety concerns during 6 months of treatment.

This finding was supported by another study published at around the same time that found 12 weeks of black cohosh given to postmenopausal women had no impact on oestrogen markers in serum and no effect on pS2 (a potential marker of breast cancer activity) or cellular morphology in nipple aspirate fluid.84

An observational retrospective cohort study investigated breast cancer patients treated at German institutions.85 Of 18 861 patients, a total of 1102 had received therapy with an isopropanolic extract of black cohosh. The mean overall observation time was 3.6 years. Black cohosh was not linked with an increased risk of recurrence, but instead was associated with prolonged disease-free survival (hazard ratio 0.83).

Contraindications

A traditional contraindication is pregnancy and lactation (see below), except to assist with childbirth.

A systematic review of the safety and efficacy of black cohosh in patients with cancer concluded that black cohosh appears to be safe in breast cancer patients without risk for liver disease, although further research is needed.33

Black cohosh is contraindicated in patients with pre-existing liver disease.

Special warnings and precautions

As noted above, caution should be exercised in patients with oestrogen-sensitive malignant tumours, especially when using doses at the higher end of the range.

Patients on long-term black cohosh therapy should be monitored for signs and symptoms of liver damage.

Interactions

The antiproliferative effect of black cohosh extract in combination with tamoxifen was assessed in vitro on 17-beta-oestradiol-stimulated MCF-7 human breast cancer cells.86 Dilutions of black cohosh extract in the range 10–3 to 10–5 augmented the antiproliferative action of 10–5 tamoxifen. Whether this interaction also applies in vivo has not been established.

Although a black cohosh extract and six triterpenoid glycosides isolated from it were shown to inhibit the key drug metabolising cytochrome P450 enzyme CYP3A4 in vitro,87 two human drug interaction studies have indicated this does not occur in people. One study found that a black cohosh extract (40 mg twice daily, standardised to 2.5% triterpene glycosides) did not affect CYP3A, which is involved in the metabolism of about half of all pharmaceutical drugs.88 The other study screened a black cohosh extract (1090 mg twice daily, each capsule standardised to 0.2% triterpene glycosides) for effects on the drug metabolising enzymes CYP3A4/5, CYP1A2, CYP2E1 and CYP2D6 in 12 healthy volunteers who took the extract for 28 days. There was a statistically significant inhibition of only CYP2D6, but the magnitude of the effect was small (around 7%) and deemed unlikely to be of clinical relevance.89 A subsequent study conducted by the same group, in which 18 healthy volunteers took a standardised black cohosh extract for 14 days, found no evidence for an effect on CYP2D6.90 A separate drug interaction study (using digoxin as a probe drug) found no evidence for effects on the drug transporter P-glycoprotein in 16 healthy volunteers who took a black cohosh extract (equivalent to 40 mg herb per day) for 2 weeks.91

The potential for black cohosh to alter the response to radiation therapy and four common anticancer drugs (docetaxel, doxorubicin, cisplatin and 4-hydroperoxycyclophosphamide – 4-HC, an analogue of cyclophosphamide that is active in cell culture) was studied in vitro in a mouse breast cancer cell line. Black cohosh extract increased the cytotoxicity of docetaxel and doxorubicin, decreased the cytotoxicity of cisplatin and did not alter the effects of 4-HC or radiation.92 The relevance of these findings to the human use of black cohosh is uncertain.

Use in pregnancy and lactation

Category B2 – no increase in frequency of malformation or other harmful effects on the fetus from limited use in women. Animal studies are lacking.

The traditional position is generally that black cohosh should not be taken during pregnancy except to assist with birth. According to the British Herbal Compendium black cohosh is contraindicated in pregnancy;93 however, this restriction is not listed in the Commission E.94 Black cohosh was widely used by the Eclectics in traditional Western herbal medicine as a partus preparator, if taken in the last weeks of pregnancy.5

In a 1999 survey of certified nurse-midwives in the US, black cohosh was used by 45% of the 90 respondents who used herbal medicine to stimulate labour. Adverse effects attributed to use of blue cohosh and black cohosh were not separately assigned and included nausea, increased meconium-stained fluid and transient fetal tachycardia.95

After a normal labour, a female infant was not able to breathe spontaneously and sustained CNS hypoxic-ischaemic damage. A midwife had attempted induction of labour using a combination of blue cohosh and black cohosh given orally (dosage undefined) at around 42 weeks’ gestation.96 It was not possible to identify the herbal preparation as the causative agent; however, this reaction may have been due to the blue cohosh (Caulophyllum thalictroides) rather than to the black cohosh (see Chapter 5).

A prospective, epidemiological study investigated the influence of first trimester use of medications and vaccines in the 1950s on the occurrence of congenital malformations and fetal survival in approximately 3200 pregnancies. Black cohosh was used in 1 of 266 pregnancies where a malformation occurred and in 2 of 532 pregnancies from the comparison groups. The dose and duration of use of black cohosh and the nature of the malformation were not specified.97 Black cohosh could not be identified as a causative agent.

Black cohosh use should be strongly discouraged during breastfeeding. This consideration is based on a possible oestrogenic effect. The British Herbal Compendium contraindicates black cohosh during lactation.93 However, the Commission E does not list this restriction.94

Effects on ability to drive or operate machinery

No adverse effects expected.

Side effects

General side effects

High doses of black cohosh can cause a frontal headache, with a dull, full or bursting feeling. This headache is the most characteristic effect observed when giving even therapeutic doses.98 A review published in 2000 found that mild gastrointestinal upset was the most frequent minor adverse event reported in clinical studies (average of 5.6% of patients across five studies). Other minor adverse events reported in clinical studies included headache, vertigo, weight gain, mastalgia, heavy feeling in the legs and a stimulant effect. Vaginal bleeding has also been reported.2

Two reviews published in 2003 confirm that adverse events with black cohosh are rare, mild and reversible. Gastrointestinal upsets and rashes are the most common adverse events. There is record of a few serious adverse events, including hepatic and circulatory conditions, but causality could not be determined.99,100 Details of some of the case reports follow.

A case was reported in 2001 of a woman diagnosed with grade 1 endometrioid adenocarcinoma of the endometrium ‘whose history was notable for extensive use of supplemental phytoestrogens’. Herbs included chaste tree, dong quai, black cohosh and licorice.101 No causality was demonstrated.

A 45-year-old woman who had been taking separate bottled products of black cohosh, Vitex agnus-castus and evening primrose oil for 4 months had three nocturnal seizures within a 3-month period. The patient had also consumed one to two beers 24 to 48 h prior to each incident.102 It was not established whether the herbal preparations caused the seizures.

A 26-year-old woman presented at a hospital with chest pain. Her heart rate and blood pressure dropped temporarily during the course of monitoring. Her urine digoxin level was ‘elevated’ at 0.9 ng/mL (but within the normal therapeutic range – 0.5 to 2.0 ng/mL). However, she was not taking digoxin. In addition to the contraceptive pill, she was taking a herbal preparation containing black cohosh, skullcap (Scutellaria lateriflora), lousewort (Pedicularis canadensis), hops (Humulus lupulus), valerian (Valeriana officinalis) and cayenne pepper (Capsicum annuum). The product was not available for analysis. The chest pain had started during her shift as a topless dancer, during which she had consumed four alcoholic drinks, but no illicit drugs.103 This study inappropriately speculated on ‘digoxin-like factors’ with cardiotonic activity claimed to be ‘commonly’ found in herbal teas. The source of the patient’s symptoms remains a mystery, but factors that interfere with digoxin assays, yet are without cardiotonic activity, have been reported in some herbs. (Refer to the Ginseng (Eleutherococcus senticosus) monograph by way of example.)

A case report exists of a 54-year-old woman who developed severe asthenia and high blood levels of creatine phosphokinase (230 to 237 U/L), lactate dehydrogenase (504 to 548 U/L) and total cholesterol, while taking a supplement derived from black cohosh for the management of vasomotor symptoms related to menopause. Notably the woman had previously taken the product for 12 months with no alteration in biochemistry and had restarted the product after a 3-month break. The black cohosh tablets contained 20 mg of dried rhizome and root extract. No other cause could be identified for her symptoms and she was advised to discontinue the product, after which a progressive normalisation of biochemical parameters and improvement in clinical symptoms occurred.104

A case report described a 56-year-old woman diagnosed with cutaneous pseudolymphoma after taking a black cohosh product for 12 months. The localised erythematous plaques on her arms and legs appeared after 6 months and completely disappeared with withdrawal of the product for 3 months.105

Other adverse reactions attributed to black cohosh use include cutaneous vasculitis106 and coagulation activation with fluid retention (secondary to a transient autoimmune hepatitis).107

Liver injury

On the 9 February 2006, the Australian TGA announced the following:

The Therapeutic Goods Administration (TGA) reviewed the safety of Black cohosh (Cimicifuga racemosa) following reports of possible liver problems internationally and in Australia. At the time of the review, there were 47 cases of liver reactions worldwide, including 9 Australian cases. In Australia, 4 patients were hospitalised, including two who required liver transplantation. Although some reports are confounded by multiple ingredients, by more than one medication or by other medical conditions, there is sufficient evidence of a causal association between Black cohosh and serious hepatitis. However, considering the widespread use of Black cohosh, the incidence of liver reaction appears to be very low. Following the safety review, the TGA has decided that medicines containing Black cohosh should include the following label statement: ‘Warning: Black cohosh may harm the liver in some individuals. Use under the supervision of a healthcare professional’.

On July 18, 2006, the Medicines and Healthcare Products Regulatory Agency (MHRA) in the UK issued a press release stating that all black cohosh products sold there should carry the following label warning: ‘Black cohosh may rarely cause liver problems. If you become unwell (yellowing eyes/skin, nausea, vomiting, dark urine, abdominal pain, unusual tiredness) stop taking immediately and seek medical advice. Not suitable for patients with a previous history of liver disease.’

Notwithstanding the Australian TGA’s claim at the time concerning the number of cases of liver damage linked to black cohosh (which include the adverse reaction reports filed with the UK MHRA and other health authorities) only five papers or letters had been published purporting to demonstrate a link between black cohosh (Actaea racemosa) ingestion and subsequent liver injury. It is important to closely examine these published reports since, because of the process of peer review, these represent the best-documented evidence of any association with liver injury. The first publication, from doctors at the Princess Alexandra Hospital in Brisbane, Australia, described six patients with evidence of severe hepatitis that was linked to taking a range of herbal products.108 Two of these patients were taking black cohosh, although one was also taking other herbs including skullcap, a herb which can be substituted by Teucrium species, a known hepatotoxic genus.109

The one case attributed to black cohosh alone (Case 1) was truly dramatic. Of the cases reported, the most serious illness occurred in this 47-year-old woman who was taking black cohosh for menopausal symptoms. She required liver transplantation even though, according to the publication, the patient had been taking the black cohosh for just 1 week. Histological examination of her liver confirmed severe hepatitis and early fibrosis. The patient did not exhibit eosinophilia and had no signs of any systemic disturbance. Serology for hepatitis A, B and C was negative, but rechallenge with the herb was not performed ‘for ethical reasons’. The dose of black cohosh taken was not specified, neither was the product.

The second publication, also from Australia, describes a 52-year-old woman with acute liver failure (Case 2).110 She had been taking an herbal formula containing 1:1 liquid extracts prescribed by a pharmacist. Black cohosh 1:1 was 10% of the mixture and the daily dose of the combination was 7.5 mL twice a day. The patient underwent successful liver transplantation. Although the authors stated that: ‘Extensive investigation excluded other recognised causes of liver failure’, they provided no details of what these investigations were. Analysis by the TGA was said to confirm the presence of golden seal, black cohosh and Ginkgo in the herbal mixture. Other stated ingredients were ground ivy and oats seed.

The phenomenon of idiosyncratic hepatic reactions to drugs is well documented. It also appears that this reaction does occur to certain herbs, for example chaparral (Larrea tridentata) and germander (Teucrium species). By definition, such reactions are rare and unpredictable and are not dose related. There are two types of idiosyncratic hepatic injury: hypersensitivity and aberrant metabolism. The former develops 1 to 5 weeks after exposure to the drug and, since it is immune-mediated and acute, also involves a systemic reaction including rash, fever and eosinophilia. The latter takes weeks to months to develop and symptoms are confined to the liver. Diagnosis of drug-induced idiosyncratic liver injury (DILI) is very difficult and relies largely on circumstantial evidence. Factors taken into account include a temporal association, exclusion of other possible causes, a consistent latency period to those described above, presence or absence of hypersensitivity (systemic) features, positive response to drug removal (dechallenge), positive response to rechallenge and a positive lymphocyte stimulation test (this last factor is quite controversial). Complicating this is the fact that DILI can mimic every known human liver disease.111 There are many confounding factors that could lead to incorrect associations between ingested medications or herbs and idiosyncratic liver injury. Many viruses that cause liver disease are still to be identified112 and there are no tests for them. Even known viruses are not always tested for. For example, a Dutch study published this year found that hepatitis E virus was a significant cause of unexplained hepatitis.113 Occult coeliac disease has been suggested as a cause of unexplained raised ALT and AST.114 Rare liver diseases may not be excluded.115,116 Other environmental factors could be implicated.

The experience of a liver transplant unit highlights some key issues behind the history and incidence of severe acute hepatitis (fulminant hepatic failure, FHF). All adult cases of FHF presenting to the Victorian Liver Transplant Unit (Australia) from 1988 to 2002 were analysed. Eighty patients (mostly female) were referred, at a rate of approximately one case per million population per year. Mean age was about 38 years. Most cases were due to paracetamol poisoning (36%) or idiopathic hepatitis (34%).110 Only five of the 80 cases were classified as drug induced, making this causality a rare factor. Other main causes included hepatitis A (three cases), hepatitis B (eight cases) and Wilson’s disease (six cases). The 27 cases (34%) of hepatitis due to unknown causes (idiopathic) is a surprising rate. These cases are also described as non-A non-B hepatitis, since patients are not positive for hepatitis A or B. In the USA one study found that the most common cause of FHF was non-A non-B (idiopathic) hepatitis.117 (Note that this US study was published in 1995, well before the dramatic rise in herbal use in that country.) Presumably unidentified infectious or environmental factors could cause these cases of idiopathic hepatitis. However, the authors of the Australian study state: ‘The strong female predominance of cases argues against a viral cause and raises the possibility that hormonal factors are involved, or that the condition is linked to autoimmune liver diseases. There is clearly a need for large, detailed, multicentre epidemiological studies, to provide further clues to a possible aetiology/ies of this syndrome.’

The demographics of idiopathic hepatitis (female, late 30s to early 50s) and black cohosh use strongly overlap. Hence, there is a distinct possibility that some patients who develop idiopathic hepatitis might also be coincidentally taking black cohosh. The herb could then be mistakenly attributed as the cause.

Since these initial series of cases attributing idiosyncratic hepatotoxicity to black cohosh, more cases have been reported. There have also been several publications analysing these and the earlier case reports, especially from the team headed by Teschke.

In 2009 the group rigorously analysed all 69 reported cases (at the time) and found no likelihood of causality in 68.118 Most cases were marred by confounding variables, misreported data and a lack of critical information.119 In particular, there was a lack of identification of the herb involved in the initial cases.120

Of high relevance here are the findings of Health Canada. From January 2005 to March 2009, Health Canada received six domestic reports of liver adverse reactions suspected of being associated with black cohosh. Analysis of three products associated with these reports revealed that they did not contain authentic black cohosh. Their phytochemical profiles were consistent with the presence of other related herbal species. A review of the authenticity of all licensed products containing black cohosh resulted in the voluntary withdrawal of several products not containing authentic black cohosh, including the products reported in four of the adverse reaction cases.121

Studies in black cohosh users have also sought to understand its impact on the liver. A prospective, longitudinal study recruited 100 healthy postmenopausal women from a hospital in Egypt. The women received black cohosh extract (40 mg/day) for relief of menopausal symptoms and were followed up for 12 months. Eighty-seven women completed the study, which included evaluation of total hepatic blood flow and liver function. The study sought to investigate potential mechanisms of hepatotoxicity: compromise of blood flow to the liver or a direct toxic effect on liver cells. The following results were obtained after 12 months of treatment:

• No significant changes in hepatic artery blood flow, portal vein blood flow or total hepatic blood flow

• No significant changes in any liver function tests

• A significant reduction in the prevalence, daily frequency and severity of hot flushes, compared with baseline.122

A meta-analysis of five trials involving 1117 women found no evidence that the isopropanolic extract of black cohosh has an adverse effect on liver function.123

Overdosage

According to early data, ingestion of 5 g of the herb or 12 g of the fluid extract can cause nausea, vomiting, violent headache, vertigo, joint pain, red eyes and weak pulse. Visual and nervous disturbances have also been noted.8,124,125 Some of these effects may have been due to the past adulteration of black cohosh with the poisonous plants red baneberry (Actaea spicata) and white cohosh or white baneberry (A. panchypoda (A. alba)).126 However, in the absence of any further information such doses of black cohosh are not recommended.

Safety in children

No information available, although the Eclectics did describe use in children for fever.5

Regulatory status in selected countries

Black cohosh is covered by a positive Commission E monograph with the following applications: neurovegetative complaints of premenstrual, dysmenorrhoeic or climacteric origin.

Black cohosh is on the UK General Sale List, with a maximum single dose of 200 mg. However, it does require a warning statement as noted above. Black cohosh products have achieved Traditional Herbal Registration in the UK with the traditional indication of relief of symptoms of the menopause.

Black cohosh does not have GRAS status. However, it is freely available as a ‘dietary supplement’ in the USA under DSHEA legislation (1994 Dietary Supplement Health and Education Act). Black cohosh has been present in OTC menstrual drug products. The FDA advises: ‘based on evidence currently available, there is inadequate data to establish general recognition of the safety and effectiveness of these ingredients for the specified uses’.

Black cohosh is not included in Part 4 of Schedule 4 of the Therapeutic Goods Act Regulations of Australia and is freely available for sale provided products carry a warning regarding possible liver damage.

References

1. Compton JA, Culham A, Jury SL. Taxon. 1998;47:593–634.

2. American Herbal Pharmacopoeia. Black cohosh rhizome: Actaea racemosa L. syn. Cimicifuga racemosa (L.) Nutt.: Analytical, Quality Control, and Therapeutic Monograph. Santa Cruz: American Herbal Pharmacopoeia, 2002.

3. German Federal Minister of Justice. German Commission E for Human Medicine Monograph, Bundes-Anzeiger (German Federal Gazette) no. 43, dated 02.03.1989.

4. British Herbal Medicine Association. British Herbal Pharmacopoeia. Cowling: BHMA, 1983. p. 66

5. Felter HW, Lloyd JU. King’s American Dispensatory, 18th ed., rev 3. Portland: 1905. Reprinted by: Eclectic Medical Publications, 1983;1:528–533.

6. Harnischfeger G, Stolze H. Erfahrungsheilkunde. 1981;30(6):439–444.

7. Chiej R. The Macdonald Encyclopedia of Medicinal Plants. London: Macdonald, 1984. Entry no. 87

8. Grieve MA, A Modern Herbal, New York, Dover Publications, 1971;vol 1. p. 211

9. Information on file. Warwick, Queensland 4072, Australia: MediHerb Pty Ltd; 2003.

10. Hostettmann K, Marston A. Chemistry & Pharmacology of Natural Products: Saponins. Cambridge: Cambridge University Press, 1995. p. 280

11. Wagner H, Bladt S. Plant Drug Analysis: A Thin Layer Chromatography Atlas, 2nd ed. Berlin: Springer-Verlag, 1996. p. 336

12. Gizycki H. Z Exp Med. 1944;113:635–644.

13. Jarry H, Harnischfeger G. Planta Med. 1985;51(1):46–49.

14. Jarry H, Ch Gorkow, Wuttke W, Loew D, Rietbrock N, eds. Phytopharmaka in Forschung und klinischer Anwendung, Darmstadt, Steinkopff (Verlag), 1995:104.

15. Jarry H, Harnischfeger G, Düker EM. Planta Med. 1985;51(4):316–319.

16. Duker EM, Kopanski L, Jarry H, et al. Planta Med. 1991;57(5):420–424.

17. Jarry H, Leonhardt S, Duls C, et al. 23rd International LOF-Symposium on Phyto-Oestrogens. Belgium: University of Gent, 1999.

18. Jarry H, Ch Gorkow, Wuttke W, Loew D, Rietbrock N, eds. Phytopharmaka in Forschung und klinischer Anwendung, Darmstadt, Steinkopff (Verlag), 1995:109–110.

19. Einer-Jensen N, Zhao J, Andersen KP, et al. Maturitas. 1996;25(2):149–153.

20. Zava DT, Dollbaum CM, Blen M. Proc Soc Exp Biol Med. 1998;217(3):369–378.

21. Liu J, Burdette JE, Xu H, et al. J Agric Food Chem. 2001;49(5):2472–2479.

22. Gaube F, Wolfl S, Pusch L, et al. BMC Pharmacol. 2007;7:11.

23. Bolle P, Mastrangelo S, Perrone F, et al. J Steroid Biochem Mol Biol. 2007;107(3–5):262–269.

24. Chan BY, Lau KS, Jiang B, et al. Bone. 2008;43(3):567–573.

25. Seidlová-Wuttke D, Jarry H, Wuttke W. Planta Med. 2007;73:995.

26. Kolios L, Schumann J, Sehmisch S, et al. Planta Med. 2010;76(9):850–857.

27. Qiu SX, Dan C, Ding LS, et al. Chem Biol. 2007;14(7):860–869.

28. Nesselhut T, Schellhase C, Dietrich R, et al. Arch Gynecol Obstet. 1993;254(1–4):817–818.

29. Nesselhut T. Expert Forum on Remifemin®: Report and Results From Endocrinology Expert Forum in Luneburg. Salzgitter (Ringelheim): Schaper & Brummer GmbH & Co., 1993.

30. Einbond LS, Shimizu M, Xiao D. Breast Cancer Res Treat. 2004;83(3):221–231.

31. Einbond LS, Su T, Wu HA. Int J Cancer. 2007;121(9):2073–2083.

32. Einbond LS, Shimizu M, Nuntanakorn P, et al. Planta Med. 2006;72(13):1200–1206.

33. Walji R, Boon H, Guns E, et al. Support Care Cancer. 2007;15:913–921.

34. Hostanska K, Nisslein T, Freudenstein J, et al. Anticancer Res. 2005;25(1A):139–147.

35. Jarry H, Stromeier S, Wuttke W, et al. Planta Med. 2007;73(2):184–187.

36. Seidlová-Wuttke D, Thelen P, Wuttke W. Planta Med. 2006;72(6):521–526.

37. Jarry H, Metten M, Spengler B, et al. Maturitas. 2003;44(Suppl 1):S31–S38.

38. Burdette JE, Liu J, Chen SN, et al. J Agric Food Chem. 2003;51(19):5661–5670.

39. Rhyu MR, Lu J, Webster DE, et al. J Agric Food Chem. 2006;54(26):9852–9857.

40. Reame NE, Lukacs JL, Padmanabhan V, et al. Menopause. 2008;15(5):832–840.

41. Schmid D, Gruber M, Woehs F, et al. J Pharm Pharmacol. 2009;61(8):1089–1096.

42. Yang CL, Chik SC, Li JC, et al. J Med Chem. 2009;52(21):6707–6715.

43. Schmid D, Woehs F, Svoboda M, et al. Can J Physiol Pharmacol. 2009;87(11):963–972.

44. Wojcikowski K, Stevenson L, Leach D, et al. J Altern Complement Med. 2007;13(1):103–109.

45. Burdette JE, Chen SN, Lu ZZ, et al. J Agric Food Chem. 2002;50(24):7022–7028.

46. Hemmi H, Ishida N. J Pharmacobiodyn. 1980;3(12):643–648.

47. Genazzani E, Sorrentino L. Nature. 1962;194:544–545.

48. Yoo JS, Jung JS, Lee TH, et al. Korean J Pharmacogn. 1995;4(26): 355

49. Einbond LS, Soffritti M, Esposti DD, et al. Fundam Clin Pharmacol. 2009;23(3):311–321.

50. van Breemen RB, Liang W, Banuvar S, et al. Clin Pharmacol Ther. 2010;87(2):219–225.

51. Bachmann GA. J Reprod Med. 2005;50(3):155–165.

52. Rossouw JE, Anderson GL, Prentice RL, et al. JAMA. 2002;288(3):321–333.

53. Knöss W. Assessment Report on Cimicifuga racemosa (L.) Nutt., rhizoma. London: European Medicines Agency, 2010.

54. Palacio C, Masri G, Mooradian AD. Drugs Aging. 2009;26(1):23–36.

55. Borrelli F, Ernst E. Pharmacol Res. 2008;58(1):8–14.

56. Shams T, Setia MS, Hemmings R, et al. Altern Ther Health Med. 2010;16(1):36–44.

57. Stoll W. Therapeuticum. 1987;1:23–31.

58. Wuttke W, Seidlová-Wuttke D, Gorkow C. Maturitas. 2003;44(suppl 1):S67–S77.

59. Osmers R, Friede M, Liske E, et al. Obstet Gynecol. 2005;105(5 Pt 1):1074–1083.

60. Frei-Kleiner S, Schaffner W, Rahlfs VW, et al. Maturitas. 2005;51(4):397–404.

61. Newton KM, Reed SD, LaCroix AZ, et al. Ann Intern Med. 2006;145(12):869–879.

62. Bai W, Henneicke-von Zepelin HH, Wang S, et al. Maturitas. 2007;58(1):31–41.

63. Chung DJ, Kim HY, Park KH, et al. Yonsei Med J. 2007;48(2):289–294.

64. Uebelhack R, Blohmer JU, Graubaum HJ, et al. Obstet Gynecol. 2006;107(2 Pt 1):247–255.

65. Rotem C, Kaplan B. Gynecol Endocrinol. 2007;23(2):117–122.

66. Sammartino A, Tommaselli GA, Gargano V, et al. Gynecol Endocrinol. 2006;22(11):646–650.

67. Verhoeven MO, van der Mooren MJ, van de Weijer PH, et al. Menopause. 2005;12(4):412–420.

68. Briese V, Stammwitz U, Friede M, et al. Maturitas. 2007;57(4):405–414.

69. Amsterdam JD, Yao Y, Mao JJ, et al. J Clin Psychopharmacol. 2009;29(5):478–483.

70. Hernández Muñoz G, Pluchino S. Maturitas. 2003;44(suppl 1):S59–S65.

71. Jacobson JS, Troxel AB, Evans J, et al. J Clin Oncol. 2001;19(10):2739–2745.

72. Bebenek M, Kemmler W, von Stengel S, et al. Menopause. 2010;17(4):791–800.

73. Mills SY, Jacoby RK, Chacksfield M, et al. Br J Rheumatol. 1996;35(9):874–878.

74. Shahin AY, Ismail AM, Zahran KM, Makhlouf AM. Reprod Biomed Online. 2008;16(4):580–588.

75. Shahin AY, Ismail AM, Shaaban OM. Reprod Biomed Online. 2009;19(4):501–507.

76. Korn WD. Six-month Oral Toxicity Study with Remifemin Granulate in Rats Followed by an Eight-week Recovery Period. Hannover, Germany: International Bioresearch, 1991.

77. Genazzani E, Sorrentino L. Nature. 1962;194(4828):544–545.

78. Beuscher N. Z Phytother. 1995;16:301–310.

79. Davis VL, Jayo MJ, Ho A, et al. Cancer Res. 2008;68(20):8377–8383.

80. Rebbeck TR, Troxel AB, Norman S, et al. Int J Cancer. 2007;120:1523–1528.

81. Obi N, Chang-Claude J, Berger J, et al. Cancer Epidemiol Biomarkers Prev. 2009;18(8):2207–2213.

82. Brasky TM, Lampe JW, Potter JD, et al. Cancer Epidemiol Biomarkers Prev. 2010;19(7):1696–1708.

83. Hirschberg AL, Edlund M, Svane G, et al. Menopause. 2007;14(1):89–96.

84. Ruhlen RL, Haubner J, Tracy JK, et al. Nutr Cancer. 2007;59(2):269–277.

85. Zepelin HH, Meden H, Kostev K, et al. Int J Clin Pharmacol Ther. 2007;45(3):143–154.

86. German Patent DE 196 52 183 CI. Verwendung eines Extraktes aus Cimicifuga racemosa: Schaper & Brümmer.

87. Tsukamoto S, Aburatani M, Ohta T. Evid Based Complement Alternat Med. 2005;2(2):223–226.

88. Gurley B, Hubbard MA, Williams DK, et al. J Clin Pharmacol. 2006;46(2):201–213.

89. Gurley BJ, Gardner SF, Hubbard MA, et al. Clin Pharmacol Ther. 2005;77(5):415–426.

90. Gurley BJ, Swain A, Hubbard MA, et al. Mol Nutr Food Res. 2008;52(7):755–763.

91. Gurley BJ, Barone GW, Williams DK, et al. Drug Metab Dispos. 2006;34(1):69–74.

92. Rockwell S, Liu Y, Higgins SA. Breast Cancer Res Treat. 2005;90(3):233–239.

93. British Herbal Medicine Association. British Herbal Compendium. Bournemouth, BHMA; 1992. pp. 34–36.

94. Blumenthal M, ed. The Complete German Commission E Monographs: Therapeutic Guide to Herbal Medicines. Austin: American Botanical Council, 1998.

95. McFarlin BL, Gibson MH, O’Rear J, et al. J Nurse Midwifery. 1999;44(3):205–216.

96. Gunn TR, Wright IM. N Z Med J. 1996;109(1032):410–411.

97. Mellin GW. Am J Obst Gynec. 1964;90(7, Pt 2):1169–1180.

98. Felter HW. The Eclectic Materia Medica, Pharmacology and Therapeutics, 1922. Portland: Eclectic Medical Publications, 1983. pp. 466–469

99. Dog TL, Powell KL, Weisman SM. Menopause. 2003;10(4):299–313.

100. Huntley A, Ernst E. Menopause. 2003;10(1):58–64.

101. Johnson EB, Muto MC, Yanushpolsky EH, et al. Obstet Gynecol. 2001;98(5 Pt 2):947–950.

102. Shuster J. Hosp Pharm. 1996;31(12):1553–1554.

103. Scheinost ME. J Am Osteopathic Assoc. 2001;101(8):444–446.

104. Minciullo PL, Saija A, Patafi M, et al. Phytomedicine. 2006;13(1–2):115–118.

105. Meyer S, Vogt T, Obermann EC, et al. Dermatology. 2007;214(1):94–96.

106. Ingraffea A, Donohue K, Wilkel C, Falanga V. J Am Acad Dermatol. 2007;56(suppl 5):S124–S126.

107. Zimmermann R, Witte A, Voll RE, et al. Climacteric. 2010;13(2):187–191.

108. Whiting PW, Clouston A, Kerlin P. MJA. 2002;177(8):440–443.

109. Mills S, Bone K. The Essential Guide to Herbal Safety. USA: Churchill Livingstone, 2005. pp. 581–584

110. Lontos S, Jones RM, Angus PW, et al. MJA. 2003;179(7):390–391.

111. Pishvaian AC, Trope BW, Lewis JH. Curr Opin Gastroenterol. 2004;20(3):208–219.

112. Bowden D, Moaven LD, Locarnini S. MJA. 1996;164:87–89.

113. Waar K, Herremans M, Vennema H. J Clin Virol. 2005;33(2):145–149.

114. Bardella M, Vecchi M, Conte D. Hepatology. 1999;29(3):654–657.

115. Tordjmann T, Grimbert S, Genestie C. Gastroenterol Clin Biol. 1998;22(3):305–310.

116. Gaya D, Thorburn D, Oien K. J Clin Pathol. 2003;56(11):850–853.

117. Hoofnagle J, Carithers R, Shapiro C. Hepatology. 1995;21:240–252.

118. Teschke R, Bahre R, Genthner A, et al. Maturitas. 2009;63(4):302–314.

119. Teschke R. Menopause. 2010;17(2):426–440.

120. Teschke R, Schwarzenboeck A, Schmidt-Taenzer W, et al. Ann Hepatol. 2011;10(3):249–259.

121. Black cohosh products and liver toxicity: update. Canadian Adverse Reaction Newsletter. 2010:1–2.

122. Nasr A, Nafeh H. Fertil Steril. 2009;92(5):1780–1782.

123. Naser B, Schnitker J, Minkin MJ, et al. Menopause. 2011;18(4):366–375.

124. Mills SY. The A-Z of Modern Herbalism. London: Thorsons, 1989. p. 39

125. Blaschek W, Ebel S, Hackenthal E, et al. HagerROM 2002: Hagers Handbuch der Drogen und Arzneistoffe. Heidelberg: Springer, 2002.

126. Grieve M, A Modern Herbal, New York, Dover Publications, 1971;vol 1. pp. 81–82

Boswellia

(Boswellia serrata Roxb. ex Colebr.)

Synonyms

Boswellia glabra Roxb. (botanical synonym), Boswellia, Indian frankincense, Indian olibanum (Engl), Salaibaum (Ger), Baswellie-dentelee (Fr), (Dan) Sallaki (Sanskrit) Salai guggal (Hindi).

What is it?

The Boswellia shrubs and small trees belong to the same family (Burseraceae) as the trees that produce myrrh and are native to the dry tropics of Africa (especially the northeast) and Asia. The fragrant resins exuded from many species of Boswellia are known as frankincense or oleo-gum olibanum and have been used since antiquity for incense and embalming liquids. (Boswellia serrata resin is closely related to the Biblical frankincense (B. carterii).) As well as a long tradition of therapeutic use, particularly in Ayurveda, Boswellia serrata has recently been tested in clinical studies for a wide range of inflammatory conditions. Research has centred on the triterpenoids, especially the boswellic acids, which are considered to be responsible for the observed anti-inflammatory and antiarthritic activities of the resin. Studies on B. carterii have generally not been included in this review.

Effects

Reduces inflammation in a variety of body tissues; possibly disease modifying in osteoarthritis.

Traditional view

In Ayurveda, Boswellia resin, known as Salai guggal, is mainly used as an astringent and anti-inflammatory agent (when applied topically) and as a stimulant and expectorant (internal use).1 Therapeutic applications include pulmonary diseases, especially if chronic, rheumatic disorders, diarrhoea, dysentery, piles, dysmenorrhoea, gonorrhoea, syphilis and liver disorders. It is also used for general weakness and to improve appetite.2,3

Summary actions

Anti-inflammatory, antiarthritic.

Can be used for

Indications supported by clinical trials

As an anti-inflammatory agent in asthma, inflammatory bowel disease (Crohn’s and ulcerative colitis), rheumatoid arthritis (mixed results) and osteoarthritis. There is some clinical evidence to suggest Boswellia may reduce oedema in patients with certain malignant brain tumours.

May also be used for

Extrapolation from pharmacological studies

Other conditions where leukotrienes and cytokines play an important role as inflammatory mediators. Disorders characterised by elevated levels of leukotrienes include cystic fibrosis, psoriasis, allergic rhinitis, lupus, gout, urticaria, liver cirrhosis, multiple sclerosis and chronic smoking. The value of Boswellia for these conditions has not been established and recently the clinical relevance of the in vitro inhibition of 5-lipoxygenase by boswellic acids has been questioned. Boswellia may have clinically significant anti-tumour activity.

Other applications

Its anti-inflammatory properties and ability to cross the blood-brain barrier (especially for beta-boswellic acid) suggest a possible role for Boswellia in the prevention of Alzheimer’s disease.

Preparations

The dried oleo-gum resin or a dry extract standardised for boswellic acids in tablet or capsule form. Boswellia resin requires a high content of alcohol for extraction (usually 90%) similar to myrrh and ginger. For this reason, Boswellia is more conveniently dispensed as a tablet or capsule rather than as a tincture, given the relatively high doses required.

Dosage

The dosage for Boswellia is 200 to 400 mg of extract three times a day, taken with meals. This extract is typically standardised to have a boswellic acid content of 60% to 70% and the dose corresponds to an equivalent resin intake of 2.4 to 4.8 g.

Duration of use

There are no known problems with long-term consumption. However, wherever possible the cause of the inflammation should be addressed, rather than relying only on palliation with Boswellia.

Summary assessment of safety

No major problems have been associated with the ingestion of normal doses and there are no documented interactions with conventional drugs. Typical adverse reactions occur with a low frequency and include diarrhoea and allergy.

Technical data

Botany

The Burseraceae family, to which both the Boswellia and Commiphora genera belong, contains trees or shrubs with prominent resin ducts in the bark. When the bark is cut, the secretion exudes and solidifies to a gum-like consistency after exposure to the air. Boswellia serrata is a deciduous, medium-sized tree native to the dry hills of India. The leaves are opposite, sessile and have serrated edges. Flowers occur in auxiliary racemes, shorter than the leaves, and are small, aromatic and white. The fruit is three-angled, splits into three valves and contains a single compressed seed.14

Adulteration

There are no documented adulterants, but presumably other species of Boswellia are possible candidates. The assessment of boswellic acid levels in standardised extracts is sometimes determined by simple acid-base titration. This leads to the possibility that a common food acid such as citric acid could be added as an adulterant to falsely elevate the level of boswellic acids measured by this means.

Key constituents

The oleo-gum-resin of Boswellia serrata contains pentacyclic triterpene acids (mainly beta-boswellic acid and the acetyl-boswellic acids: acetyl-beta-boswellic acid, acetyl-11-keto-beta-boswellic acid (AKBA) and 11-keto-beta-boswellic acid),5 and tetracyclic triterpene acids.6

Other constituents include an essential oil, terpenols, monosaccharides, uronic acids, sterols and phlobaphenes.7 The oils from Boswellia species consist of monoterpenes and sesqui-terpenes: alpha-thujene can be a major component of the oil obtained from Boswellia serrata.8 The tetracyclic triterpenoid 3-oxo-tirucallic acid (unrelated to the boswellic acids) has also been identified as a component of Boswellia.9

image

Pharmacodynamics

The relevant pharmacodynamics of Boswellia (including B. carterii) and the boswellic acids have been the subject of two comprehensive reviews.10,11

Anti-inflammatory and related activity

Laboratory studies on Boswellia extract and boswellic acids have demonstrated their ability to both prevent and relieve inflammation. Anti-inflammatory and antiarthritic activity was demonstrated in several models:12 carrageenan-, histamine- and dextran-induced oedema, carrageenan-induced pleurisy,13 adjuvant arthritis, formaldehyde- and bovine serum albumin (BSA)-induced arthritis14 and sodium urate gouty arthritis. Oral doses of Boswellia extract from 25 to 200 mg/kg were typically tested. The anti-inflammatory effect was not affected by removal of the adrenal glands, indicating the pituitary-adrenal axis was not involved. No significant activity occurred in cotton pellet-induced granuloma, which is more sensitive to steroidal than non-steroidal anti-inflammatory agents. In the developing adjuvant polyarthritis model, boswellic acids significantly reduced the development of secondary lesions. The reversal of syndromes of established arthritis indicated that it could control damage arising from immunological mechanisms.12

In adjuvant arthritis, administration of Boswellia extract or boswellic acids decreased the urinary excretion of connective tissue metabolites15 and had a beneficial effect on glycohydrolases and lysosomal stability.16,17 Treatment also reduced the degradation of glycosaminoglycans. From these results it is likely that Boswellia has beneficial effects in inflammatory conditions by suppressing proliferating tissue and preventing the degradation of connective tissue.18 Anti-inflammatory activity was also observed for B. carterii extract (450 to 900 mg/kg/day for 7 days, oral) in inflammation induced by complete Freund’s adjuvant.19 Topical application of a boswellic acid ointment demonstrated a dose-dependent anti-inflammatory effect in both acute and chronic experimental models, including croton-oil- induced mouse ear oedema and adjuvant arthritis in rats.20

Oral administration of boswellic acids reduced the development of 24 h delayed-type hypersensitivity reaction and complement fixing antibody in mice.21 Oral administration of boswellic acids, particularly AKBA, before treatment with galactosamine/endotoxin significantly reduced serum enzyme activity and potential liver damage in mice.22 (This is a model of increased leukotriene synthesis.) A pain-relieving activity was reported for Boswellia extract,23,24 but this finding was not confirmed in later studies.12 Weak antipyretic activity was, however, observed.12 AKBA (50 to 200 mg/kg, oral doses) produced a dose-dependent and significant analgesic effect in several different experimental models of nociception and potentiated the analgesic effect of selective cyclo-oxygenase inhibitors such as nimesulide.25

Conflicting results were observed in experimental models of gastrointestinal inflammation. One study found that Boswellia extract did not ameliorate symptoms of colitis in mice,26 whereas in another AKBA and high-dose Boswellia extract significantly reduced tissue injury in rats with experimental ileitis.27 AKBA was also found to confer protection in experimental murine colitis induced by dextran sodium sulphate, possibly by interfering with P-selectin-mediated recruitment of inflammatory cells.28

Boswellic acids were described as belonging to the group of slow-acting antiarthritic medications and inhibited latex-induced rat paw inflammation at oral doses of 50 to 100 mg/kg.29 Unlike conventional non-steroidal anti-inflammatory drugs (NSAIDs), ulcerogenic effects were not detected for Boswellia extract in animal models, and the extract even exhibited a protective effect on experimentally induced ulcers in rats.30 Other side effects of NSAIDs, such as prolonged gestation and parturition and diarrhoea, were not observed for the boswellic acids.12

Mechanism of action

The clinically relevant anti-inflammatory mechanisms for Boswellia are not fully understood. It appears to possess activity across a wide range of inflammatory diseases. While earlier studies have suggested the basis for anti-inflammatory activity is 5-lipoxygenase inhibition, this understanding has been challenged in recent times.

In early research, boswellic acids significantly inhibited the stimulated release of leukotrienes from intact human neutrophils, with AKBA being the most potent.31 Boswellia extract also reduced the formation of leukotrienes by inhibiting the enzyme 5-lipoxygenase in vitro.32

Boswellic acids appeared to exert a specific in vitro inhibitory activity on 5-lipoxygenase, with little effect on cyclo-oxygenase (which produces prostaglandins) or 12-lipoxygenase, although some inhibitory activity on both these enzymes has been recently found in human platelets.33 The mechanism of action is therefore quite distinct from conventional NSAIDs, which inhibit prostaglandin production. Compounds that inhibit 5-lipoxygenase usually do so because they are antioxidants. However, the action of the boswellic acids does not rely on antioxidant properties and for these reasons it is described as novel and specific.34,35 It was concluded that Boswellia could be a useful treatment in leukotriene-mediated inflammation and hypersensitivity-based disorders. Some degradation products of boswellic acids are also potent inhibitors of 5-lipoxygenase.36

There is a suggestion from in vitro research that the action of Boswellia on 5-lipoxygenase is biphasic.37 Concentrations greater than 10 to 15 µg/mL inhibited the enzyme in stimulated neutrophils, as noted previously. In contrast, lower concentrations of extracts (1 to 10 µg/mL) potentiated 5-lipoxygenase product formation. It was proposed in one study that this stimulatory effect on 5-lipoxygenase activity may be due (at least in part) to 3-oxo-tirucallic acid and related compounds, rather than the boswellic acids.38,39

A 2009 study found that while the 11-keto-boswellic acids efficiently suppressed 5-lipoxygenase activity in isolated neutrophils, this effect was absent in whole human blood.40 A single dose (800 mg) of Boswellia extract to 12 healthy male volunteers failed to suppress leukotriene B4 (a product of 5-lipoxygenase) in plasma. The authors suggested that, since leukotrienes do not play a major role in osteoarthritis and inflammatory bowel disease, a lack of clinical activity on 5-lipoxygenase is actually consistent with the observed value of Boswellia for these disorders. Their findings certainly question the relevance of 5-lipoxygenase inhibition to the pharmacological activity of Boswellia. However, unpublished results quoted in a recent review claimed inhibition of cysteine-leukotriene formation in stimulated granulocytes ex vivo from healthy humans given 1200 mg of a commercial Boswellia extract.11 The inhibitory effect was more than 90% with a maximum 6 h after intake, which corresponds with the known pharmacokinetics of boswellic acids.

That the anti-inflammatory mechanism of Boswellia is not confined to the inhibition of leukotriene formation was highlighted in a 2006 review.41 There is suggestion from in vitro studies that Boswellia can alter the production of pro-inflammatory cytokines. AKBA and acetyl-alpha-boswellic acid downregulated tumour necrosis factor alpha (TNF-alpha) in stimulated peripheral monocytes by inhibiting NF-kappaB signalling.42 A related species (Boswellia carterii) inhibited Th1 cytokines and promoted Th2 cytokines in vitro.43 One mechanism by which TNF-alpha causes inflammation is by potently inducing the expression of adhesion molecules such as VCAM-1 (vascular cell adhesion molecule 1). This was completely prevented by Boswellia extract in vitro for isolated human microvascular endothelial cells.44

Boswellic acids were also found to inhibit another pro-inflammatory enzyme in vitro: human leukocyte elastase (HLE). The dual inhibition of both 5-lipoxygenase and HLE is apparently unique to these compounds.45 They also possess anticomplementary activity in the classical and alternative complement pathways. A key enzyme of the classical complement pathway (C3-convertase) was inhibited.46,47 (Complement is a blood-based enzymatic cascade that serves to amplify, or complement, other inflammatory processes in the body. It is particularly implicated in the inflammation that occurs in rheumatoid arthritis.)

Like HLE, cathepsin G (catG) is another serine protease found in neutrophil granules that may participate in the breakdown of ingested pathogens and other inflammatory responses. CatG inhibitors have potential in the treatment of inflammatory conditions such as asthma, emphysema, psoriasis and rheumatoid arthritis.10 Boswellic acids exhibited a high and specific binding affinity for catG in vitro and potently suppressed the proteolytic activity of catG in a competitive and reversible manner.48 Related serine proteases were significantly less sensitive in vitro, including HLE, or not affected at all. Boswellic acids inhibited chemoinvasion but not chemotaxis of challenged neutrophils in vitro and suppressed catG induced calcium mobilisation in human platelets in vitro. Oral administration of a defined Boswellia extract to 12 healthy volunteers (800 mg single dose) and to five patients with Crohn’s disease (2400 mg/day) significantly lowered the catG activity of various blood samples.11 This research represents an intriguing development in the understanding of the anti-inflammatory activity of Boswellia.

Anti-allergic activity

An extract of Boswellia containing 60% AKBA was evaluated for anti-allergic and mast cell stabilising activity using passive rat paw anaphylaxis and induced degranulation of mast cells as the experimental in vivo models.49 The extract inhibited the anaphylaxis reaction in a dose-dependent manner at oral doses of 20, 40 and 80 mg/kg, but was not as strong as dexamethasone (0.27 mg/kg). Similarly, a dose-dependent inhibition of mast cell degranulation was observed at the above doses.

Anticancer activity

Boswellia extract and boswellic acids have shown anticancer activities in cell cultures, including inhibition of cell growth and DNA synthesis.50,51 The induction of differentiation and apoptosis (possibly due to topoisomerase I inhibition) suggests that boswellic acids may be useful in the treatment of leukaemia.5254

The ability of boswellic acids (such as boswellic acid acetate and AKBA) to induce in vitro apoptosis has been demonstrated in the following tumour cell lines: myeloid leukaemia cells,55 metastatic melanoma and fibrosarcoma cells,56 various leukaemia, haematological and brain tumour cell lines,57 colon cancer cells,58 liver cancer Hep G2 cells59 and malignant glioma.60 In addition AKBA was found to be cytotoxic towards meningioma cells in vitro.61,62 A useful and extensive review of the anticancer activity of the boswellic acids is available,63 as is a discussion of their molecular targets in this context.64

Topical application of Boswellia extract with the tumour promoter 12-O-tetradecanoylphorbol-13-acetate (TPA) inhibited the expected formation of skin tumours in mice.65,66 Boswellic acids have also been shown to inhibit tumour growth in vivo using a rat brain tumour model (glioma), albeit at quite high doses (720 mg/kg boswellic acids).67 The effect was dose dependent. In mice carrying prostate cancer cell tumours, systemic doses of AKBA inhibited tumour growth and triggered apoptosis in the absence of systemic toxicity.68

Other activity

Boswellia resin demonstrated lipid lowering activity in vivo.6971 It is interesting that another Ayurvedic remedy, the resin from a plant in the same family, Commiphora mukul, has demonstrated hypocholesterolaemic activity both in vivo and in clinical trials, although a recent trial has cast doubt on its clinical efficacy.72

Oral doses of AKBA (100 mg/kg) and nimesulide (2.42 mg/kg) for 15 days significantly reversed the age-induced deterioration of memory, cognitive performance and meta-function in mice.73

The 11-methylene analogue of 11-keto-boswellic acid (KBA), namely beta-boswellic acid, potently induced calcium mobilisation in platelets in vitro.74 Pivotal protein kinases were activated, resulting in functional platelet responses such as thrombin generation, liberation of arachidonic acid and aggregation.

Pharmacokinetics

Boswellic acids have also been shown to cross the blood-brain barrier in rats.75 Permeation studies of Boswellia extract in the in vitro Caco-2 model of intestinal absorption found poor permeability for AKBA and moderate absorption of KBA.76 Most of these compounds were retained in the Caco-2 monolayer. In rat liver microsomes and hepatocytes, as well as in human liver microsomes, KBA but not AKBA, underwent extensive phase I metabolism.77 This was verified in vivo, where it was found that KBA undergoes extensive first-pass metabolism, whereas AKBA does not. Hence, metabolism (e.g. deacetylation) is not mainly responsible for the relatively low bioavailability of AKBA.

The human bioavailability of boswellic acids has been established in several pharmacokinetic studies. These indicate that beta-boswellic acid exhibits relatively better bioavailability than KBA and AKBA. Twelve healthy adult men were given capsules containing 333 mg of Boswellia extract after a 7-day washout period.78 Venous blood samples, drawn at various times after administration of the herb, were analysed for KBA. A mean peak plasma level of 2.72±0.18 µM was reached at 4.50±0.55 h, with an elimination half-life of 5.97±0.95 h. These results suggested that Boswellia is best taken orally every 6 h and that this should achieve steady-state plasma levels after approximately 30 h.

In a randomised, open, single-dose, two-way crossover study, 12 healthy male volunteers received 786 mg of Boswellia extract either with or without a standard high-fat meal.79 Plasma concentrations of boswellic acids were measured up to 60 h after oral dosing. Administration in conjunction with a high-fat meal led to a substantial improvement in the bioavailability of the boswellic acids. For example, the maximum concentration for AKBA and KBA respectively was 6.0 and 83.8 ng/mL for the fasted conditions versus 28.8 and 227 ng/mL with food. However, as might be expected, the time at which this and other maxima were reached was delayed by the meal. In contrast, a pilot study involving six healthy volunteers found an average concentration of KBA of 43 ng/mL 2 h after the administration of 500 mg of a Boswellia extract.80 No relationship to meals was specified.

Steady-state concentrations of boswellic acids in the plasma of Crohn’s disease patients receiving 2400 mg/day of a Boswellia extract were 6.35±1.0 µM for beta-boswellic acid, 0.33±0.1 µM for KBA and 0.04±0.01 µM for AKBA.11

As part of a method validation process, 10 different boswellic acids were found in the plasma of a brain tumour patient (glioblastoma multiforme) who took 3144 mg/day of Boswellia extract for 10 days.81 The highest concentration was found for beta-boswellic acid at 10.1 µM.

Clinical trials

Arthritis

A significant improvement in symptoms was observed for 60 rheumatoid arthritis (RA) patients after receiving 6 to 8 weeks of treatment with boswellic acids in an open label study.82 After reviewing the successful results of preclinical toxicology and efficacy studies, an uncontrolled clinical trial was undertaken at the orthopaedic department of Government Medical College, Jammu, India.83,84 Results for 175 RA patients were ‘excellent’ for 14% and ‘good’ for 44%. Most patients taking Boswellia had some improvement in symptoms such as pain, stiffness and poor grip strength. Improvement occurred after 2 to 4 weeks of treatment.

In another uncontrolled study, Boswellia extract was given to 30 patients with RA.85 The percentage of patients with detectable C-reactive protein was 63% initially, but this decreased to 47% after 6 months of treatment, a significant finding. This suggests that Boswellia may have a disease-modifying effect in RA.

The clinical efficacy of a herbomineral formulation containing Withania somnifera, Boswellia serrata, Curcuma longa (turmeric) and a zinc complex was evaluated in a randomised, double blind, placebo-controlled, crossover study in 42 patients with osteoarthritis.86 The treatment period was 3 months, with a 15-day washout period before crossover and then another 3 months of treatment. Clinical efficacy was evaluated every fortnight on the basis of such findings as severity of pain, morning stiffness, joint score, disability score and grip strength. Treatment with the herbomineral formulation produced a significant drop in pain (p<0.001) and disability (p<0.05). However, X-ray assessment of joints did not show any significant changes.

The above herbomineral formulation was also evaluated in a randomised, double blind, placebo-controlled, cross-over trial of 20 RA patients for a period of 3 months. Significantly greater relief of pain, decreased morning stiffness, Ritchie Articular Index, joint score and erythrocyte sedimentation rate (ESR) were observed for those treated with the product. Seroconversion occurred for most of the treated patients, but radiological assessment did not show significant changes in either group.87

Etzel in 1996 reviewed 12 controlled clinical trials conducted in Germany or India (including those cited above) that assessed the effects of Boswellia in the treatment of RA.88 Of these studies, only two were double blind, placebo-controlled trials, so results were difficult to interpret overall. However, summarising the results of different studies, Etzel suggested the following:

• There was a benefit from Boswellia treatment over placebo in patients suffering from RA for several years and in patients who responded poorly to conventional treatment.

• Boswellia was apparently as effective as gold therapy for RA.

• Some children suffering from chronic juvenile arthritis particularly benefited from Boswellia treatment.

• Tolerance was good and side effects were mild, such as diarrhoea and urticaria.

These positive outcomes contrast with a subsequent German study that found, under double blind, placebo-controlled conditions, that Boswellia was not more effective than placebo in patients with chronic polyarthritis (presumably mainly RA).89 The authors of this study reserved their judgement, suggesting that a larger trial than the one they conducted was necessary to confirm or reject the use of Boswellia in RA sufferers, especially since the dropout rate in their trial was quite high.

An extract of Boswellia standardised to 40% boswellic acids by HPLC (high-performance liquid chromatography) yielded good results in the treatment of osteoarthritis.90 A randomised, double blind, placebo-controlled, crossover study was conducted to assess the efficacy, safety and tolerability of the Boswellia extract (1000 mg/day) in 30 patients with osteoarthritis of the knee, 15 each receiving active treatment or placebo for 8 weeks. After the first intervention, washout was given and then the groups were crossed over to receive the opposite intervention for 8 weeks. All patients receiving herbal treatment reported a significant decrease in knee pain, increased knee flexion and increased walking distance. The frequency of swelling in the knee joint was substantially decreased, but radiologically there was no change. Boswellia was well tolerated by the patients, except for minor gastrointestinal adverse reactions.

Since the boswellic acids were considered to be specific, non-redox inhibitors of 5-lipoxygenase and hence leukotriene biosynthesis, research attention as noted above had previously focused on the effects of Boswellia in inflammatory joint diseases such as RA. Despite this research focus on RA, herbal clinicians also favoured the use of Boswellia in osteoarthritis. The results of the above trial provided the first good evidence to support this application. What was striking about the trial was the substantial clinical benefit observed. Results were highly statistically significant (p<0.001) and changes in treatment parameters were quite large. For example, in the first 8-week treatment period before crossover, the pain index in the Boswellia group fell from 2.7±0.45 to 0.26±0.45, the loss of movement index was reduced from 2.8±0.41 to 0.30±0.48 and the swelling index went from 1.1±0.91 to zero. The group receiving the placebo treatment after crossover showed substantial deterioration over the ensuing 8 weeks, suggesting that the 21-day washout period before crossover was insufficient. Although this is a flaw in the experimental design, it suggests a substantial residual therapeutic benefit after stopping Boswellia.

A prospective, randomised study conducted in India assessed the efficacy and tolerability of Boswellia extract in comparison to valdecoxib, a selective COX-2 inhibitor.91 Sixty-six patients aged between 40 and 70 years with primary osteoarthritis of the knee, diagnosed according to the criteria of the American College of Rheumatology, were recruited. X-rays confirmed the diagnosis. Patients were assessed by the WOMAC scale at baseline and at monthly intervals until 1 month after discontinuation of 6 months’ therapy. The WOMAC (Western Ontario and McMaster Universities Osteoarthritis Index) scale consists of questions based on the three symptoms (pain, stiffness and difficulty in performing daily activities), to which the patient assigns a number between 0 and 100. Patients received either 1000 mg standardised Boswellia extract (containing 40% boswellic acids, which corresponded to a dose of 400 mg/day of boswellic acids) or valdecoxib 10 mg/day. There were 33 patients in each treatment group. During the trial patients were permitted to continue receiving physiotherapy and take a rescue medication (ibuprofen).

Results indicated that Boswellia extract was as effective as valdecoxib for knee osteoarthritis. In comparison with valdecoxib, Boswellia had a slower onset of action but the effect persisted after the discontinuation of therapy (unlike valdecoxib). The implications of this trial are that Boswellia is effective for knee osteoarthritis but needs to be taken for at least 2 months to establish the full clinical effect. Furthermore, the slow onset and slow washout of benefit suggests it could be disease modifying.

In a trial published in 2008, 75 patients with knee osteoarthritis received either Boswellia extract (containing 100 or 250 mg/day of an AKBA-enriched selective boswellic acid extract) or placebo for 90 days. Boswellia conferred a clinically and statistically significant dose-response improvement in pain and physical function scores. Symptom alleviation was faster in the higher dose Boswellia group (as early as 7 days) and a significant reduction in synovial fluid levels of matrix metalloproteinase-3 (a cartilage-degrading enzyme) was also observed for the Boswellia groups.92

A later study evaluated the activity of this AKBA-enriched selective boswellic acid extract at 100 mg/day against a similar extract with enhanced bioavailability (also at 100 mg/day) and a placebo in 60 patients with knee osteoarthritis.93 A double blind, randomised, placebo-controlled design was used, with a trial period of 90 days. At the end of the study, both Boswellia extracts conferred clinically and statistically significant improvements in pain and physical function scores versus placebo (various p values). Significant improvements were recorded as early as 7 days for the enhanced bioavailability extract, which demonstrated better activity than the other extract. By 90 days, some clinical changes for the enhanced bioavailability extract were particularly marked. For example, the WOMAC pain subscale fell by 69% (p<0.0001 versus placebo) and the WOMAC stiffness subscale by 70.1% (p<0.0001 versus placebo) from baseline.

A formulation containing Withania, Boswellia, turmeric and ginger was shown to be effective in osteoarthritis in a double blind, randomised, placebo-controlled trial involving 99 patients.94 Concurrent NSAIDs or analgesics were not permitted. Significant improvements were noted for pain (visual analogue scale) (p<0.05) and a modified WOMAC score (p<0.01) by the end of trial (32 weeks). There was no significant difference in type and number of adverse effects between active and placebo, although 28 patients did not complete the trial.

Ulcerative colitis

A group of Indian and German scientists conducted an open, non-randomised pilot study to test the hypothesis that Boswellia might be beneficial for the treatment of ulcerative colitis. For the study, 34 patients (18 with grade II and 16 with grade III colitis) were given 350 mg of Boswellia resin three times a day, and eight patients (five with grade II and three with grade III colitis) were given the drug sulfasalazine, 1 g three times a day. The authors commented that the cost of the drug was responsible for the small control group. Symptoms such as abdominal pain, loose stool, mucus and blood improved in both groups, with results for the control sulfasalazine group slightly better than the Boswellia group. Sigmoidoscopic examination for the grading of the disease and rectal biopsies both showed substantial improvements in the two treatment groups, and there was no statistically significant difference between them. About 80% of the patients receiving Boswellia went into remission. The authors concluded that, although they could not statistically confirm the superiority of Boswellia over sulfasalazine, the data suggest that it is at least not inferior.95

Twenty patients with chronic colitis received Boswellia gum resin (900 mg/day for 6 weeks) and another 10 patients were given sulfasalazine (3 g/day for 6 weeks) in an open label trial.96 Out of 20 patients treated with Boswellia, 18 went into remission (90% compared with 60% for sulfasalazine).

Asthma

In a double blind, placebo-controlled clinical study, 80 patients with chronic asthma were treated with 900 mg/day of Boswellia, gum resin or placebo for 6 weeks.97 Only 27% of patients in the control group showed improvement, whereas 70% of patients taking Boswellia improved. After taking Boswellia, positive changes were observed for shortness of breath, number of attacks and respiratory capacity as well as indicators of inflammation. Comparing the Boswellia group with the placebo group, there was a significant improvement in forced expiratory volume (in 1 second), a measure of bronchial obstruction (p=0.0001). Peak expiratory flow rate, a measure of lung capacity, was also significantly increased (p=0.0001). The number of asthma attacks was lower in the Boswellia group (p=0.0001). Additionally, Boswellia treatment showed substantial improvements (compared with placebo) in secondary outcome parameters such as rhonchi, eosinophil count, ESR and respiratory rate (p<0.05). Two patients who received Boswellia complained of epigastric pain and nausea.

It should not be assumed that inhibition of leukotrienes is the only mechanism by which Boswellia might provide beneficial effects for asthma, especially given the antiallergic activity from experimental models.

Crohn’s disease

The safety and efficacy of a Boswellia extract was compared against mesalazine for the treatment of 102 patients with active Crohn’s disease in an 8-week randomised, double blind European study.98 The primary clinical outcome measured was the change in Crohn’s Disease Activity Index (CDAI). After therapy with Boswellia extract (3.6 g/day) the average CDAI was reduced by 90, compared with a reduction of 53 for the mesalazine group (4.5 g/day). The authors concluded that the Boswellia extract was as effective as mesalazine, which is the current anti-inflammatory treatment for this disorder. Considering the observed fewer side effects and better safety profile of Boswellia, they suggested that this novel herbal treatment appears to be superior to mesalazine in terms of a risk-benefit evaluation.

In contrast, a 52-week double blind, placebo-controlled randomised trial in 108 patients with Crohn’s disease found 2400 mg/day of a Boswellia extract was not superior to placebo in terms of maintaining remission.99 However, only 66 patients completed the trial due to early termination and the extract did not seem to deliver high plasma levels of boswellic acids relative to other studies (see under Pharmacokinetics).11 Boswellia treatment was well tolerated.

Brain tumours

Malignant brain tumours produce highly active forms of leukotrienes and this causes localised fluid build-up in the brain around the tumour, which damages healthy nerve cells. Twelve patients with malignant glioma, a type of brain tumour, were given 3600 mg/day of Boswellia extract (standardised to 60% boswellic acids) for 7 days prior to surgery.100 Ten patients showed a decrease in fluid around the tumour, with an average reduction of 30% in eight of the 12 patients. Signs of brain damage decreased during the treatment; one patient became worse. Vomiting as a side effect was observed in one patient. This resulted in the European Commission declaring Boswellia as an orphan drug (a drug with no sponsors to fund the registration process) for the treatment of oedema resulting from brain tumours.75

Nineteen children and adolescents with intracranial tumours received palliative therapy with Boswellia extract at a maximum dose of 126 mg/kg/day for up to 26 months.101 All patients were previously treated with conventional therapy. An anti-oedematous effect was demonstrated by MRI in one patient. Five of the 19 children reported an improvement of general health (perhaps a placebo effect). Some objective improvement, sometimes transient, was observed in seven patients.

Twelve patients with brain tumours and progressive oedema caused by either the tumour or treatment were given 3600 mg/day Boswellia extract for 4 weeks.102 Oedema was reduced in five patients. Of five patients with treatment-related leukoencephalopathy, the clinical improvement following Boswellia was sustained for several months.

Forty-four patients with primary or secondary malignant cerebral tumours were randomly assigned to radiotherapy plus either 4200 mg/day Boswellia extract or placebo in a double blind trial.103 Compared with baseline and measured immediately after the end of radiotherapy, a greater than 75% reduction in cerebral oedema was observed in 60% of the patients receiving Boswellia versus 26% receiving placebo (p=0.023). The dexamethasone dose during radiotherapy did not significantly differ between groups. The tumour/oedema volume ratio decreased only in the Boswellia group, suggesting an antitumour effect in addition to the anti-oedema activity. However, progression-free survival did not differ between the groups. Nonetheless, the better tumour response to radiotherapy was an unexpected finding. Common adverse events associated with radiotherapy were similar in both groups, although gastrointestinal discomfort was probably higher in the Boswellia group. While boswellic acids could be detected in patients’ serum, the level of AKBA was quite low and could not be detected. Compliance with the high dose of extract used was an issue.

Other conditions

Collagenous colitis is a form of microscopic colitis of unknown aetiology and pathogenesis. The main symptom is chronic watery diarrhoea with few or no endoscopic abnormalities. Patients (n=31) with proven collagenous colitis were randomised to receive either 400 mg of Boswellia extract three times daily or placebo for 6 weeks in a double blind, randomised placebo-controlled trial.104 Clinical remission at the end of the trial was higher in the Boswellia group (64% versus 26% for protocol, p=0.04; 44% versus 27% intention-to-treat, p=0.25). The authors concluded that results were promising and larger trials are necessary to confirm any benefit from Boswellia for this disorder.

A multicentre veterinary clinical trial was conducted by 10 practising veterinarians in Switzerland involving 29 dogs with degenerative chronic joint and spinal osteoarthritis.105 An extract of Boswellia was administered in food at a dose of 400 mg/10 kg/day over 6 weeks. A statistically significant reduction in signs such as intermittent lameness and stiff gait were reported after 6 weeks. In five dogs reversible brief episodes of diarrhoea and flatulence occurred, but only in one case was a relationship to the Boswellia treatment suspected.

Toxicology and other safety data

Toxicology

Toxicity studies have generally shown that boswellic acids possess very low acute toxicity and cause no adverse effects after chronic administration. The oral and intraperitoneal LD50 was greater than 2 g/kg in mice and rats. No significant changes were observed in general behaviour, or in clinical, haematological, biochemical and pathological data after chronic oral administration.106 A Boswellia extract enriched with 30% AKBA exhibited an oral LD50 >5 g/kg in rats and was classified as non-irritant to the skin.107 A subacute toxicity study of the same extract over 90 days at up to 2.5% of feed indicated no adverse effects. However, one study involving experimentally induced colitis in mice found hepatotoxic effects for a methanolic extract at 1% of feed for 21 days, which was also supported by in vitro data.26

Contraindications

None known.

Special warnings and precautions

Caution in patients with a known allergic tendency. This is based on the fact that other resinous herbs such as myrrh are known to cause various allergic reactions and that allergic reactions have been reported for Boswellia (see below).

Interactions

No interactions have been reported in the literature.

Extracts of the oleo-gum, resin of Boswellia carterii, Boswellia frereana, Boswellia sacra and Boswellia serrata, were identified as equally potent, non-selective inhibitors of the major drug-metabolising CYP enzymes 1A2/2C8/2C9/2C19/2D6 and 3A4 in vitro.108 Although the boswellic acids could be identified as moderate to potent inhibitors of the applied CYP enzymes, they were not the major CYP inhibitory principles. The clinical significance of these in vitro findings is uncertain.

Use in pregnancy and lactation

Category B1 – no increase in frequency of malformation or other harmful effects on the fetus from limited use in women. No evidence of increased fetal damage in animal studies.

Treatment of pregnant rats with a boswellic acid fraction (250 to 1000 mg/kg, oral route) revealed no significant effects on gestation period, litter size and weight of offspring at birth. No gross morphological or skeletal abnormalities were recorded. This generation of offspring in turn produced a normal number of normal offspring.106

Boswellia is likely to be compatible with breastfeeding.

Effects on ability to drive and use machines

No adverse effects are expected.

Side effects

Contact dermatitis has been caused by Boswellia spp.109 Boswellia was well tolerated in clinical trials for the treatment of RA and Crohn’s disease. Very mild side effects such as diarrhoea and urticaria were reported.88

Overdosage

No incidents found in the published literature.

Safety in children

No information available but adverse effects are not expected.

Regulatory status in selected countries

In the UK Boswellia is not included on the General Sale List and it was not included in the Commission E assessment in Germany.

Boswellia does not have GRAS status in the USA. However, it is freely available as a ‘dietary supplement’ in the USA under DSHEA legislation (Dietary Supplement Health and Education Act of 1994).

In Australia Boswellia is not included in Part 4 of Schedule 4 of the Therapeutic Goods Regulations and is freely available for sale.

References

1. Thakur RS, Puri HS, Husain A. Major Medicinal Plants of India. Lucknow: Central Institute of Medicinal and Aromatic Plants, 1989. p. 123

2. Kapoor LD. CRC Handbook of Ayurvedic Medicinal Plant. Boca Raton: CRC Press, 1990. p. 83

3. Bharatiya Vidya Bhavan’s Swami Prakashananda Ayurveda Research Centre. Selected Medicinal Plants of India, Bombay, Chemexcil, 1992;67–68.

4. Mabberley DJ, ed. The Plant Book. Cambridge: Cambridge University Press, 1997. pp. 99, 107

5. Pardhy RS, Bhattacharyya SC. Indian J Chem Sect B. 1978;16(3):176–178.

6. Pardhy RS, Bhattacharyya SC. Indian J Chem Sect B. 1978;16(3):174–175.

7. Ammon HPT. Eur J Med Res. 1996;1(8):369–370.

8. Lawrence BM. Progress in essential oils. In Essential Oils 1992–1994. Carol Stream: Allured Publishing Corporation; 1995. pp. 20–23

9. Boden SE, Schweizer S, Bertsche T, et al. Mol Pharmacol. 2001;60(2):267–273.

10. Abdel-Tawab M, Werz O, Schubert-Zsilavecz M. Clin Pharmacokinet. 2011;50(6):349–369.

11. Ammon HP. Phytomedicine. 2010;17(11):862–867.

12. Singh GB, Singh S, Bani S. Phytomedicine. 1996;3(1):81–85.

13. Sharma ML, Khajuria A, Kaul A, et al. Agents Actions. 1988;24(1–2):161–164.

14. Sharma ML, Bani S, Singh BG. Int J Immunopharmacol. 1989;11(6):647–652.

15. Kesava Reddy G, Dhar SC, Singh GB. Agents Actions. 1987;22(1–2):99–105.

16. Kesava Reddy G, Dhar SC, Singh GB. Leather Sci. 1986;33:192–199.

17. Kesava Reddy G, Dhar SC. Ital J Biochem. 1987;6(4):205–217.

18. Kesava Reddy G, Chandrakasan G, Dhar SC. Biochem Pharmacol. 1989;38(20):3527–3534.

19. Fan AY, Lao L, Zhang RX, et al. J Altern Complement Med. 2005;11(2):323–331.

20. Singh S, Khajuria A, Taneja SC, et al. Phytomedicine. 2008;15(6–7):400–407.

21. Sharma ML, Kaul A, Khajuria A, et al. Phytother Res. 1996;10(2):107–112.

22. Safayhi H, Mack T, Ammon HPT. Biochem Pharmacol. 1991;41(10):1536–1537.

23. Kar A, Menon MK. Life Sci. 1969;8(19):1023–1028.

24. Menon MK, Kar A. Planta Med. 1971;19(4):333–341.

25. Bishnoi M, Patil CS, Kumar A, et al. Indian J Exp Biol. 2006;44(2):128–132.

26. Kiela PR, Midura AJ, Kuscuoglu N, et al. Am J Physiol Gastrointest Liver Physiol. 2005;288(4):G798–G808.

27. Krieglstein CF, Anthoni C, Rijcken EJ, et al. Int J Colorectal Dis. 2001;16(2):88–95.

28. Anthoni C, Laukoetter MG, Rijcken E, et al. Am J Physiol Gastrointest Liver Physiol. 2006;290(6):G1131–G1137.

29. Gupta OP, Sharma N, Chand D. J Pharmacol Toxicol Methods. 1994;31(2):95–98.

30. Singh S, Khajuria A, Taneja SC, et al. Phytomedicine. 2008;15(6–7):408–415.

31. Wildfeuer A, Neu IS, Safayhi H, et al. Arzneimittelforschung. 1998;48(6):668–674.

32. Ammon HP, Mack T, Singh GB, et al. Planta Med. 1991;57(3):203–207.

33. Siemoneit U, Hofmann B, Kather N, et al. Biochem Pharmacol. 2008;75(2):503–513.

34. Safayhi H, Mack T, Sabieraj J, et al. J Pharmacol Exp Ther. 1992;261(3):1143–1146.

35. Safayhi H, Sailer ER, Ammon HP. Mol Pharmacol. 1995;47(6):1212–1216.

36. Schweizer S, von Brocke AF, Boden SE, et al. J Nat Prod. 2000;63(8):1058–1061.

37. Safayhi H, Boden SE, Schweizer S, et al. Planta Med. 2000;66(2):110–113.

38. Altmann A, Poeckel D, Fischer L, et al. Br J Pharmacol. 2004;141(2):223–232.

39. Altmann A, Fischer L, Schubert-Zsilavecz M, et al. Biochem Biophys Res Commun. 2002;290(1):185–190.

40. Siemoneit U, Pergola C, Jazzar B, et al. Eur J Pharmacol. 2009;606(1–3):246–254.

41. Ammon HP. Planta Med. 2006;72(12):1100–1116.

42. Syrovets T, Buchele B, Krauss C, et al. J Immunol. 2005;274(1):498–506.

43. Chevrier MR, Ryan AE, Lee DY, et al. Clin Diagn Lab Immunol. 2005;12(5):575–580.

44. Roy S, Khanna S, Shah H, et al. DNA Cell Biol. 2005;24(4):244–255.

45. Safayhi H, Rall B, Sailer ER, et al. J Pharmacol Exp Ther. 1997;281(1):460–463.

46. Kapil A, Moza N. Int J Immunopharmacol. 1992;14(7):1139–1143.

47. Knaus U, Wagner H. Phytomedicine. 1996;3(1):77–81.

48. Tausch L, Henkel A, Siemoneit U, et al. J Immunol. 2009;183(5):3433–3442.

49. Pungle P, Banavalikar M, Suthar A, et al. Indian J Exp Biol. 2003;41(12):1460–1462.

50. Han R. Stem Cells. 1994;12(1):53–63.

51. Huang M-T, Shao Y, Ma W, et al. Proc Ann Meet Am Assoc Cancer Res. 1997;38:A2465.

52. Jing Y, Nakajo S, Xia L, et al. Leuk Res. 1999;23(1):43–50.

53. Hoernlein RF, Orlikowsky T, Zehrer C, et al. J Pharmacol Exp Ther. 1999;288(2):613–619.

54. Syrovets T, Buchele B, Gedig E, et al. Mol Pharmacol. 2000;58(1):71–81.

55. Xia L, Chen D, Han R, et al. Mol Cancer Ther. 2005;4(3):381–388.

56. Zhao W, Entschladen F, Liu H, et al. Cancer Detect Prev. 2003;27(1):67–75.

57. Hostanska K, Daum G, Saller R. Anticancer Res. 2002;22(5):2853–2862.

58. Liu JJ, Nilsson A, Oredsson S, et al. Carcinogenesis. 2002;23(12):2087–2093.

59. Liu JJ, Nilsson A, Oredsson S, et al. Int J Mol Med. 2002;10(4):501–505.

60. Glaser T, Winter S, Groscurth P, et al. Br J Cancer. 1999;80(5–6):756–765.

61. Park YS, Lee JH, Harwalkar JA, et al. Adv Exp Med Biol. 2002;507:387–393.

62. Park YS, Lee JH, Bondar J, et al. Planta Med. 2002;68(5):397–401.

63. Shah BA, Qazi GN, Taneja SC. Nat Prod Rep. 2009;26(1):72–89.

64. Poeckel D, Werz O. Curr Med Chem. 2006;13(28):3359–3369.

65. Huang M-T, Badmaev V, Xie J-G, et al. Proc Ann Meet Am Assoc Cancer Res. 1997;8:A2464.

66. Huang MT, Badmaev V, Ding Y, et al. Biofactors. 2000;13(1–4):225–230.

67. Winking M, Sarikaya S, Rahmanian A, et al. J Neurooncol. 2000;46(2):97–103.

68. Syrovets T, Gschwend JE, Buchele B, et al. J Biol Chem. 2005;280(7):6170–6180.

69. Zutshi U, Rao PG, Kaur S, et al. Indian J Pharmacol. 1986;18(3):182–183.

70. Singh GB, Singh S. J Molec Cell Cardiol. 1994;25(suppl 3):188.

71. Pandey RS, Singh BK, Tripathi YB. Indian J Exp Biol. 2005;43(6):509–516.

72. Szapary PO, Wolfe ML, Bloedon LT, et al. JAMA. 2003;290(6):765–772.

73. Bishnoi M, Patil CS, Kumar A, et al. Methods Find Exp Clin Pharmacol. 2005;27(7):465–470.

74. Poeckel D, Tausch L, Altmann A, et al. Br J Pharmacol. 2005;146(4):514–524.

75. Reising K, Meins J, Bastian B, et al. Anal Chem. 2005;77(20):6640–6645.

76. Krüger P, Kanzer J, Hummel J, et al. Eur J Pharm Sci. 2009;36(2–3):275–284.

77. Krüger P, Daneshfar R, Eckert GP, et al. Drug Metab Dispos. 2008;36(6):1135–1142.

78. Sharma S, Thawani V, Hingorani L, et al. Phytomedicine. 2004;11(2–3):255–260.

79. Sterk V, Buchele B, Simmet T. Planta Med. 2004;70(12):1155–1160.

80. Shah SA, Rathod IS, Suhagia BN, et al. J Chromatogr B. 2007;848(2):232–238.

81. Büchele B, Simmet T. J Chromatogr B. 2003;795(2):pp. 355–pp. 362.

82. Singh, Hardayal, Singh GP, et al. Abstract presented at International Conference on Clinical Pharmacology and Therapeutics, Bombay: 1987; pp. 20–22.

83. Proceedings of the Symposium of Recent Advances in Mediators of Inflammation and Anti-Inflammatory Agents. Jammu: Council of Scientific and Industrial Research, Regional Research Laboratory; 1984.

84. Gupta VN, Yadav DS, Jain MP, et al. Indian Drugs. 1987;24(5):221–231.

85. Eleventh European Congress of Rheumatology. 1987; 5 (supplement issue):175.

86. Kulkarni RR, Patki PS, Jog VP, et al. J Ethnopharmacol. 1991;33(1–2):91–95.

87. Kulkarni RR, Patki PS, Jog VP, et al. Indian J Pharmacol. 1992;24(2):98–101.

88. Etzel R. Phytomed. 1996;3(1):91–94.

89. Sander O, Herborn G, Rau R. Z Rheumatol. 1998;57(1):11–16.

90. Kimmatkar N, Thawani V, Hingorani L, et al. Phytomedicine. 2003;10(1):3–7.

91. Sontakke S, Thawani V, Pimpalkhute S, et al. Indian J Pharmacology. 2007;39(1):27–29.

92. Sengupta K, Alluri KV, Satish AR, et al. Arthritis Res Ther. 2008;10:R85.

93. Sengupta K, Krishnaraju AV, Vishal AA, et al. Int J Med Sci. 2010;7(6):366–377.

94. Chopra A, Lavin P, Patwardhan B, et al. J Clin Rheumatol. 2004;10(5):236–245.

95. Gupta I, Parihar A, Malhotra P, et al. Eur J Med Res. 1997;2(1):37–43.

96. Gupta I, Parihar A, Malhotra P, et al. Planta Med. 2001;67(5):391–395.

97. Gupta I, Gupta V, Parihar A, et al. Eur J Med Res. 1998;3(11):511–514.

98. Gerhardt H, Seifert F, Buvari P, et al. Z Phytother. 2001;22:69–75.

99. Holtmeier W, Zeuzem S, Preiss J, et al. Inflamm Bowel Dis. 2011;17(2):573–582.

100. Winking M, Boeker DK, Simmet TH. J Neurooncol. 1996;30(2):P39.

101. Janssen G, Bode U, Breu H, et al. Klin Padiatr. 2000;212(4):189–195.

102. Streffer JR, Bitzer M, Schabet M, et al. Neurology. 2001;56(9):1219–1221.

103. Kirste S, Treier M, Wehrle SJ, et al. Cancer. 2011;117(16):3788–3795.

104. Madisch A, Miehlke S, Eichele O, et al. Int J Colorectal Dis. 2007;22(12):1445–1451.

105. Reichling J, Schmokel H, Fitzi J, et al. Schweiz Arch Tierheilkd. 2004;146(2):71–79.

106. Singh GB, Bani S, Singh S. Phytomedicine. 1996;3(1):87–90.

107. Lalithakumari K, Krishnaraju A, Sengupta K, et al. Toxicol Mech Methods. 2006;16(4):199–226.

108. Frank A, Unger M. J Chromatogr A. 2005;1112(1–2):255–262.

109. Mitchell J, Rook A. Botanical Dermatology: Plants and Plant Products Injurious to the Skin. Vancouver: Greengrass, 1979. pp. 144–145

Buchu

(Agathosma betulina (Bergius) Pill.)

Synonyms

Barosma betulina (Bergius) Bartl. et Wendl. (botanical synonym), bucco (Engl), Barosmae folium (Lat), Bukkostrauch, Buccoblätter (Ger), buchu (Fr), diosma (Ital), bukko (Dan).

What is it?

Buchu is a South African herb extensively employed as a urinary antiseptic and may also be used in laxative and carminative formulas. The Hottentots use buchu leaves to perfume their bodies. (The Agathosma genus is native to South Africa, especially the southwest Cape region.) The essential oil is a component of artificial fruit flavours, especially blackcurrant, and it may be found in trace amounts in a wide variety of food products and beverages.

The leaves are the part most commonly used for therapeutic purposes and should be harvested whilst the plant is flowering and fruiting. They possess a strongly aromatic taste and a curious blackcurrant-peppermint-like odour.

Effects

Disinfects the urinary tract and acts as a mild diuretic.

Traditional view

Buchu was used to treat gravel, inflammation and catarrh of the bladder.1 According to the Eclectics, buchu is an aromatic stimulant and tonic that promotes the appetite, relieves nausea and flatulence, and acts as a diuretic and diaphoretic. Its principal use was in the treatment of chronic diseases of the genitourinary tract including chronic inflammation of the mucous membranes of the bladder, irritable conditions of the urethra, for urinary discharges (particularly mucopurulent discharges), abnormally acidic urine with a constant desire to urinate and little relief from micturition, and incontinence associated with a diseased prostate.2

Summary actions

Urinary antiseptic, mild diuretic.

Can be used for

Traditional therapeutic uses

Urinary tract infection, dysuria, cystitis, urethritis and prostatitis.

May also be used for

Other applications

Buchu may be included as an aroma or taste enhancer in herbal tea mixtures.3

Preparations

Dried leaf as a decoction, liquid extract, tincture, tablets or capsules for internal use. As with all essential oil-containing herbs, use of the fresh plant or carefully dried herb is advised. Keep covered if infusing the herb to retain the essential oil.

Dosage

• 3 to 6 g/day of the dried leaf or as an infusion

• 2 to 4 mL/day of 1:2 liquid extract, 5 to 10 mL/day of 1:5 tincture.

Duration of use

May be taken long term for most applications.

Summary assessment of safety

May occasionally cause gastrointestinal irritation if taken on an empty stomach. Other species of Agathosma contain high levels of pulegone and are contraindicated in pregnancy.

Technical data

Botany

Agathosma betulina, a member of the Rutaceae (citrus) family, is a low shrub that can grow to a height of 2 m. The leaves are rhomboid-obovate in shape, 12 to 20 mm long and 4 to 25 mm broad, with a blunt and recurved apex. Numerous small oil glands are scattered throughout the lamina and large oil glands are situated at the base of each marginal indentation and at the apex. The flowers have five whitish petals and the brown fruits contain five carpels.4 The leaves become brittle and coriaceous when dried and when moistened become mucilaginous.4 They have a spicy odour and a pungent and spicy taste.5

Adulteration

The Khoi-San (a native South African tribe) name ‘buchu’ is applied to any aromatic herb or shrub that they find suitable for use as a dusting powder. Hence many species, including those not in the Agathosma genus, are recorded as buchu. The genus Diosma is often referred to as wild buchu, but was only used in cases where true buchu (Agathosma) was unprocurable.6

Adulterants include: Agathosma crenulata, A. serratifolia, A. ericifolia, Adenandra fragrans,7Empleurum serrulatum, Psoralea olbiqua and Myrtus communis.8 Other species also traded as medicinal buchu have included Diosma oppositifolia (Diosma succulentum), Agathosma pulchella, Empleurum unicapsulare (Empleurum ensatum) and the so-called anise buchu (possibly A. variabilis).9

Although A. crenulata has been used traditionally as true buchu, it is not suitable for therapeutic use due to the lower diosphenol and higher pulegone and isopulegone contents of the essential oil.10,11

The preferred medicinal buchu (Agathosma betulina) may be distinguished from A. crenulata and A. serratifolia by the ratio of leaf length to leaf width.7 This distinction does not hold for buchu hybrids (considerable hybridisation has taken place between A. betulina and A. crenulata), where chemical analysis of the essential oil is required.12 Use of buchu hybrids is not generally acceptable, since the level of pulegone in the essential oil varies enormously.13

From chemotaxonomic studies, two chemotypes of A. betulina have been identified11:

• Diosphenol chemotype: high concentration of diosphenol (total diosphenol isomers ≥22%), low isomenthone content (<29%)

• Isomenthone chemotype: high concentration of isomenthone (>31%), low diosphenol content (total diosphenol isomers ≤0.3 %).

No chemotypes of A. crenulata have been found. The diosphenol chemotype is probably preferable for therapeutic use.

Key constituents

• Essential oil (2%), consisting mainly of the monoterpene diosphenol. Other components include: limonene, (−)-isomenthone, (+)-menthone, (−)-pulegone, terpinen-4-ol and the sulphur-containing p-menthan-3-on-8-thiol, which is responsible for the blackcurrant flavour5

• Flavonoids, especially diosmin and rutin.14

As noted under Adulteration, other species such as Agathosma crenulata are not suitable for medicinal use due to the lower diosphenol and higher pulegone contents in their essential oils.10

image

Pharmacodynamics

Antimicrobial activity

An early in vitro study demonstrated some activity for the alcoholic extract of buchu against microflora typical of urinary tract infections.15 However, only the essential oil showed considerable activity against all the test organisms.

More recently poor or absent antimicrobial activity was observed for buchu essential oil against five different microorganisms using the zone of inhibition technique.16 This finding was somewhat supported by a later study that found only moderate activity, with minimum inhibitory concentrations (MIC) for the oil varying from 4 to 32 mg/mL (depending on the microorganism tested for Bacillus cereus, Staphylococcus aureus, Klebsiella pneumoniae and Candida albicans), as determined by a microtitre plate dilution method.17 In fact, the methanol:dichloromethane (1:1) extract of buchu leaf was more active than its hydrodistilled essential oil in this test model (MIC range against the same organisms 2 to 4 mg/mL).18

In an attempt to better reflect in vivo activity, extracts of buchu were treated with simulated gastric and intestinal fluids. The water extract showed a doubling of antimicrobial activity after it was subjected to simulated intestinal conditions. Intestinal transport simulation using the Caco-2 cell monolayer revealed good bioavailability for the compounds present in this extract.19,20

Other activity

At a high concentration buchu essential oil had an initial spasmogenic action on isolated guinea pig ileum, followed by spasmolysis. Cyclic AMP generation and the blocking of calcium channels were suggested as possible mechanisms for the spasmolytic activity, based on the experimental probes used.16

A relatively strong inhibition of 5-lipoxygenase was observed in vitro for the essential oil (IC50=50.4 μg/mL), but not the methanol:dichloromethane (1:1) extract.17,20

Pharmacokinetics

No human or animal data available. See above under Antimicrobial activity for limited in vitro information.

Clinical trials

No clinical studies on the urinary antiseptic and diuretic effects traditionally attributed to buchu are available.

Unpublished results from a double blind, placebo-controlled clinical trial involving 30 healthy male volunteers found that a buchu oil gel applied three times daily reduced swelling following exercise-induced muscle damage.20

Toxicology and other safety data

Toxicology

There are no reports of cases of poisoning and only in vitro toxicology data are available. Here the toxicities of buchu essential oil and extract were evaluated using the MTT (3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide) cellular viability assay in Graham cells (transformed human kidney epithelial cells).17,18,20 While the extract demonstrated no toxicity at the concentrations tested (IC50>100 μg/mL), the oil was highly active in disrupting the viability of test cells.

Contraindications

None required on current evidence.

Special warnings and precautions

Best not used during lactation without professional advice.

Interactions

No precautions required on current evidence.

Use in pregnancy and lactation

Category B2 – no increase in frequency of malformation or other harmful effects on the fetus from limited use in women. Animal studies are lacking.

The British Herbal Compendium 1992 suggests that buchu is contraindicated in pregnancy.14 However, this would only be the case for buchu substitutions (such as A. crenulata) that contain much higher levels of pulegone in their essential oil (see above).

Buchu is compatible with breastfeeding but caution should be exercised as it contains an essential oil that may pass into breast milk. As the therapeutic effects provided by buchu are rarely required in infants, its effects in this patient group are unknown, but probably benign.

Effects on ability to drive and use machines

None known.

Side effects

Occasional gastrointestinal intolerance and irritation if taken on an empty stomach.14

Overdosage

No reports of poisoning occur in published literature.9

Safety in children

No information available, but adverse effects are not expected.

Regulatory status in selected countries

Buchu is covered by a null Commission E monograph. It is on the UK General Sale List.

Buchu does not have GRAS status. However, it is freely available as a ‘dietary supplement’ in the USA under DSHEA legislation (1994 Dietary Supplement Health and Education Act). Buchu has been present as an ingredient in OTC weight control drug products and orally administered menstrual drug products. The FDA, however, advises that: ‘based on evidence currently available, there is inadequate data to establish general recognition of the safety and effectiveness of these ingredients for the specified uses’.

Buchu is not included in Part 4 of Schedule 4 of the Therapeutic Goods Act Regulations of Australia and is freely available for sale.

References

1. Grieve M, A Modern Herbal, New York, Dover Publications, 1971;Vol 1. pp. 133–134

2. Felter HW, Lloyd JU. King’s American Dispensatory, 18th ed. 3rd rev, Vol 1. 1905, Portland: Reprinted by Eclectic Medical Publications; 1983. pp. 891–892.

3. German Federal Minister of Justice. German Commission E for Human Medicine Monograph, Bundes-Anzeiger (German Federal Gazette), No. 22a, dated 01.02.1990.

4. Evans WC. Trease and Evans’ Pharmacognosy, 14th ed. London: WB Saunders, 1996. p. 272

5. Bisset NG, ed. Herbal Drugs and Phytopharmaceuticals. Stuttgart: Medpharm Scientific Publishers, 1994. pp. 102–103

6. Watt JM, Breyer-Brandwijk MG. The Medicinal and Poisonous Plants of Southern and Eastern Africa: Being an Account of Their Medicinal and Other Uses, Chemical Composition, Pharmacological Effects and Toxicology in Man and Animal, 2nd ed. Edinburgh: Livingstone; 1962. pp. 909–910

7. Spreeth AD. J S Afr Bot. 1976;42(2):109–119.

8. Anonymous. Flavour Ind. 1970;1(6):379–382.

9. Blaschek W, Ebel S, Hackenthal E, et al. HagerROM 2002: Hagers Handbuch der Drogen und Arzneistoffe. Heidelberg: Springer, 2002.

10. Kaiser R, Lamparsky D, Schudel P. J Agric Food Chem. 1975;23(5):943–950.

11. Collins NF, Graven EH, van Beek TA, et al. J Essent Oil Res. 1996;8(3):229–235.

12. Blommaert KLJ, Bartel E. J South Afr Bot. 1976;42(2):121–126.

13. Mills S, Bone K, eds. The Essential Guide to Herbal Safety. St Louis: Elsevier Churchill Livingstone, 2005. p. 296

14. British Herbal Medicine Association., British Herbal Compendium, Bournemouth, BHMA, 1992;Vol 1. pp. 43–45

15. Didry N, Pinkas M. Plantes Med Phytother. 1982;16(4):249–252.

16. Lis-Balchin M, Hart S, Simpson E. J Pharm Pharmacol. 2001;53:579–582.

17. Viljoen AM, Moolla A, van Vuuren SF, et al. J Essent Oil Res. 2006;18(suppl):2–16.

18. Moolla A, van Vuuren SF, van Zyl RL, et al. South Afr J Bot. 2007;73:588–592.

19. Viljoen AM, Vermaak I, Hamman J, et al. South Afr J Bot. 2007;73:319–320.

20. Moolla A, Viljoen AM. J Ethnopharmacol. 2008;119(3):413–419.

Bugleweed and European bugleweed

(Lycopus virginicus L., Lycopus europaeus L.)

Synonyms

Lycopus europaeus: gipsywort, bitter bugle, gypsy herb, marsh horehound. Lycopus virginicus: Virginia bugleweed, sweet bugleweed, Virginia horehound.

What is it?

Two main species of Lycopus are used therapeutically. Bugleweed (L. virginicus) is a native of the eastern United States and European bugleweed (L. europaeus) is one of the two European species.1 The traditional uses of hyperthyroidism and heart palpitations were given some scientific support by experiments in the 1940s and 1950s that found influences on dampening thyroid function. Since then there have been several studies investigating the mode of action and active constituents. While these investigations have been informative, the relevance of their findings is hampered by the dominance of in vitro studies and animal models where the herb was administered by injection. Also lacking at present are properly designed and conducted clinical trials. Hence, despite the scientific interest and theoretically plausible mechanisms, the rationale for the use of this herb still largely rests on traditional treatises, both from the United States and Europe, with a positive Commission E monograph.

Effects

May interfere with the effects of thyroid stimulating hormone (TSH) leading to reduced thyroxine production by the thyroid; alleviates palpitations and tachycardia caused by an overactive thyroid; possible inhibition of female fertility.

Traditional view

Traditional uses, especially for L. virginicus, included nervous tachycardia, Graves’ disease with cardiac involvement, thyrotoxicosis with difficult breathing, tachycardia and tremor2 and exophthalmic goitre.3 It was also recommended for organic and functional cardiac disease, abnormally active circulation and rapid pulse with high temperature.3 Other clinical uses included chronic cough, irritating and wet cough, pneumonia, bronchitis,2,3 restlessness, insomnia and anxiety.3

Bugleweed, particularly L. europaeus, is considered in European herbal medicine to have antithyroid activity.4 Being less powerful than conventional drugs, it is recommended for mild thyroid hyperfunction and can be used as a long-term treatment.5

Summary actions

TSH antagonist, antithyroid, mild sedative, possible antigonadotropic agent.

Can be used for

Indications supported by clinical trials

Hyperthyroidism, especially Graves’ disease and associated symptoms such as tachycardia and rapid pulse (open label trials, weak evidence). ‘Vegetative dystonia’ which is essentially anxiety characterised by cardiac and digestive symptoms (open label trials, weak evidence).

Traditional therapeutic uses

Hyperthyroidism as above, but especially with cardiac involvement; also for restlessness, insomnia and anxiety.

Preparations

Dried herb for an infusion, tincture or liquid extract for internal use.

Dosage

• 3 to 9 g/day of dried aerial parts or by infusion

• 2 to 6 mL/day of a 1:2 liquid extract or equivalent in tablet or capsule form

• 6 to 18 mL/day of a 1:5 tincture.

Duration of use

May be taken long-term, but only if appropriate.

Summary assessment of safety

Contraindicated in pregnancy and lactation and in patients with an underactive thyroid. Do not administer with thyroid hormone preparations or during the application of thyroid diagnostic procedures using radioactive isotopes.

Technical data

Botany

L. europaeus is a perennial herb spreading by branching stolons. The quadrangular, pubescent stems are erect, simple or branched, 30 to 100 cm tall. Leaves are opposite, short-petioled and ovate-lanceolate to elliptic, up to 12 cm long; the lower leaves are pinnate, the upper leaves crenate. Flowers are about 4 mm long, white with purple dots and grow in dense whorls at the base of the uppermost leaves.1,6

L. virginicus is a perennial, 15 to 60 cm tall. Stems are quadrangular, erect and pubescent, producing stolons at the base. The leaves are opposite, ovate or oblong-lanceolate, coarsely serrate but entire near the base, 2 to 5 cm long. Flowers are small, purplish, growing in axillary whorls, with a mint-like odour.

Adulteration

Leonurus marrubiastrum L. may be mistaken for L. europaeus, especially when wildcrafted.7

Key constituents

L. virginicus and L. europaeus aerial parts contain flavonoids and phenolic acids, such as derivatives of cinnamic acid (including caffeic acid), although the pattern varies in each plant.810

Pharmacodynamics

Antithyroid and antigonadotropic activities

It is probably most useful to review the pharmacological research into the antihormonal activity of Lycopus in an approximate chronological order, as there is certainly an aspect of unfolding discovery to the story.

Madaus, Cook and Albus in 1941 were able to show in experimental investigations that the effects of L. europaeus and L. virginicus L. were similar.11 Lycopus countered the typical weight decrease and tachycardia from thyroxine in the rabbit in an acute model. Results from another model also suggested a direct thyroxine antagonism: thyroxine antagonises the temperature fall induced by novocaine injection in the guinea pig and this effect was cancelled by Lycopus administration. Thyroxine causes an increased oxygen requirement as a result of increased metabolism and Lycopus countered this effect in rats. Next to this direct antithyroxine effect, the authors also showed effects on TSH activity. After repeated injection of TSH, oxygen consumption in the guinea pig increased by around 25%. Simultaneous administration of Lycopus extract reduced this to about 10%, but only at the highest dosage.12

Pharmacodynamic research on Lycopus began in earnest in the early 1960s when Kemper and co-workers found that an aqueous extract of L. virginicus inhibited the actions of gonadotropic hormones and TSH in vivo (based on organ weights) after administration by injection to guinea pigs and rats.13,14 However, a preparation of L. europaeus had no influence on thyroid metabolism in rats after oral doses, as assessed using radioactive iodine.15

A paper published in 1970 claimed, on the basis of in vivo experiments using doses administered by injection, that an oxidised form of lithospermic acid was the antigonadotropic principle in L. virginicus.16 However, it was the work of the late Dr Hilke Winterhoff and team that found a different basis for this and the antithyroid activity in a series of studies published from 1976 to recently. Lithospermic acid was shown to have little antihormonal activity compared to the freeze-dried extract of Lycopus.17,18 This freeze-dried aqueous extract of Lycopus (species not specified) given alone by injection or with TSH to infant female rats led to a marked fall in circulating thyroid hormones.19 When TRH and L. virginicus extract were co-administered (at different sites) by injection to rats, TSH levels were significantly lower after 10 minutes when compared to the TRH and saline controls.20 As a result, thyroid hormone levels were suppressed and significantly lower 3 h later. Pretreatment of rats with dried extract for 4 days resulted in a drop in peripheral thyroid hormone levels.20

In the euthyroid rat, injections (50 mg/kg) of freeze-dried extracts of both L. europaeus and L. virginicus greatly reduced serum and pituitary TSH levels.21 Effects were less clear in hypothyroid rats. Serum prolactin was also reduced by L. virginicus injection, but only at 400 mg/kg. L. europaeus injection (50 mg/kg) did significantly lower pituitary (but not serum) prolactin levels.

The active components suggested by Winterhoff and team as responsible for these effects were the oxidation products of phenolic compounds such as rosmarinic acid, which are abundant in Lycopus species.22 These showed marked antigonadotropic activity in a rat model after injection. This line of research was further progressed by this group in collaboration with others, including several in vitro investigations conducted in the 1980s. The antigonadotropic activity of ether-extracted fractions of L. virginicus aqueous extracts was tested in a suitable model (details not specified).23 Within the ether extracts, significant activity was only found after the addition of an oxidising agent (up to a 10-fold increase). This oxidised and active form was identified to contain oxidation products of rosmarinic acid. Freeze-dried aqueous extracts of L. virginicus and L. europaeus inhibited the binding of bovine TSH to human thyroid plasma membranes and inhibited the stimulation of adenylate cyclase.24 Auto-oxidation products of the 3,4-dihydroxycinnamic acid derivatives (such as caffeic, rosmarinic, chlorogenic and ellagic acids) were also active in the same model.25 It was suggested that these compounds form adducts with TSH, resulting in a greatly reduced or absent ability to bind to the TSH receptor.26 In later research, rosmarinic acid and a freeze-dried aqueous extract of L. virginicus also inhibited forskolin-induced activation of adenylate cyclase in cultured rat thyroid cells.27

The impact of Lycopus on the peripheral conversion of T4 to T3 was investigated in vitro using iodothyronine deiodinases from rat liver microsomes.28 Preparations of L. virginicus, especially the ether fraction of hot aqueous extracts, markedly inhibited the enzyme in a dose-dependent manner. Subsequent investigation found that the active principles exhibited chemical characteristics of phenolcarboxylic acids (e.g. rosmarinic acid). Curiously, oxidation of extracts with potassium permanganate decreased this activity.29

In all samples of Graves’ immunoglobulin G (IgG) tested, incubation with L. virginicus or L. europaeus or their auto-oxidised constituents decreased the TSH-binding inhibitor activity in vitro. This was in a dose-dependent manner and with some degree of specificity.30 The plant extracts and their auto-oxidised constituents also inhibited the biological responses to Graves’ IgG in vitro.

The efficacy of injections of L. virginicus extracts for up to 3 days at inhibiting thyroid hormone levels and thyroid weight varied with the method of preparation.31 Extraction with boiling water or ethanol yielded inactive extracts, whereas oxidation with potassium permanganate re-established activity. The reduction in prolactin levels observed in an earlier study was also confirmed (as assessed by assay 6 h after a single intravenous injection). It was shown in a subsequent study that several phenolic plant ingredients mentioned already, but additionally luteolin-7-beta-glucuronide,32 confer antigonadotropic activity. However, their oxidation products are likely to be more active.33 The suggestion that L. europaeus contains lithospermic acid was also refuted. Oligomeric (short polymers) oxidation products of caffeic acid (cyclolignan derivatives) with antigonadotropic activity were subsequently identified as likely active components.34,35

As well as the previously noted antagonism of TSH, results from a series of experiments suggested that injections of L. europaeus are capable of suppressing thyroid hormone secretion by a direct intrathyroidal attack.36 Additional phenolic acids (including isoferulic acid and ferulic acid) as well as caffeic acid were active in this TSH-antagonism model.

One reservation that can be expressed about the above research is that most of the results have been generated using in vitro models or in vivo models following injection of the herbal extracts or relevant compounds. This was addressed by Winterhoff’s team in a 1994 publication where a 70% ethanolic extract L. europaeus was administered by oral means to rats, albeit at quite high doses of 200 and/or 1000 mg/kg.37 Diverse endocrine parameters were measured between 3 and 24 h after oral doses and compared with intraperitoneal doses (of L. virginicus). Oral application of the extract caused a long-lasting decrease in T3 levels, presumably due to reduced peripheral T4 deiodination. A pronounced reduction of T4 and TSH was observed, as well as LH (luteinising hormone), indicating a central action for the extract.

The impact of oral L. europaeus (2×5 mg or 2×200 mg/kg per day for up to 8 weeks of a dried aqueous-ethanolic extract) on cardiac symptoms in hyperthyroid rats was compared with the beta-blocking drug atenolol.38 Even the lower dose of the herb countered the raised body temperature, whereas the reduced body weight and increased food intake was not influenced by any treatment. No significant changes in thyroid hormone levels or TSH were observed. Lycopus (in general both doses) and atenolol reduced the increased heart rate, blood pressure and heart size, but the drug was considerably more active with respect to heart rate. Interestingly, Lycopus given to normal rats at 2×200 mg/kg per day had no impact on any of the above parameters. Collectively the above findings suggest that the herb only antagonises the effects of increased thyroid hormone levels, and does this without altering depressed TSH and elevated thyroxine levels, at least in the model used.

This work was subsequently extended in two publications using three rat models of differing severity of hyperthyroidism induced by differing applications of T4. Findings were similar to the above study in both publications, with Lycopus extract, extract fractions and powdered herb having almost the same potency as beta-blockers (atenolol and propranolol) in reducing body temperature.39,40 The effects on beta-adrenoreceptor density, blood pressure and heart rate were less distinct, but still significant. In the later publication there was some suggestion that Lycopus powder had inhibited T4 to T3 conversion.40 (Note that this result may have been more determined by the model used than the form of Lycopus, since the powder was only tested in one model of hyperthyroidism.)

Summarising the research to date, results from in vitro experiments or in vivo models with dosage by injection suggest that Lycopus extracts or its components at relatively high doses antagonised or reduced pituitary hormones (TSH, gonadotropins, prolactin), reduced thyroxine production, reduced peripheral conversion of T4 to T3 and antagonised the binding and effects of Graves’ IgG. In terms of phytochemical components responsible for these effects, oligomeric oxidation products of phenolic acids such as caffeic acid were found to be most active. As a consequence, freshly prepared aqueous-ethanolic extracts were not found to be active, as they are devoid of such products. However, some of the above results (TSH, T4 and T3 reductions) were confirmed in an oral-dose model using high doses of a commercial aqueous-ethanolic extract. Perhaps this extract was active because phenolic acid oxidation products had formed in the time since its manufacture. More recently, animal models using realistic oral doses of a commercial aqueous-ethanolic extract have provided quite a different perspective. Here it was found that Lycopus could antagonise the cardiac effects of induced hyperthyroidism without any corresponding change in thyroid hormones or TSH. Lycopus given orally to normal rats at such doses had no impact on thyroid function. The phytochemical components responsible for the activities observed in these later experiments have not been explored to date.

Other activity

A methanolic extract of L. europaeus was one of the three most active of 122 traditional Chinese herbs screened for in vitro activity against xanthine oxidase (the enzyme that catalyses purine metabolites to uric acid) with an IC50 of 26 μg/mL.41 Two new isopimarane diterpenes were isolated from L. europaeus that were capable of potentiating the in vitro activities of tetracycline and erythromycin against resistant strains of Staphylococcus aureus possessing multidrug efflux mechanisms.42 Aqueous and methanolic extracts of L. europaeus showed high radical scavenger capacity compared to 17 other medicinal Labiatae species tested.43

Pharmacokinetics

No relevant data found.

Clinical trials

Early clinical studies were largely observational in nature and, while they could be construed to provide weak evidence by modern standards, some interesting and sometimes striking results were documented. The following brief review is not comprehensive, as many such studies were published in the 1950s and early 1960s. Throughout these and the later trials, the doses of Lycopus administered were quite low.

Mattausch reported in 1943 on the treatment of about 200 patients with mild hyperthyroidism. A decline in pulse frequency was observed and, following a longer treatment duration, also a normalisation of other symptoms.44 A group of 50 patients with symptoms of hyperthyroidism, or ‘vegetative dystonia’ that with more exacting clinical investigation turned out to be latent hyperthyroidism, were treated with Lycopus.45 The thyroid uptake of radioactive iodine was evaluated in 32 of these patients. This demonstrated an objective improvement in thyroid function in 15 cases; eight of the other 17 exhibited symptom improvement with no corresponding objective change. Overall for the 50 cases, pulse rates were normalised and insomnia was alleviated. Another study found reduced symptoms and pulse rate, and increased body weight in 53 hyperthyroid patients treated with Lycopus.46

One hundred patients with severe hyperthyroidism were treated with Lycopus prior to surgery, rather than with Lugol’s solution.47 The appearance of the gland in the operation field showed relatively less vascularisation, and postoperative reactions were notably mild.

As alluded to above, Lycopus has been used to treat ‘vegetative dystonia’, a disturbance of autonomic nervous system function. In a series of 123 cases this disturbance was attributed to elevated thyroid function.48 Patients were treated with a commercial Lycopus tincture (12 drops twice a day). After 14 days, the pulse rate had dropped by an average of 11%. Based on subjective symptoms there was a clear improvement in 83 patients after 14 days, which by 28 days had increased to 102 patients. Symptoms such as dermographism, tremor, hair loss and sweating were improved. A significant weight gain was also observed.

In a more recent observational study of 43 patients (21 with latent hyperthyroidism and 22 with ‘vegetative dystonia’), Lycopus dried extract (10 mg/day equivalent to 40 mg of herb) was given for 21 days.49 Of the 34 patients with cardiac symptoms, 27 reported improvement in palpitations and/or benign arrhythmias. However, thyroid parameters were unchanged, including in another 20 healthy volunteers who received the herbal treatment.

A controlled, open study examined the effect of 40 mg/day L. europaeus extract on thyroid function and associated symptoms in 62 patients with hyperthyroidism of unspecified causes.50 The study population consisted of patients with a TSH <1.0 mU/L and hyperthyroidism-associated symptoms. The main clinical outcome measured was 24-h urinary T3 and T4 excretion after around 12 days of therapy. The study found that T4 excretion was significantly increased by Lycopus (p=0.032). There was also a trend to increased T3 excretion that did not achieve clinical significance. This occurred in conjunction with a reduction of some symptoms, specifically increased heart rate in the morning. However, TSH and serum T3 and T4 levels were not changed. The authors proposed that a renal mechanism was responsible for the increased T4 clearance. Apart from the lack of a placebo group, a key weakness of this study was the entry point of TSH <1.0 mU/L, which meant that patients with both clinical and sub-clinical hyperthyroidism were included. Also the causes of hyperthyroidism were not differentiated.

Toxicology and other safety data

Toxicology

Pressed juice of L. europaeus was lethal to male mice (0.75 mL corresponding to 7.5 g fresh plant, route unknown but probably injection). Intravenous injection of 1 mL of L. virginicus pressed juice was lethal, but 3 mL given orally caused no toxic symptoms.51

Caffeic acid has been listed as a possible carcinogen on the basis of in vivo studies in mice and rats using two oral dosage regimes: 2.1 g/kg (males), 3.1 g/kg (females) for 96 weeks; 0.7 g/kg and 0.8 g/kg over 104 weeks.52 Given the small amount present in the herb, this research is unlikely to be relevant to the therapeutic application of bugleweed.7 Moreover, caffeic acid is a common constituent of many plant foods, including fruit, vegetables and coffee, principally in conjugated forms such as chlorogenic acid.

Contraindications

Thyroid hypofunction, enlargement of the thyroid without functional disorder,53 pregnancy and lactation.51

Special warnings and precautions

Caution is advised in women wishing to conceive.

Interactions

Interference with uptake of radioactive isotopes has been observed in humans.51 Bugleweed should not be administered concurrently with preparations containing thyroid hormone and iodine supplements, although those concerns are largely theoretical and, given the therapeutic uses of Lycopus, their co-administration is clinically unlikely.53

Use in pregnancy and lactation

Category C – has caused or is associated with a substantial risk of causing harmful effects on the fetus or neonate without causing malformations.

Reduction in serum thyroid hormones in vivo suggests that Lycopus should not be taken during pregnancy. Treatment with Lycopus in mice and rats has reduced the number of offspring.51

Lycopus is contraindicated in breastfeeding. Decreased milk supply has been observed after administration of L. virginicus in suckling rats.51 Although the human relevance of the effects of Lycopus on serum prolactin in experimental models is not clear, these herbs should not be taken while breastfeeding due to the possibility of both prolactin reduction in the mother and antithyroid constituents passing into breast milk.

Effects on ability to drive and use machines

No negative influence is expected.

Side effects

In rare cases, extended therapy and high (undefined) dosages of bugleweed preparations have resulted in an enlargement of the thyroid. Sudden discontinuation of bugleweed preparations can cause increased symptoms of the disease (hyperthyroid function).53 The following side effects have been reported in the literature between 1941 to 1968 from clinical usage of bugleweed alone or combined with motherwort (Leonurus cardiaca): headache, increase in size of thyroid, and occasionally an increase in hyperthyroid symptoms including nervousness, tachycardia and loss of weight. Increase in thyroid size was observed in patients with goitre not due to thyroid malfunction. The incidence of headache could probably be avoided by reducing the dosage.51 Not all trials resulted in such side effects.

Overdosage

No incidents found in published literature.

Regulatory status in selected countries

In the UK bugleweed is included on the General Sale List and is covered by a positive Commission E Monograph.

In the USA bugleweed does not have GRAS status. However, it is freely available as a ‘dietary supplement’ under DSHEA legislation (Dietary Supplement Health and Education Act of 1994).

Bugleweed is not included in Part 4 of Schedule 4 of the Therapeutic Goods Regulations in Australia and is freely available for sale.

References

1. Applequist W. The Identification of Medicinal Plants. In A Handbook of the Morphology of Botanicals in Commerce. USA: American Botanical Council; 2006.

2. British Herbal Medicine Association’s Scientific Committee. British Herbal Pharmacopoeia. Bournemouth: BHMA, 1983.

3. Felter HW, Lloyd JU. King’s American Dispensatory, 18th ed. rev 3, 1905, Portland. Reprinted Eclectic Medical Publications; 1983.

4. Madaus G. Lehrbuch der biologischen Heilmittel, Vol 2, Leipzig, 1938. Reprinted New York Verlag; 1976. pp. 1807–1809.

5. Weiss RF. Herbal Medicine, translated by Meuss AR from the 6th German Edition of Lehrbuch der Phytotherapie. Beaconsfield: Beaconsfield Publishers; 1988.

6. Harvey R. Br J Phytother. 1995/96;4(2):55–65.

7. Blaschek W, Ebel S, Hackenthal E, et al. HagerROM 2002: Hagers Handbuch der Drogen und Arzneistoffe. Heidelberg: Springer, 2002.

8. Kartnig T, Bucar F. Planta Med. 1995;61(4):392.

9. Bucar F, Kartnig T, Paschek G, et al. Planta Med. 1995;61(5):489.

10. Kartnig T, Bucar F, Neuhold S. Planta Med. 1993;59(6):563–564.

11. Madaus G, Koch E, Albus G. Z Gesamte Exp Med. 1941;109:411–424. In: Vonhoff C, Winterhoff H. Z Phytother 2006;27(3):110–119

12. Koch FE. Madaus Jahresbericht VI 107–112. In: Vonhoff C, Winterhoff H. Z Phytother 2006;27(3):110–119.

13. Kemper F, Loeser A. Acta Endocrinol. 1961;38(2):200–206.

14. Kemper F, Loeser A, Richter A. Arzneimittelforschung. 1961;11:92–94.

15. Hartenstein H, Müller WA. Hippokrates. 1961;32:284–288.

16. Wagner H, Hörhammer L, Frank U. Arzneimittelforschung. 1970;20(5):705–713.

17. Kemper FH, Winterhoff H, Sourgens H, et al. Planta Med. 1978;33(3):311.

18. Niehaus KD, Winterhoff H, Kemper FH. Naunyn Schmiedebergs Arch Pharmacol. 1976;293(suppl):R39.

19. Winterhoff H, Sourgens H, Gumbinger HG, et al. Acta Endocrinol. 1979;91(suppl 225):43.

20. Sourgens H, Winterhoff H, Gumbinger HG, et al. Acta Endocrinol. 1980;94(suppl 234):49.

21. Sourgens H, Winterhoff, Gumbinger HF, et al. Planta Med. 1982;45(2):78–86.

22. Gumbinger HG, Winterhoff H, Sourgens H, et al. Contraception. 1981;23(6):661–666.

23. Gumbinger HG, Winterhoff H. Naunyn Schmiedebergs Arch Pharmacol. 1980;311(supp1):R52. Abstract 208

24. Auf’mkolk M, Ingbar JC, Amir SM. Endocrinology. 1984;115(2):527–534.

25. Auf’mkolk M, Amir SM, Kubota K, et al. Endocrinology. 1985;116(5):1687–1693.

26. Auf’mkolk M, Amir SM, Kubota K, et al. Endocrinology. 1985;116(5):1677–1686.

27. Kleemann S, Winterhoff H. Planta Med. 1990;56(6):683.

28. Köhrle J, Auf’mkolk M, Winterhoff H, et al. Acta Endocrin. 1981;96(suppl 240):15–16.

29. Auf’mkolk M, Köhrle J, Gumbinger H, et al. Horm Metab Res. 1984;16(4):188–192.

30. Auf’mkolk M, Ingbar JC, Kubota K, et al. Endocrinology. 1985;116(5):1687–1693.

31. Sourgens H, Winterhoff H, Gumbinger HG, et al. Planta Med. 1982;45(6):78–86.

32. Gumbinger HG, Winterhoff H, Wylde R, et al. Planta Med. 1992;58(1):49–50.

33. Winterhoff H, Gumbinger HG, Sourgens H. Planta Med. 1988;54(2):101–106.

34. Nahrstedt A, Albrecht M, Wray V, et al. Planta Med. 1990;56(4):395–398.

35. John M, Gumbinger HG, Winterhoff H. Planta Med. 1990;56(1):14–18.

36. Frömbling-Borges A. Z Phytother. 1990;11(1):1–6.

37. Winterhoff H, Gumbinger HG, Vahlensieck U, et al. Arzneimittelforschung. 1994;44(1):41–45.

38. Vonhoff C, Baumgartner A, Hegger M, et al. Life Sci. 2006;78(10):1063–1070.

39. Vonhoff C, Biller A, Nahrstedt A. et al., 51st Annual Congress of the Society for Medicinal Plant Research, Germany, 217: August 31–September 4, 2003. Abstract P267.

40. Vonhoff C, Winterhoff H. Z Phytother. 2006;27(3):110–119.

41. Kong LD, Cai Y, Huang WW, et al. J Ethnopharmacol. 2000;73(1–2):199–207.

42. Gibbons S, Oluwatuyi M, Veitch NC, et al. Phytochemistry. 2003;62(1):83–87.

43. López V, Akerreta S, Casanova E, et al. Plant Foods Hum Nutr. 2007;62(4):151–155.

44. Mattausch F. Kippokrates. 1943;10:168–170. In: Vonhoff C, Winterhoff H. Z Phytother 2006;27(3):110–119

45. Lobenhofer G. Münch Med Wochenschr. 1953;95(51):1375–1376.

46. Gottlieb J. Folia Clin Int. 1952;2(9):420–423.

47. Sittig H. Münch Med Wochenschr. 1995;97(25):826. 829–832

48. Ackermann VJ. Dtsch Med J. 1959;10(12):409–411.

49. Scheck R, Biller A. Natura Med. 2000;15(5):31–36.

50. Beer A-M, Wiebelitz KR, Schmidt-Gayk H. Phytomed. 2008;15(1–2):16–22.

51. De Smet P.A.G.M., Keller K, Hansel R, et al, eds., Adverse Effects of Herbal Drugs, Berlin, Springer-Verlag, 1993;Vol 2. pp. 245–251

52. WHO, International Agency for Research on Cancer, IARC Working Group on the Evaluation of Carcinogenic Risks to Humans, Lyon, IARC, 1993;Vol 56. p. 115

53. Blumenthal M, ed. The Complete German Commission E Monographs: Therapeutic Guide to Herbal Medicines. Austin: American Botanical Council, 1998. pp. 98–99

Bupleurum

(Bupleurum falcatum L.)

Synonyms

Hare’s ear root (Engl), Bupleuri radix (Lat), chai hu (Chin), saiko (Jap), siho (Kor), segl-hareøre (Dan).

What is it?

Several species of Bupleurum have been officially used in traditional Chinese medicine (TCM), mainly B. falcatum L., B. chinense DC and B. scorzonerifolium Willd. Although other species may be used, the toxic plant B. longiradiatum should not be used medicinally. Pharmacological research on Bupleurum root highlights an anti-inflammatory activity that appears to be mediated through the enhanced release and potentiation of hormones from the adrenal cortex. There are still many gaps in the research evidence, but it is possible that Bupleurum acts to mobilise the body’s own equivalent of steroidal anti-inflammatory mechanisms.

Effects

Supports the body’s anti-inflammatory responses; modulates immune function; protects the liver, stomach and kidneys from toxic damage.

Traditional view

In TCM Bupleurum is classified as a herb that resolves Lesser Yang Heat patterns, relaxes constrained Liver Qi, disharmony between the Liver and the Spleen and raises the Yang Qi in Spleen or Stomach Deficiency. Hence Bupleurum is used to treat alternating chills and fever, liver enlargement, prolapse of the uterus and rectum and irregular menstruation.1 Traditional texts list its properties as bitter and cool and it acts as a diaphoretic (in fever management) and to regulate and restore gastrointestinal and liver function.

Summary actions

Anti-inflammatory, hepatoprotective, antitussive, diaphoretic.

Can be used for

Indications supported by clinical trials

Influenza, common cold, feverish conditions (uncontrolled studies).

Traditional therapeutic uses

Alternating chills and fever; liver enlargement; prolapse of the uterus and rectum; epigastric pain, nausea, indigestion; irregular menstruation. Often combined with Astragalus for debility and prolapse.

May also be used for

Extrapolations from pharmacological studies

Chronic inflammatory disorders, especially autoimmune disease involving the liver or kidneys; acute or chronic liver diseases, chemical liver damage, poor liver function.

Preparations

Dried root for decoction, liquid extract, tablets and capsules; powdered root.

Dosage

• 3 to 12 g/day of the dried root by decoction

• 4 to 8 mL/day of the 1:2 liquid extract or its equivalent.

Duration of use

May be used in the long term if taken within the recommended dosage.

Summary assessment of safety

Minor side effects may occur in sensitive individuals.

Technical data

Botany

The Bupleurum genus is a member of the Umbelliferae (Apiaceae, carrot) family and consists of shrubs and herbs with entire leaves, often parallel veined, hence the name hare’s ear.2Bupleurum chinensis grows 45 to 85 cm high and may or may not be branched. Leaves are alternate, broad-linear to broad-lanceolate (3 to 9 cm long, 0.6 to 1.3 cm wide) with a marginal vein. Umbels are compound, axillary and terminal and contain yellow flowers. Fruit is ovoid, the brown root is conical, 6 to 15 cm long and 0.3 to 0.8 cm in diameter.3

Key constituents

Bupleurum falcatum:

• Triterpenoid saponins (saikosaponins a, b1, b2, b3, b4, c, d, e and f) and their sapogenins4
• Phytosterols,4 pectin-like polysaccharides (bupleurans).5

Since the highest levels of saikosaponins are found in Bupleurum falcatum (2.8%) and B. chinensis (1.7%), these species are preferred.6,7 Saikosaponins a and d are considered to be the most biologically active.

Adulteration

Although other species of Bupleurum may be used, the toxic plant B. longiradiatum should not be used medicinally.6,8

image

Pharmacodynamics

Many pharmacological studies have been conducted on the isolated saikosaponins. Although these studies have relevance to the medicinal use of Bupleurum, some important qualifications should be considered. In much of the research, saikosaponins were tested in vitro or administered by injection. Since a major active form of saikosaponins in the bloodstream is probably the sapogenins (i.e. saponin aglycones), the relevance of the in vitro research to the oral use of Bupleurum is uncertain. Similarly, the relevance of injected saikosaponins to the oral use of Bupleurum is not entirely clear, although it appears that oral doses do have some similar but milder activity.911