Sie sind auf Seite 1von 29

This article appeared in a journal published by Elsevier.

The attached
copy is furnished to the author for internal non-commercial research
and education use, including for instruction at the authors institution
and sharing with colleagues.
Other uses, including reproduction and distribution, or selling or
licensing copies, or posting to personal, institutional or third party
websites are prohibited.
In most cases authors are permitted to post their version of the
article (e.g. in Word or Tex form) to their personal website or
institutional repository. Authors requiring further information
regarding Elsevier’s archiving and manuscript policies are
encouraged to visit:
http://www.elsevier.com/copyright
Author's personal copy

Journal of Ethnopharmacology 138 (2011) 286–313

Contents lists available at SciVerse ScienceDirect

Journal of Ethnopharmacology
journal homepage: www.elsevier.com/locate/jethpharm

Review

Phyllanthus amarus: Ethnomedicinal uses, phytochemistry and pharmacology:


A review
Jay Ram Patel a , Priyanka Tripathi b,1 , Vikas Sharma a , Nagendra Singh Chauhan a , Vinod Kumar Dixit a,∗
a
Department of Pharmaceutical Sciences, Dr. Harisingh Gour Vishwavidyalaya, Sagar 470003, M.P., India
b
Shri Ramnath Singh Mahavidyalaya (Pharmacy), Gormi, Bhind 477660, M.P., India

a r t i c l e i n f o a b s t r a c t

Article history: Ethnopharmacological relevance: Phyllanthus amarus Schum. & Thonn. belongs to the family Euphorbiaceae
Received 3 March 2011 is a small herb well known for its medicinal properties and widely used worldwide. P. amarus is an
Received in revised form important plant of Indian Ayurvedic system of medicine which is used in the problems of stomach,
20 September 2011
genitourinary system, liver, kidney and spleen. It is bitter, astringent, stomachic, diuretic, febrifuge and
Accepted 23 September 2011
antiseptic. The whole plant is used in gonorrhea, menorrhagia and other genital affections. It is useful in
Available online 29 September 2011
gastropathy, diarrhoea, dysentery, intermittent fevers, ophthalmopathy, scabies, ulcers and wounds.
Materials and methods: The present review covers a literature across from 1980 to 2011. Some infor-
Keywords:
Phyllanthus amarus
mation collected from traditional Ayurvedic texts and published literature on ethanomedicinal uses of
Hepatoprotective Phyllanthus amarus in different countries worldwide.
Anti-inflammatory Results: Phytochemical studies have shown the presence of many valuable compounds such as lignans,
Anticancer flavonoids, hydrolysable tannins (ellagitannins), polyphenols, triterpenes, sterols and alkaloids. The
Diuretics extracts and the compounds isolated from P. amarus show a wide spectrum of pharmacological activ-
Nephroprotective ities including antiviral, antibacterial, antiplasmodial, anti-inflammatory, antimalarial, antimicrobial,
Antioxidant anticancer, antidiabetic, hypolipidemic, antioxidant, hepatoprotective nephroprotective and diurectic
Antiviral
properties.
Antibacterial
Conclusion: The present review summarizes information concerning the morphology, ecology, ethnophar-
Antihyperglycemic
Antihypercholesterolemic macology, phytochemistry, biological activities, clinical applications and toxicological reports of P.
amarus. This review aims at gathering the research work undertaken till date on this plant in order
to provide sufficient baseline information for future works and commercial exploitation.
© 2011 Elsevier Ireland Ltd. All rights reserved.

Abbreviations: 2AA, 2-aminoanthracene; 2NF, 2-nitrofluorene; 3D7, chloroquine-sensitive strain of Plasmodium falciparum; 4-NQO, 4-nitroquinolone-1-oxide; ABTS, 2,2 -
azino-bis (3-ethylbenz-thiazoline-6-sulfonic acid); AF2, 2-aminofluorene; AH, aniline hydroxylase; ALP, alkaline phosphatase; ALT, alanine transaminase; AP-1, transcription
factors; AST, aspartate transaminase; CAT, catalase; CD4, T-helper cell; CFA, complete freund’s adjuvant; COX-2, cyclooxygenase; CTX, cyclophosphamide; DHBV, duck
hepatitis B virus; DHHDP, 1-galloyl-2,3-dehydrohexahydroxydiphenyl; DLA, Dalton’s lymphoma ascites tumor; DMN, dimethylnitrosamine; DMSO, dimethyl sulphoxide;
DNA, deoxy ribonucleic acid; DPPH, 2-diphenyl-1-picrylhydrazyl; EAC, Ehrlich ascites carcinoma; EC50 , effective concentration 50%; ENNG, N-ethyl-N-nitrosoguanidine; FRAP,
ferric reducing antioxidant power; FT-IR, Fourier transform infrared spectroscopy; GGT, ␥-glutamyl transpeptidase; GPX, glutathione peroxidase; GSH, cellular glutathione;
GST, glutathione-S-transferase; HBeAg, hepatitis B effective antigen; HBsAg, hepatitis B suppresive antigen; HBV DNA, hepatitis B viral DNA; HBV, hepatitis B virus; HCC,
hepatocellular carcinoma; HE, hexane extract; HepA, Hepatitis A; HepA2 , Hepatitis A2 ; HIV, human immunodeficiency virus; HPLC, high-performance liquid chromatography;
HPTLC, high-performance thin layer chromatography; HTG, hepatic triglyceride; i.p., intraperitoneal injection; IC50 , inhibitory concentration 50%; ID50 , inhibitory dose 50%;
IFN-a/c, interferon; IL-1, interleukin-1; IL-10, interleukin-10; IL-12, interleukin-12; IL-18, interleukin-18; iNOS, endotoxin-induced nitric oxide; IR, infra red; KC, Kupffer
cells; LPO, lipid peroxidation; LPS, lipopolysaccharides; MDA, malondialdehyde; MDR, multi-drug resistance; MICs, minimum inhibitory concentrations; mRNA, messenger
ribo nucleic acid; NDEA, N-nitrosodiethylamine; NF-kB, NF-kappa ␤; NMR, nuclear magnetic rasonance; 4-NQO, 4-nitroquinolone-1-oxide; P. amarus, Phyllanthus amarus; P.
debilis, Phyllanthus debilis; P. fraternus, Phyllanthus fraternus; P. kozhikodianus, Phyllanthus kozhikodianus; P. maderaspatensis, Phyllanthus maderaspatensis; P. niruri, Phyllanthus
niruri; P. urinaria, Phyllanthus urinaria; PAF, platelet activating factor; PCV, packed cell volume; PGE2 , prostaglandin E2 ; P.O., per oral; ROS, reactive oxygen species; SCGE, single
cell gel electrophoresis; SEF, supercritical-fluid extraction; SL, silymarin; SOD, superoxide dismutase; STG, serum triglyceride; STZ, streptozotocin; TBARS, thiobarbituric acid
reactive substances synthase; TLC, thin layer chromatography; TNF-␣, tumor necrosis factor ␣; UV, ultra violet; WBC, white blood carpuscles; WHV, Woodchuck hepatitis virus.
∗ Corresponding author. Tel.: +91 9425647546.
E-mail address: vkdixit2011@rediffmail.com (V.K. Dixit).
1
Present address: Division of Pharmaceutics, Central Drug Research Institute, Lucknow (U.P.) 226001, India.

0378-8741/$ – see front matter © 2011 Elsevier Ireland Ltd. All rights reserved.
doi:10.1016/j.jep.2011.09.040
Author's personal copy

J.R. Patel et al. / Journal of Ethnopharmacology 138 (2011) 286–313 287

Contents

1. Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 287
2. Historical perspectives . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 288
3. Botanical description and vernacular names . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 290
4. Biogeography and ecology . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 291
5. Pharmacognostic characters . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 291
6. Phytochemical studies . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 292
7. Analytical techniques . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 293
8. Pharmacological activity . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 295
8.1. Antiamnesic activity . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 295
8.2. Antibacterial activity . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 295
8.3. Anticancer activity . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 299
8.4. Anti-diarrhoeal, gastroprotective and antiulcer activity . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 300
8.5. Antifungal activity . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 301
8.6. Analgesic, anti-inflammatory, anti-allodynic and anti-oedematogenic activity . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 301
8.7. Antinociceptic activity. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 301
8.8. Antioxidant activity . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 302
8.9. Antiplasmodial activity . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 302
8.10. Antiviral activity . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 303
8.11. Clinical studies . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 304
8.12. Aphrodisiac activity . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 305
8.13. Contraceptive effect . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 305
8.14. Diuretic and antihypertentive activity . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 305
8.14.1. Clinical study . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 305
8.15. Hepatoprotective activity . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 305
8.16. Hypoglycemic and hypocholesterolemic activities . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 306
8.16.1. Clinical study . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 307
8.17. Immunomodulatory activity . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 307
8.18. Nephroprotective activity . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 307
8.19. Radioprotective effect . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 307
8.20. Spasmolytic activity . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 308
8.21. Effect on reproductive organs . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 308
9. Toxicological assessment and contraindications . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 308
10. Conclusion . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 309
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 309

1. Introduction whole plant is used in gonorrhea, menorrhagia and other genital


affections. It is useful in gastropathy, diarrhoea, dysentery, inter-
Bhumyaamalaki (Phyllanthus amarus Schum. & Thonn., Euphor- mittent fevers, ophthalmopathy, scabies, ulcers and wounds. It is
biaceae), which is widely spread throughout the tropical and also used as a good tonic. The Spanish name ‘chanca piedra’ means
subtropical countries of the world including India is most com- “stone breaker or shatter stone.” In South America, ‘chanca piedra’
monly used in the Indian Ayurvedic system of medicine in has been used to eliminate gall bladder and kidney stones, and to
problems of stomach, genitourinary system, liver, kidney and treat gall bladder infections (Foo and Wong, 1992), cardiovascular
spleen. P. amarus has been described in Ayurveda by the Sanskrit problems (Chevallier, 2000), and also a remedy around the world
name – Bhoomyaamalakee, Taamalakee and Bhoodhatree. It was for influenza (Foo, 1993a). P. amarus has a long history of use in
described to have the properties of Rasa, Guna, Veerya and Vipaaka. the treatment of liver, kidney and bladder problems, diabetes and
The Ayurvedic literature has shown its uses as Kaasahara (antitus- intestinal parasites. In Suriname (Northeastern part of South Amer-
sive), Shwaasahara (antispasmodic, antidyspnoic), Kaphapittahara ica), P. amarus is always sold as fresh and dry plant material in the
(which relieves the Kapha Pitta Dosha), Pipaasaaghna (which herb markets. Decoctions are used in herbal baths and after labor,
relieves Polydipsia), Raktapittahara (hemorrhage disease), Paan- cramps, asthma, uterus complaints and to treat stomachache (May,
duhara (antianemic), Kaamalaahara (which cures jaundice), 1982; Titjari, 1985; Heyde, 1990; Sedoc, 1992; Nanden, 1998). It is
Kushthaghna (indicated in leprosy), Daahaghna (refrigerant, a restoration herb and used as an appetizer and as tonic. It is also
relieves burning sensation), Kshatakshayaghna (indicated in used as colic. The plant, when boiled with the leaves, is considered
Trauma) and Mootrarogahara (which cures urinary disorders). The to be a diuretic and is used in treatment of diabetes, dysentery, hep-
use of P. amarus is gaining momentum because of its novel antiviral atitis, menstrual disorders, and skin disorders (Wessels Boer et al.,
activity against hepatitis B virus and for several other biological 1976; Tirimana, 1987; Heyde, 1968, 1990). Plant extracts are used
activities such as kidney and gallbladder stones, for cold, flu, tuber- as blood purifiers, for light malaria fevers and anemia. It helps to
culosis, and other viral infections; liver diseases and disorders release phlegm (Heyde, 1990) and to combat fever (Nanden, 1998).
including hepatitis, jaundice and liver cancer (Unander et al., 1993). This herb can be used for constipation also (Tjong and Young, 1989).
It also acts against liver cell toxicity and improves the immune P. amarus elaborates different classes of organic compounds of
system of patients and has been found effective against hepatitis medicinal importance including alkaloids, flavonoids, hydrolysable
A (Jayaram et al., 1997). P. amarus is often used in the traditional tannins (Ellagitannins), major lignans, polyphenols, triterpenes,
system of medicine for a variety of ailments including dropsy, dia- sterols and volatile oil. Many lignans were isolated from the
betes, jaundice, asthma and bronchial infections (Foo and Wong, plant viz., phyllanthin (a bitter constituent) and hypophyllan-
1992). In the Ayurvedic system of medicine it is used in problems thin (a non bitter constituent) (Row et al., 1967). The highest
of stomach, genitourinary system, liver, kidney and spleen. It is amounts of phyllanthin (0.7% w/w) and hypophyllanthin (0.3%
bitter, astringent, stomachic, diuretic, febrifuge and antiseptic. The w/w) have been reported in leaves whereas, in the stem these
Author's personal copy

288 J.R. Patel et al. / Journal of Ethnopharmacology 138 (2011) 286–313

are in minor quantities present (Sharma et al., 1993). Lig- Table 1


Ethnomedicinal uses of P. amarus in different countries worldwide.
nans isolated from P. amarus are phyllanthin, hypophyllanthin,
niranthin, phyltetralin, nirtetralin, isonirtetralin, hinokinin, linte- Region Ethnomedicinal uses
tralin, isolintetralin, demethylenedioxy-niranthin, 5-demethoxy- Amazonia Anodyne, apertif, blennorrhagia, carminative,
niranthin etc., flavonoïds such as gallocatechin, rutin, quercetin- colic, diabetes, digestive, diuretic, dropsy,
3-O-glucopyranoside, phyllanthusiin, quercetin, kaempferol dysentery, dyspepsia, emmenagogue, fever, flu,
3-␤-d-glucopyranoside, kaempferol etc., ellagitannins include gallstones, gonorrhea, itching, jaundice, kidney
ailments, kidney stones, laxative, malaria,
geraniin, amariin, furosin, geraniinic acid B, amariinic acid, amaru-
proctitis, stomachache, tenesmus, tonic,
lone, repandusinic acid A, corilagin, isocorilagin, elaeocarpusin, tumor, vaginitis, vermifuge
phyllanthusiin A, B, C, D and melatonin; securinega-type alka- Aruba Blood purifier
loids such as isobubbialine and epibubbialine and sterol such as Bahamas/Caribbean Antihepatotoxic, cold, constipation, fever, flu,
stomachache, typhoid, flatulence, vermifuge,
amarosterol A, amarosterol B.
appetizer
P. amarus had been reported to have pharmacological Barbados Abortifacient
effects such as antimicrobial, antiviral activities against hepati- Brazil Abortifacient, ache (joint), albuminuria,
tis B, chemoprotective, antimutagenic and hypoglycaemic agent. analgesic, antibacterial, anticancerous,
Methanolic extract of P. amarus exhibited immunomodulatory antidiabetic, anti-inflammatory, antilithic,
antispasmodic, antiviral, aperient, arthritis,
activity. Ellagitannins (geraniin and corilagin) were shown to be
biliary conditions, bladder problems, bladder
the most potent mediators of the antiviral HIV activity. Phyllan- stones, calculi, catarrh (liver and kidney),
thin and hypophyllanthin present in P. amarus exhibited antitumor chologogue, cystitis, deobstruent, diabetes,
activities against EAC in Swiss albino mice, cytotoxic effects on diaphoretic, digestion stimulant, diuretic,
fever, gallbladder, gallstones, gastritis,
K-562 cells, and hepatoprotective and antioxidant effects. The
gastrointestinal problems, gout, hepatitis,
present review assesses the potential of P. amarus in relation to its hydropsy, hypertension, hypoglycemic,
traditional uses and in terms of findings based on modern bioscien- jaundice, kidney, colic, kidney stones, liver,
tific research. The link between conventional remedies and recent malaria, muscle relaxant, obesity, prostatitis,
research in various areas has been well established in other plants purgative, renal colic, renal problems,
stomachic, sudorific, tonic, uric acid excess,
which facilitate to determine effective mode of action of plant
urinary problems, uterine relaxant
derived products. The plant is known to contain several pharmaco- Cuba Oedema and malaria
logical important biomolecules whose efficacy is well established Haiti Carminative, colic, digestive, diuretic, fever,
by several biochemical and pharmacological studies. This review indigestion, malaria, spasmolytic,
stomachache, stomachic, tenesmus
intent to compile various studies on this plant and critically eval-
India Anemia, asthma, astringent, bronchitis,
uates the issues related to ethnopharmacology, phytochemistry, conjunctivitis, cough, deobstruent, dropsy,
pharmacology, clinical studies and toxicology of P. amarus. diabetes, diarrhoea, diuretic, dysentery, fevers,
Table 1 represents ethnomedicinal uses of P. amarus in differ- eye disorders, galactagogue, genitourinary
ent countries and Table 2 represents the ethnomedicinal uses of P. disorders, gonorrhea, hepatitis, jaundice,
leucorrhea, menorrhagia, oligogalactia,
amarus used by different tribes of countries worldwide.
itchness, skin ulcers, sores, swelling,
ringworm, scabies, stomachic, thirst,
tuberculosis, tumor (abdomen), urogenital
2. Historical perspectives tract infections, warts, appetizer, dyspepsia
Indonesia Colic, cough, diuretic, eye diseases (external),
kidney diseases, stomachache, toothache,
P. amarus has been indexed in majority of published phytochem- veneral diseases
ical, pharmacological and ethno-botanical reviews and research Island of North Caicos Fever, prevention of intestinal worms
articles till date with different named. This species, P. amarus has Jamaica Diabetes, dysentery, diuretic, oedema,
been confused with Phyllanthus niruri Linn. In the past, Linnaeus’s gonorrhea, jaundice, stomachache
Malaya Caterpillar sting, dermatosis, diarrhoea,
P. niruri, though well defined in the ‘Hortus Cliffortianus’, became
diuretic, emmenagogue, itching, miscarriage,
confused owing to his erroneous reduction of other species to the purgative, renosis, syphilis, vertigo
synonyms of the former and subsequent misidentification by other Nigeria Malaria, chronic stomach pains, oral or vaginal
authors. The commonest weedy species so mistaken for P. niruri by thrush, alcoholic liver disease, hyperglycaemia,
Muller and others was defined as Phyllanthus swartzii Kosteletzsky urinary tract infection and venereal disease.
Taken with honey as aphrodisiac
in 1836, based on P. niruri of Swartz; but the earliest name of this Peru Calculus, diuretic, emmenagogue, gallstones,
species appears to be P. amarus Schum. & Thonn. For a full discus- hepatitis, kidney pain, kidney problems,
sion on the botanical description and nomenclature of this species kidney stones, renal problems, urinary
reference has been made to the excellent discussion by Webster infections, vermifuge
Trinidad Diuretic, veneral diseases
(1957). The earliest botanical descriptions could be observed from
United States Analgesic, bronchitis, chologogue, deobstruent,
South India, Sri Lanka and Indonesia which have cited Phyllanthus diabetes, fever, gallbladder problems,
urinaria and P. niruri respectively (Van Rhede, 1690; Rumphius, gallstones, gout, hepatitis, hypertension,
1750). In the Ayurvedic system in India, no definitive botanical kidney problems, kidney stones, liver disease,
description exists in the original literature and the plants indicated uric acid excess, urinary tract infections
Elsewhere Analgesic, antipyretic, appetite stimulant,
by many of the old Sanskrit words have been lost in course of his- blennorrhagia, bruises, chologogue, cough,
tory (Handa et al., 1951; Chopra et al., 1958). It was the opinion cuts, diabetes, diarrhoea, diuretic, dropsy,
of Dymock (1886) and Dymock et al. (1893) that the same com- dysentery, dyspepsia, emmenagogue, eye
mon names in a number of Indian languages apply for both P. niruri diseases, fever, gallstones, gonorrhea, itch,
jaundice, kidney disease, kidney stones,
and P. urinaria. Similar reports have stated that even the rather
laxative, malaria, menorrhagia, menstrual
visually different P. urinaria was often used interchangeably with problems, poultice, purgative, rectitis,
those species earlier known as P. niruri in the traditional system stomachache, tonic, tuberculosis, urinary tract
of India (Van Rhede, 1690; Dymock, 1886; Kirtikar and Basu, 1975) infections, vaginitis, venereal diseases
and suggested that the practioners did not differentiate it according
Author's personal copy

J.R. Patel et al. / Journal of Ethnopharmacology 138 (2011) 286–313 289

Table 2
Ethnomedicinal uses of Phyllanthus amarus Schum. & Thonn.

S.N. Place Local name Plant part Disease Method of use Reference
used

1 Dharapuram Taluk, Keelanelli Whole plant 1. Migraine (1) Whole plant is boiled in Balakrishnan et al. (2009)
Tamil Nadu, India 2. Jaundice gingelly oil, filtered and
applied on the head
(2) The fresh root is used with
water. Paste or fresh roots are
given orally
2 Paliyar tribals in Keelanelli Leaves Jaundice Leaf paste is given internally Ignacimuthu et al. (2008)
Theni district of
Tamil Nadu, India
3 Eastern part of Bhumiamla Whole plant, Gonorrhea and Decoction of leaves, sugar and Upadhyay et al. (2010)
Rajasthan, India leaves syphilis cumin seeds are taken orally to
Skin diseases, treat gonorrhea and syphilis.
malaria Leaves are crushed with salt to
make paste and applied locally
against skin diseases. Plant is
crushed into paste, mixed with
seed powder of pepper, candy
and water and taken as a
refrigerant. Decoction of whole
plant is taken as an
antimalarial
4 Uttara kannada, Nelli Whole plant Malaria Not stated Kuppusamy and Murugan (2010)
Western Ghats,
India
5 Eastern region of Nelanelli Root juice Jaundice Root juice is taken orally with Rajakumar and Shivanna (2009)
Shimoga district, cow’s milk early in the
Karnataka, India morning for 1 week
6 Dindigul District, Kizhnelli Leaves Menstrual problem Leaf extract with milk and Samuel and Andrews (2010)
Tamil Nadu, Indian onion is given during night,
three times once in 3 days
7 Buldhana district; Bhui-awala Whole plant Jaundice Extract, one spoonful per day Ahirrao and Patil (2010)
Maharashtra, for 3 days
Indian
8 North Andaman Nallesari Whole plant Jaundice Handful of leaves is crushed Prasad et al. (2008)
Island, India with a pinch of turmeric and
one teaspoonful of extract is
taken orally for 3–5 days
9 Sivagangai district, Keelaanelli Leaves 1. Diabetes Diabetes: Shanmugam et al. (2009)
Tamil Nadu, India 2. Jaundice (A) Leaves of Piper betle,
Cynodon dactylon, Azadirachta
indica and P. amarus are dried
and powdered with the stem
park of Syzygium cumini. The
powder is boiled in water and
the extract is given orally
(B) Leaf extracts of A. indica
and P. amarus are mixed and
given orally
Jaundice:
(A) Leaves of C. dactylon and P.
amarus are grounded with the
fruits of Piper nigrum and
extracted. The extract is given
orally
(B) Leaves of Eclipta alba, P.
amarus and Leucas aspera are
grounded and extracted. The
extract is given orally
(C) Leaf extracts of C. dactylon
and P. amarus are mixed and
given orally
10 Shimoga district of Nela nelli Leaves 1. Jaundice (1) Leaf paste with cardamom Mahishi et al. (2005)
Karnataka, India (Bhumyamalaki) 2. Chronic is taken internally, two tea
dysentery spoons daily
(2) Leaves are ground with
Acacia Senegal leaves, add
sugar and give orally, or tender
leaves ground with cow’s milk
curd given orally, for 2–5 days,
before food
11 Kattunaykas tribes Kila nelli Whole plant Jaundice 15 mL whole plant juice is Udayan et al. (2007)
of Mudumalai taken internally in empty
Wildlife Sanctuary, stomach along with one
Nilgiris district tumbler goat’s milk against
Tamil Nadu, India jaundice
Author's personal copy

290 J.R. Patel et al. / Journal of Ethnopharmacology 138 (2011) 286–313

Table 2 (Continued)

S.N. Place Local name Plant part Disease Method of use Reference
used

12 Kancheepuram Keezhanelli Leaves Jaundice Fresh leaves are ground and Muthu et al. (2006)
district Tamil mixed with a cup of cow or
Nadu, India goat’s milk and taken
internally to cure jaundice
13 India Bhumi amalaki Whole plant Liver disease, 20% whole plant and 10% Samy et al. (2008)
dyspepsia, Plumbago zeylanica (roots). 4 g
anorexia, moderate of powdered mixer is given to
constipation, the patient twice daily, half an
chronic colitis, hour before meals with water
irritable bowel
syndrome, urinary
tract infection
14 Northern India Bhui amla Whole Jaundice, Not stated Kala et al. (2006)
aphrodisiac,
dysentery
15 Sitamata Wildlife Not stated Leaves Syphilis, Leaf paste and decoction of Jain et al. (2005)
Sanctuary of gonorrhea, leaves
Chittorgarh and jaundice
Udaipur district
Rajasthan, India
16 Esan North East Abenaghe Leaves Stomachache Ground leaves with pepper and Idu et al. (2008a)
local govt. area of salt. Half of cup is taken twice
Edo State, Nigeria daily
17 Delta State Nigeria Ibuko-oyeke Leaves Stomachache The leaves are infused in Idu et al. (2008b)
alcohol and drunk as a remedy
for stomachache
18 Akwa Ibom State in Oyomokiso, Leaves Malaria Boiled in water as decoction. Ajibesin et al. (2008)
Nigeria aman keeden Use internally 4 times a day for
5 days
19 South West Nigeria Eyin olobe Whole plant Diabetes Decoction Abo et al. (2008)
20 West Africa Hlinvi Arial Diabetes, fever, Not stated Adjanohoun et al. (1986)
malaria
21 Semi-arid Quebra-pedra Leaves Kidney problems Decoction of leaves soaking Cartaxo et al. (2010)
Northeasten Brazil drink
22 Dangme West Ofobi okpabi Whole plant Malaria Boil about 50 g of plant in 1 L of Asase et al. (2010)
district of Ghana water and drink a cupful of
decoction three times daily
after meals until recovered.
Children should take half of the
dose. Sweeten with honey or
sugar if desired. The decoction
may cause dizziness
23 Surinamese Fini bita Whole plant Stomach-ache, Whole plant is boiled in water Van Andel and Westers (2010)
migrants in cleaning uterus, or soaked in alcohol and drunk
Netherland laxative, health to purify the blood. These
promotion, disease so-called BITAS were said to
prevention promote one’s health, purity of
blood and prevent and cure
diseases like malaria, skin
sores, diabetes and pimples.
Bitter tonics are also reported
to be popular among
HIV-positives to support their
body function
24 Akha people in Yu Jae Leaves Rashes, itches Poultice Inta et al. (2008)
Thailand and China

Number of ethnomedicinal uses of P. amarus.


Jaundice = 14, malaria = 08, diabetes = 06, skin disease = 04, dysentery = 03, fever = 03, stomachache = 03, syphilis = 03, urinary tract infection = 03, gonorrhea = 02, constipa-
tion = 02, anorexia = 01, aphrodisiac = 01, chronic colitis = 01, dyspepsia = 01, laxative = 01, menstrual problem = 01.

to the current taxonomical understanding. One of the most difficult observed that P. amarus predominates over P. debilis in Madras area
problems in organising ethnobotanical data on Phyllanthus was the suggesting that it is an introduced species over the years. Samples of
identification of the correct species for citations under P. niruri. The the plants collected from the Madras area in 1983–1988 as P. niruri
true P. niruri Linnaeus type of the genus was collected in Barbados were predominantly P. amarus with occasional plants of P. debilis
and described in 1738 by Linnaeus (Linnaeus, 1738). Specimens of as identified by Grady webster, University of California (Unander
P. niruri have never been confirmed outside the America (Webster, et al., 1991) during the collaborative studies with the group led by
1957). According to Mitra and Jain (1985) of the Botanical Survey Baruch S. Blumberg, Fox Chase Cancer Centre, Philadelphia.U.S.A.
of India, the P. niruri in Flora of British India is indeed a mixture of
three distinct species viz. P. amarus Schum. & Thonn., Phyllanthus 3. Botanical description and vernacular names
fraternus webster and Phyllanthus debilis Klein ex willd, the true P.
niruri Linn being endemic to West Indies. The specimen of P. debilis P. amarus are erect annual herbs, 10–60 cm tall; main stem sim-
was collected from Madras in 1799 (Webster, 1957). However it is ple or branched, terrete smooth or scabridulous in younger parts.
Author's personal copy

J.R. Patel et al. / Journal of Ethnopharmacology 138 (2011) 286–313 291

Table 3
Vernacular name of P. amarus worldwide (Medicinal Plants of India, 1987,
http://knol.google.com/k/pankaj-oudhia/phyllanthus-amarus-l/3nerdtj3s9l79/20).

S.No. Language Vernacular names

1 Tamil Keelanelli (Keezhanelli)


2 Hindi Bhuyiavla, Jangli amla
3 Bengali Bhuiamala, Sadahazurmani
4 Gujarathi Bhonya anmali
5 Marathi Bhuivali
6 Oriya Bhuiaola, badianala
7 Bihari Muikoa, Kantara, Pirikantaru
8 Telugu Nela uirika, Nelavusari
9 Kanada Nela – nelli, Kirunelli
10 Malayalam Kizhkkayinelli, Kilanelli
11 Rajasthani Gugario
12 Sanskrit Bhumyaamlaki, Bhoodhatree, Thamalaki
13 English Black catnip, Carry me seed, Child
pick-a-back, Gale of wind, Gulf leaf flower,
Hurricane weed, Shatterstone, Stone breaker
Fig. 1. P. amarus plant. 14 French Poudre de plomb (Ivory coast)
15 German Weisse Blattblume
16 Spanish Yerba magica (Cuba)
Cataphylls, stipules 1.5–1.9 mm long, deltoid acuminate blade 17 African names Ahlivi (Mina-Togo), Bomagua kene (Ivory
1–1.5 mm long, subulate acuminate. Leaves 3–11 × 1.5–6 mm ellip- coast), Bounou (Ivory coast), Bounou honlin
(Ivory coast), Hinlinew (West Africa),
tic oblong obovate, oblong, or even obovate, obtuse, or minutely
Mokichinento (Korokoro–East Africa),
apiculate at apex, obtuse or slightly inequilateral at base, petioles Tsekulemegbe (Ouatchi-Togo)
0.3–0.5 mm long, stipules 0.8–1.1 mm long triangular accuminate. 18 North, Central and Black catnip, carry-meseed, chanca piedra,
Flowers minutes, proximal 2–3 axis with unisexual cymules, each South American djari-bita, egg woman, fini-bita, flor
consisting of 1 male and 1 female or 2–3 males and female or 1 names escondida, gale-of-(the)-wind, hurricane
weed, quebra-pedra, quinine creole, quinine
male and 2 females flower or combination thereof; male flowers
weed, seed-under-leaf, stone breaker and
pedicals at anthesis ca 1 mm long. Calyx lobe 5, subequal each yerba de la nina (Morton, 1981)
ca 0.7 × 0.3 mm elliptic or oblong elliptic and abruptly acute at
apex hyaline with unbranched mid ribs. Disc segments 5, roundish
stames 3 (rarely 2): filaments connate into a column 0.2–0.3 mm Bahamas, (Morton, 1981; Tirimana, 1987), Philippines or India
high autheros sessile a top dehiscing longitudinally. Female flow- (Chevallier, 2000). P. amarus is a common pantropical weed that
ers; pedicles 0.8–1 mm long, obtusely 4-gonous, dialated above, grows well in moist, shady and sunny places (Cabieses, 1993;
ca 1.5 mm in fruits, calyx five lobes, subequal. Lobes sometimes Nanden, 1998). P. amarus is distributed all over India and is con-
toothed at apex. Styles 3, free, more or less spreading, and shallowly sidered as the most widely occurring Phyllanthus species in India
bifid at apex; arms divergent (Mitra and Jain, 1985). The seed cap- (Chowdhury and Rao, 2002). The presence of dioceous cymules
sules on stalks are 1–2 mm long, round, smooth, 2 mm wide, with (Mitra and Jain, 1985) at the end of the branches is considered to
six seeds. When the fruits burst open the seeds are hurled away. be a unique character, though it resembles in many respects its
Seeds are triangular (like an orange segment); light brown, 1 mm close relatives, P. debilis and P. fraternus of the same sub-section
long, with 5–6 ribs on the back (Morton, 1981; Wessels Boer et al., Swartziani (Webster, 1994). This is the only sub-section in the sec-
1976) (Fig. 1 P. amarus plant). The vernacular names of P. amarus tion Phyllanthus, which consists of most widespread herbaceous
has been given in Table 3. species throughout the tropics (Jain et al., 2003).

4. Biogeography and ecology 5. Pharmacognostic characters

The genus Phyllanthus (Euphorbiaceae) consists of about 6500 Various species of Phyllanthus are being sold in India under the
species in 300 genera, of which 200 are American, 100 African, trade name ‘Bhuiamlki’. During market surveillance of herbal drug,
70 from Madagascar and the remaining Asian and Australasian it was observed that almost all the commercial samples, either
(Webster, 1994). Numerous species of this family are native to comprise of P. amarus Schum. & Thonn. or Phyllanthus maderas-
North, Central and South America (Unander et al., 1995). The name patensis Linn. or mixture of P. amarus, P. fraternus Webster and P.
‘Phyllanthus’ means “leaf and flower” because the flower, as well maderaspatensis Linn. The species admixtures have been assessed
as the fruit, seems to become one with the leaf (Cabieses, 1993). in raw drug trade of Phyllanthus in southern India using morpho-
The taxonomic revision on this genus by Webster included closely taxonomical characters and molecular analysis. The morphological
related genera P. amarus, under the sub-section Swartiziani of the analysis of these samples revealed six different species of Phyl-
section Phyllanthus. The nomenclature, taxonomic distinctness lanthus. Seventy-six percent of the market samples contained P.
and close relatives of P. amarus were addressed in detail based on amarus as the predominant species (>95%) and thus were devoid
morphology and geographical distribution (Chowdhury and Rao, of admixtures. The remaining 24% of the shops had five different
2002). It is said to be related to P. abnormis, which is endemic to species namely P. debilis, P. fraternus, P. urinaria, P. maderaspatensis,
sandy areas in Texas and Florida of southern USA. It is therefore and Phyllanthus kozhikodianus. Species specific DNA barcode signa-
most likely that P. amarus originated in the Caribbean area as a tures were developed for Phyllanthus species using the chloroplast
vicarious species of P. abnormis of the southern United States and DNA region, psbA-trnH. The trade sample identities were validated
has spread around the tropics by trading vessels (Webster, 1957). P. and confirmed by these species specific DNA barcodes (Srirama
amarus is widely distributed in all tropical and subtropical regions et al., 2010). The detailed botanical evaluation of three species was
of the world. Paleobotanical studies have not found the exact carried out with the aim to establish the identification markers of
geographic origin of this plant. It is indigenous to the rainforests this plant. Some reliable diagnostic characters were specified as,
of the Amazon and other tropical countries like India, China, and number of sepals five in P. amarus and six in P. fraternus and P.
Author's personal copy

292 J.R. Patel et al. / Journal of Ethnopharmacology 138 (2011) 286–313

maderaspatensis; the female flowers of P. maderaspatensis are much analysis was carried out using an Agilent 6890N GC system using
larger in size as compared to P. amarus and P. fraternus, in which flame ionization detector (FID) at the temperature 300 ◦ C. Results
these are almost equal in size; TS of branchlet circular, leaf margin from analysis of the oil of P. amarus revealed that 82 identified
crenate in P. amarus and in P. fraternus branchlets have six notches compounds were responsible for 87.6% of the oil content. The oil
with serrated leaf margins, while the leaf margins are crenate and was characterized by the dominance of linalool (36.4%) and phytol
bulbous in P. maderaspatensis. Preliminary phytochemical screen- (13.0%). Other significant compounds were hexahydrofarnesyl ace-
ing of the extractives showed that the triterpenoids were noticed in tone (3.4%), pentacosane (2.5%), naphthalene (2.4%), (E)-b-ionone
the hexane extract of P. amarus and P. fraternus and in chloroform (2.3%), nonacosane (2.1%), tetracosane and octacosane (ca. 1.7%).
extracts of P. maderaspatensis, however, glycosides in the alcoholic Eight of the components were in trace amount (less than 0.1%),
extract of P. amarus and P. fraternus only. Likewise, the percent- but they are likely important in the characteristics of the oil. Two
age of sugar and tannins were quite high in P. maderaspatensis and components with relative retention index (RRI) of 2692 and 2700
tannins were almost nil in P. fraternus. On the contrary, the com- were tentatively identified as nonyl phenol isomers. The classes of
parative thin layer chromatography (TLC) finger print profiles of P. compounds present in P. amarus oil are monoterpene hydrocar-
amarus and P. fraternus were almost similar and showed eight com- bons (0.2%), oxygenated monoterpenoids (11.0%), sesquiterpene
mon bands at Rf 0.04, 0.28, 0.38, 0.44, 0.47, 0.55, 0.60, 0.63 of light hydrocarbons (1.3%), oxygenated sesquiterpenoids (3.3%), diter-
green, greyish green, grey, faint green, light green, faint pink, light penoids (8.5%), aliphatic alcohols (51.2%), fatty acids (3.9%),
green, greyish green, respectively. However, in P. maderaspatensis aldehydes (8.0%), ketones (0.5%) and esters (0.3%) (Moronkola
only three bands at Rf 0.04, 0.28 and 0.38 were observed while one et al., 2009). Two new lignans, 3-(3,4-dimethoxy-benzyl)-4-
major band of bright blue colour was observed at Rf 0.9 in P. frater- (7-methoxy-benzo[1,3]dioxol-5-yl-methyl)-dihydrofuran-2-one
nus only. Therefore, all these three species could be differentiated and 4-(3,4-dimethoxy-phenyl)-1-(7-methoxy-benzo[1,3]dioxol-
on the basis of macro and microscopic characters, physico-chemical 5-yl)-2,3-bis-methoxymethyl-butan-1-ol were isolated from
values, high pressure thin layer chromatography (HPTLC), finger- the leaves of P. amarus (Singh et al., 2009). A phytochemical
print profile, and detection of phyllanthin and hypophyllanthin investigation of methanolic extract obtained from the whole
as marker components (Khatoon et al., 2006). Macromorphology, plant of P. amarus, revealed the presence of six bioactive lig-
micromorphology, histochemical and physical pharmacognostic nans [isolintetralin (2,3-demethoxy-seco-isolintetralin diacetate),
studies of P. amarus revealed certain diagnostic uncommon char- demethylenedioxy-niranthin, 5-demethoxy-niranthin, niranthin,
acters: basal submarginal venation formed by curving of almost phyllanthin and hypophyllanthin] and one triterpene 2Z, 6Z, 10Z,
unbranched lateral veins, 4–6 angled cortical fibres (TS), 1–2 seriate 14E, 18E, 22E-farnesyl farnesol (Maciel et al., 2007). Phytochemical
xylem rays, crystals concentrated along the veins (mostly rosette), evaluation of P. amarus showed to contain high level of saponins
combination of paracytic and anomocytic stomata, sinuous epider- and tannins at 24.05 and 17.50%, respectively, but with low
mal cell walls, vessel members tailed on two ends; high frequency content of cyanogenic glycosides (1.46%). The plant contained
of crystals in leaf (87.5 mm−2 ), stomatal index, palisade ratio, etc. high percentage level of fibre (24.50%) and carbohydrate (45.52%),
Additionally, distribution of alkaloidal reaction and protein in the with approximate content of fat (6.03%), protein (6.10%) and ash
secondary xylem, extractive values, ash values, UV fluorescence (6.80%). The potassium and sodium contents were high at 150.30
were also distinctive characterstics (De and Datta, 1990). and 228.20 mg per 100 g dry weight, respectively, while magne-
sium, calcium, iron and phosphorus were all low at 2.40, 1.60, 1.65
and 1.00 mg per 100 g dry weight, respectively (Igwe et al., 2007).
6. Phytochemical studies The crude extract of phyllanthin was obtained from P. amarus
using solvents of varied polarity. The presence of pyrrolizidine
The secondary metabolites present in P. amarus are alkaloids, type of alkaloids was reported in extract of P. amarus. These are
flavonoids, hydrolysable tannins (Ellagitannins), major lignans, securinine, dihydrosecurinine, tetrahydrosecurine, securinol-B,
polyphenols, triterpenes, sterols and volatile oil. The main active phyllanthine, allosecurine, nor-securinine etc. (Kassuya et al.,
constituents of P. amarus are lignans (phyllanthin, hypophyllan- 2006). The whole plant of P. amarus has afforded new secosterols
thin, nirurin niranthin, phyltetralin, niranthine, nirtetralin etc. named as amarosterol-A characterized as 13, 14-seco-stigma
(Morton, 1981; Chevallier, 2000; Srivastava et al., 2008; Kassuya 5(6), 14(15)-diene-3-␣-ol and amarosterol-B characterized as 13,
et al., 2006; Huang et al., 2003; Maciel et al., 2007; Singh 14-seco-stigma 9(11), 14(15)-diene-3-␣-ol (Ahmad and Alam,
et al., 2009), flavonoids (quercetin, quercetrin, rutin, gallocat- 2003). Polyprenols comprising 11 and 12 isoprene units were the
echin, phyllanthusiin, kaempferol etc.), (Foo and Wong, 1992; dominant ones in the majority of the plants investigated. Out of
Foo, 1993a; Londhe et al., 2008; Morton, 1981), Ellagitannins viz. the 19 species studied the highest concentration of polyprenols
geraniin, amariin, furosin, geraniinic acid, amariinic acid, amaru- was observed in P. amarus (0.1–0.3%, dry weight) (George et al.,
lone, repandusinic acid, corilagin, isocorilagin, elaeocarpusin, 2001). Two new securinega types of alkaloids, isobubbialine
phyllanthin D gallic acid, repandusinic acid A etc. (Foo and Wong, and epibubbialine were isolated from the leaves of P. amarus, as
1992; Foo, 1993a; Foo, 1995), triterpenes (phyllanthenol, phyllan- well as the three known alkaloids, phyllanthine, securinine and
thenone, phytllantheol etc.) (Maciel et al., 2007; Foo and Wong, nor-securinine (Houghton et al., 1996). Chemical examination of
1992), alkaloids (securinine, dihydrosecurinine, tetrahydrose- the polar extractives of the aerial parts of P. amarus led to the
curinine, securinol, phyllanthine, allosecurine, nor-securinine, isolation of amariinic acid, a novel ellagitannin, together with
epibubbialine, isobubbialine, 4-methoxy dihydrosecurinine, 4- 1-O-galloyl-2, 4-dehydrohexahydroxydiphenoyl-glucopyranose,
methoxytetrahydrosecurinine, 4-hydrosecurinine etc.) (Houghton elaeocarpusin, repandusinic acid A and geraniinic acid B. The
et al., 1996; Kassuya et al., 2006; Foo and Wong, 1992), sterol structure of amariinic acid was established as the ring-opened
(amarosterol-A, amarosterol-B etc.) (Ahmad and Alam, 2003) and oxidized cyclohexentrione moiety of dehydrohexahydroxydiphe-
volatile oil (linalool, phytol etc.) (Moronkola et al., 2009). A detailed noyl attached to 0–4 of the glucose core (Foo, 1995). A novel
extraction, isolation and characterization method was optimized cyclic metabolite named amarulone was isolated from P. amarus
for phyllanthin (Hamrapurkar et al., 2009). (Foo, 1993a). Amariin, a novel hydrolysable tannin together with
The oils obtained from P. amarus were analyzed for its geraniin, corilagin, 1,6-digalloylglucopyranoside as well as rutin
constituents by means of gas chromatography (GC) and gas chro- and quercetin-3-O-glucopyranoside were isolated from the polar
matography coupled with mass spectrometry (GC/MS). The GC fraction of P. amarus. The structure of amariin was established as
Author's personal copy

J.R. Patel et al. / Journal of Ethnopharmacology 138 (2011) 286–313 293

Table 4
Phytoconstituents reported in P. amarus.

S. No. Secondary metabolites Phyto-constituents References

1 Lignans Phyllanthin, hypophyllanthin, niranthin, Morton (1981), Sharma et al. (1993),


phyltetralin, nirtetralin, isonirtetralin, Chevallier (2000), Srivastava et al.
hinokinin (2008), Kassuya et al. (2006), Huang
et al. (2003), Singh et al. (2009)
Lintetralin, isolintetralin, Maciel et al. (2007)
demethylenedioxy-niranthin,
5-demethoxy-niranthin
(3-(3,4-dimethoxy-benzyl)-4-(7-methoxy- Singh et al. (2009)
benzo[1,3]dioxol-5-yl-methyl)-dihydrofuran-
2-one,
4-(3,4-dimethoxy-phenyl)-1-(7-methoxy-
benzo[1,3]dioxol-5-yl)-2,3-bis-
methoxymethyl-butan-1-ol
2 Flavonoids Rutin, astragalin, kaempferol, Morton (1981), Foo and Wong (1992),
quercetin-3-O-glucoside, quercetin, quercitrin Foo (1993a, 1995), Londhe (2008)
3 Hydrolysable tannin Tannin precursors Gallic acid, ellagic acid, gallocatechin Foo (1993a, 1995)
(Ellagitannins)
Simple tannins 1,6-digalloylglucopyranose, 4-O-galloylquinic Foo (1993a, 1995)
acid
Complex tannins Geraniin, amariin, furosin, geraniinic acid B, Foo and Wong (1992), Foo (1993a,
amariinic acid, amarulone, repandusinic acid A, 1995)
corilagin, isocorilagin, elaeocarpusin,
phyllanthusiin A, B, C and D, melatonin
4 Alkaloids Securinine, dihydrosecurinine, Houghton et al. (1996), Kassuya et al.
tetrahydrosecurinine, securinol, phyllanthine, (2006)
allo-securine, nor-securinine, epibubbialine,
isobubbialine
4-methoxy-nor-securinine, 4-methoxy Foo and Wong (1992)
dihydrosecurinine,
4-methoxytetrahydrosecurinine, 4
hydrosecurinine
Phenazine and phenazine derivatives Foo (1993a)
5 Triterpenes 2Z, 6Z, 10Z, 14E, 18E, 22E-farnesylfarnesol Maciel et al. (2007)
Lupeol, phyllanthenol, phyllanthenone, Foo and Wong (1992)
phyllantheol, Oleanolic acid, ursolic acid
6 Sterols Amarosterol A, amarosterol B Ahmad and Alam (2003)
7 Volatile oil Linalool, phytol Moronkola et al. (2009)

1-galloyl-2,4: 3,6-bis-dehydrohexahydroxydiphenoyl- was methanol–water 66:34 (% v/v) which was very simple and cost
glucopyranoside in which the cyclohexenetrione portion of effective. The detection was carried out using variable wavelength
the dehydrohexahydroxydiphenoyl moieties were linked to the UV–vis detector set at 229 nm. Linearity for the developed method
O-3 and O-4 of the glucose moiety (Foo, 1993b). An unusual was found over the concentration range 1–50 ␮g/mL with a correla-
ellagitannin, phyllanthusiin D was isolated from the biologically tion coefficient of 0.999 (Alvari et al., 2011). An online-hyphenated
active polar fraction of P. amarus. Its structure was established high-performance liquid chromatography-photodiode array-mass
as 1-galloyl-2, 4-(acetonyl-dehydrohexahydroxydiphenoyl)-3, spectrometry (HPLC-PDA-MS) analytical method was developed
6-hexahydroxydiphenoyl-glucopyranoside (Foo and Wong, 1992). for the simultaneous determination of six lignans of therapeutic
Table 4 represents the secondary metabolites with their respective importance in four Phyllanthus spp. (P. amarus, P. maderaspatensis,
phytochemicals present in P. amarus. P. urinaria, and Phyllanthus virgatus). HPLC with monolithic reverse
phase silica column (4.6 × 100 mm) and simple isocratic elution of
7. Analytical techniques methanol–water mixed with dioxane facilitated the separation of
lignans of diverse nature such as diarylbutyrolactone, tetrahydro-
A HPLC analysis method was developed and validated to obtain furan, isomeric aryltetralin, and diarylbutane type for quantitative
an easily performable and inexpensive method for the standard- analysis. Targeted lignans viz. heliobuphthalmin lactone, virga-
ization of crude extract of P. amarus and ellagic acid. Ethanolic tusin, hypophyllanthin, phyllanthin, nirtetralin, and niranthin were
extract of whole plant of P. amarus was dissolved in DMSO, ultra- confirmed unambiguously in four Phyllanthus species by their
sonicated for 15 min, and diluted with 50% methanol. Analysis was abundant molecular adduct ions, retention time, UV, and mass
performed using water and methanol containing 0.06% TFA and spectra as compared with those of reference compounds. Advan-
the peaks were detected at 254 nm. Ellagic acid showed a linear tages and limitations of both detection techniques for qualitative
relationship in the range of 1.74–20.91 ␮g/mL and a single-point (fingerprinting) and quantitative analysis of the above mentioned
calibration was allowed. The method was shown to be precise with lignans in four Phyllanthus spp. are discussed. The method was vali-
respect to time (RSD of 1.84%, 3 days, n = 6) and concentration (RSD dated following international guidelines. The described method can
of 2.54%, three levels, n = 6). The overall mean content of ellagic acid be utilized for assays and stability tests of P. amarus extracts as well
was 2.06%. A recovery experiment was performed and it showed as traditional Indian medicine based on Phyllanthus herb (Shanker
an accuracy of 100.4% (Dhooghe et al., 2011). A simple, specific and et al., 2011). Phyllanthin was extracted from the plant P. amarus
precise RP-HPLC method has been developed and validated for the by Soxhlet and supercritical-fluid extraction (SFE) and isolated by
estimation of phyllanthin, present in P. amarus. Furthermore, the column chromatography. A HPTLC method was established and
developed method was also used to successfully quantify the phyl- validated for analysis. The method was used for quantitative anal-
lanthin in plant extract. The mobile phase optimized for RP-HPLC ysis and macro and micro fingerprinting analysis of phyllanthin.
Author's personal copy

294 J.R. Patel et al. / Journal of Ethnopharmacology 138 (2011) 286–313

Fig. 2. Structures of Lignans isolated form P. amarus.

This study also revealed that SFE enabled more efficient isola- hypophyllanthin at Rf 0.3 and 0.4 respectively, by densitometric
tion of phyllanthin than Soxhlet extraction (Hamrapurkar et al., scanning at 254 nm (Mukherjee et al., 2006). Separation of phyl-
2010; Annamalai and Laxmi, 2009). Phyllanthin was isolated from lanthin and hypophyllanthin was carried out on silica gel 60F254
the aerial parts of P. amarus by silica gel column chromatography layers eluted with hexane:acetone:ethyl acetate (74:12:8), and the
employing gradient elution with hexane–ethyl acetate solvent mix- analytes were visualised through colour development with vanillin
ture. It was obtained in high yields (1.23%), compared to reported in concentrated sulfuric acid and ethanol. Scanning and quantifica-
procedures and the purity was ascertained by HPTLC and reversed tion of spots was performed at 580 nm. Recoveries of phyllanthin
phase HPLC analysis (Krithika et al., 2009). A sensitive, selective, and hypophyllanthin were 98.7 and 97.3%, respectively (Tripathi et
and robust HPTLC method using chiral TLC plates for qualitative al., 2006). Dhalwal et al. (2006) developed a HPTLC densitometric
and quantitative analysis of phyllanthin, hypophyllanthin, niran- method for simultaneous quantitation of phyllanthin, hypophyl-
thin, and nirtetralin, the active lignans of Phyllanthus species, was lanthin, gallic acid, and ellagic acid in the whole plant of P. amarus.
developed and validated. The effectiveness and role of various sta- They were found at levels of 0.37, 1.16, 0.36, and 0.17% (w/w),
tionary phases viz TLC silica gel 60F254 , HPTLC silica gel 60F254 , and respectively. For estimation of phyllanthin and hypophyllanthin in
chiral TLC plates in the quantitation were evaluated. A precoated P. amarus, an isocratic, reversed phase (RP) HPLC procedure has
chiral TLC plate was found suitable for the simultaneous analysis of been adopted using a mixture of pH 2.8 phosphate buffer and ace-
four pharmacologically active lignans. For achieving good separa- tonitrile as mobile phase, Cyano (CN) column as stationary phase
tion, the optimized mobile phase of n-hexane-acetone-1, 4-dioxane and UV detector. The developed method showed high resolution
(9:1:0.5 by volume) was used. A densitometric determination of the (R = 1.9), accuracy and reproducibility (Murali et al., 2001). The
above compounds was carried out in reflection absorption mode at invention concerns an improved procedure for the extraction and
620 nm. Optimized chromatographic conditions provided well sep- isolation of phyllanthin from P. amarus by pulverization of the dried
arated compact bands for the tested lignans. The calibration curves leaves with calcium carbonate, percolating the ground component
were found linear in the concentration range of 100–500 ng/band using a mixture of organic solvents at room temperature to 80 ◦ C,
(Srivastava et al., 2008). Sharma et al. (1993) have developed a removing the solvent by distillation, removing the grease by precip-
reversed phase HPLC procedure for standardizing P. amarus on the itation using organic solvents and filtration, vacuum sepration of an
basis of its two bioactive lignans, phyllanthin and hypophyllanthin. n-hexane fraction, and subjecting the phyllanthin-rich fraction to
The method has been found to be sensitive, precise and efficient to silica gel column chromatography, and further purifying the phyl-
record more than 98% recovery of these two lignans. The leaves lanthin by crystallization (Chaudhuri et al., 2001). A HPTLC method
of P. amarus were found to contain the highest amount of phyl- was described for the simultaneous determination of the bioac-
lanthin (0.7% w/w) and hypophyllanthin (0.3% w/w) as compared tive lignans, phyllanthin and hypophyllanthin from the dried whole
to other parts of the plant. A method for amount determination plant powder of P. amarus (Sane et al., 1997). A TLC – densitometric
of gallic acid in P. amarus capsules was established by HPLC (Guo method has been developed for the estimation of the two lignans
et al., 2007). Methanolic extract of P. amarus and standard phyllan- of P. amarus phyllanthin and hypophyllanthin (Deb and Mandal,
thin and hypophyllanthin were developed in the chromatographic 1996). Air-dried leaves of P. amarus were extracted with 0.5 M HCl
conditions. They showed the presence of standard phyllanthin and and filtered. The filtrate was basified with 10% aqueous Na2 CO3
Author's personal copy

J.R. Patel et al. / Journal of Ethnopharmacology 138 (2011) 286–313 295

8.2. Antibacterial activity

Hexane, methanol and water extracts of aerial parts of P.


amarus were screened for antimicrobial activities against Bacil-
lus subtilis, Escherichia coli, Pseudomonas aeruginosa, Salmonella
typhi, Staphylococcus aureus and Candida albicans using the agar-
cup diffusion protocol. The aqueous and methanolic extracts of
P. amarus were active against all the test microorganisms. The
Fig. 3. Structures of flavonoids isolated from P. amarus.
methanolic extract of P. amarus also showed a broad spectrum of
activity with a minimum inhibitory concentration of 1.56 mg/mL
and extracted with CHCl3 to yield an oily residue (165 mg). Exam- against all the test microorganisms. The extracts were also screened
ination by TLC showed the presence of five Dragendorff-positive for secondary metabolites and the result indicated the presence
zones. The residue was then subjected to preparative TLC on sil- of alkaloids, saponins, tannins and terpenoids (Alli et al., 2011).
ica gel to yield five compounds (PA1–PA5). PAl (0.026% yield), The 80% methanolic extracts obtained from seven Phyllanthus sp.
PA2 (0.021% yield) and PA3 (0.018% yield w/w) were character- were evaluated for antibacterial activity using the broth micro-
ized as phyllanthine, securinine and nor-securinine, respectively dilution assay. Best antibacterial activity was obtained by P. amarus
by comparison of their spectral data with standard values. Two against S. aureus (gram-positive) with a MIC value of 17.7 ␮g/mL
new securinega-type alkaloids, isobubbialine and epibubbialine (Eldeen et al., 2011). The antibacterial activity of ethanolic extracts
were isolated from the leaves of P. amarus, as well as the three of the root and leaf of P. amarus was assessed against extend
known alkaloids, phyllanthine, securinine and nor-securinine. The spectrum ␤-lactamase (ESBL) producing E. coli isolated from the
structures of the unknown compounds were determined by means stool samples of HIV sero-positive patients with or without diar-
of UV, IR, mass and NMR spectroscopy (Houghton et al., 1996). rhoea using Bauer disc diffusion method. The strains isolated
The 70% aqueous acetone extract of the aerial part of P. amarus from both HIV sero-positive patients were susceptible to vari-
was fractionated over a column of Sephadex LH20 using aqueous ous concentrations of the extracts (5, 10, 20, 40 and 80 mg/mL).
methanol to yield various fractions subjected to repeate chro- The mean zones of inhibition of the root extracts ranged from
matographic treatment alternating between MCI-gel CHP-20 using 8.0 ± 0.33 to 25.0 ± 1.50 mm against ESBL E. coli while the mean
aqueous methanol and Sephadex LH20 with aqueous ethanol led zones of inhibition the of the leaf extracts ranged from 8.0 ± 0.50
to the isolation of amariinic acid, a novel ellagitannin, together to 26.0 ± 1.00 mm against ESBL E. coli. The root extracts showed
with 4-O-galloylquinic acid, elaeocarpusin, furosin, geraniinic acid the highest zone of inhibition (25 ± 1.50 mm) against ESBL E. coli
B, amariinic acid and potassium salt of repandusinic acid B and their at 80 mg/mL while the leaf extracts showed the highest zone of
structures were established on the basis of chemical and 13 C NMR inhibition highest (26 ± 1.00 mm) against ESBL E. coli at 80 mg/mL.
spectrum (Foo, 1995). An unusual ellagitannin was isolated from The minimum inhibitory concentration (MIC) and minimum bac-
the biologically active polar fraction of aerial part of P. amarus. The terial concentration (MBC) of the plant extracts ranging from
hydrolysable tannin fraction was obtained by column chromatog- 5–20 to 5–30 mg/mL respectively (Akinjogunla et al., 2010). P.
raphy of the water soluble portion of the 70% aqueous acetone amarus was examined against ocular infections causing bacteria P.
extract of the aerial parts of the plant on Sephadex LH20 using aque- aeruginosa, Micrococcus lylae, Bacillus licheniformis, Staphylococcus
ous methanol. Further chromatography of the fraction on MCI-gel hominis, S. aureus, Staphylococcus haemolyticus, Micrococcus luteus,
CHP-20 yielded phyllanthusiin D as an amorphous powder which Bacillus lentus, Bacillus firmus and Pseudomonas stutzeri using agar-
gave a [M-H] ion peak at m/z 991 with fast atom bombardment well diffusion method. Results revealed that P. amarus exhibited
(FAB) mass spectrometry (Foo and Wong, 1992). remarkable bioactivity against M. lylae, S. haemolyticus, B. lentus,
Chemical constituents isolated and characterized so far from P. B. firmus, P. stutzeri, P. aeruginosa and S. aureus (Koday et al., 2009).
amarus and their structures are given in Figs. 2–8. Antimicrobial properties aqueous and methanolic extracts of leaves
of P. amarus at the concentration of 100 mg/mL were tested against
E. coli, Streptococcus spp, Klebsiella spp, Pseudomonas spp and Staphy-
8. Pharmacological activity lococcus spp. aqueous and methanolic extract of P. amarus. The
crude extracts were observed to inhibit the growth of E. coli, Strep-
8.1. Antiamnesic activity tococcus and S. aureus. Methanolic extract of the plant was more
effective (6–11 mm) than aqueous extracts (5–10 mm) inhibiting
The effect of aqueous extract of leaves and stems of P. amarus the growth of pathogenic bacteria but was less potent when com-
was evaluated on cognitive functions and brain cholinesterase pared to that of ofloxacin (19 mm) and ciprofloxacin (21 mm) used
activity in male Swiss albino mice. P. amarus (50, 100 and as positive controls (Okoli et al., 2009). 80% methanolic extract of
200 mg/kg) produced a dose-dependent improvement in memory whole plant of P. amarus showed the least MIC on the tested bac-
scores of young and older mice. P. amarus also reversed successfully teria viz. B. stearothermophilus, S. aureus, B. subtilis, M. leuteus, S.
the amnesia induced by scopolamine (0.4 mg/kg, i.p.) and diazepam typhi, Enterobacter aerogens, Proteus mirabilis, and Proteus vulgaris.
(1 mg/kg, i.p.). Interestingly, brain cholinesterase activity was also The MIC and MBC were 30 and 40 ␮g/mL respectively. Ampicillin
reduced. Piracetam 400 mg/kg, i.p. was used as positive control was used as standard (Komuraiah et al., 2009). The essential oil and
(Joshi and Parle, 2007). Nootropic activity of [6]-gingerol and phyl- its fractions obtained from fresh leaves and seeds of P. amarus were
lanthin was studied in mice using elevated plus maze and passive tested on 12 microorganisms including yeast, gram-positive and
avoidance paradigm. [6]-gingerol (25 and 50 mg/kg, p.o.) and phyl- gram-negative bacteria viz. B. subtilis, Citrobacter sp., E. coli (isolate),
lanthin (7.5 and 15 mg/kg, p.o.) significantly attenuated amnestic E. coli (ATCC 25922), Enterococcus faecalis, Klebsiella pneumoniae,
deficits induced by diazepam, scopolamine (0.4 mg/kg, i.p.) and P. aeruginosa, P. mirabilis, Staphylococcus albus, S. aureus (ATCC
natural aging. [6]-gingerol and phyllanthin increased step down 25923), S. aureus (isolate) and C. albicans. All the samples of essen-
latencies significantly in the aged mice, diazepam and scopolamine tial oil and fractions demonstrated activity (11–20 mm diameter
induced amnesic mice as compared with piracetam (200 mg/kg, zone of inhibition) against the microorganisms except P. aeruginosa.
i.p.). [6]-gingerol and phyllanthin significantly decreased whole 0.05% ciprofloxacin (≥21 mm diameter zone of inhibition) was used
brain acetyl cholinesterase activity (Joshi and Parle, 2006). as positive control (Ogunlesi et al., 2009). Crude aqueous (hot and
Author's personal copy

296 J.R. Patel et al. / Journal of Ethnopharmacology 138 (2011) 286–313

Fig. 4. Strutures of hydrolysable tannins (Ellagitannins) isolated form P. amarus.


Author's personal copy

J.R. Patel et al. / Journal of Ethnopharmacology 138 (2011) 286–313 297

Fig. 4. Continued.
Author's personal copy

298 J.R. Patel et al. / Journal of Ethnopharmacology 138 (2011) 286–313

Fig. 5. Structures of alkaloids isolated from P. amarus.

cold water) and ethanolic extracts of leaves of P. amarus was inves- methanolic extract of whole plant of P. amarus at the concentration
tigated for antimicrobial activity against S. typhi. Ethanolic extracts of 5, 10, 25, 50, 100, 200, 400, 800, and 1000 mg/mL was studied
of P. amarus revealed the strongest activity against S. typhi with against drug resistant pathogenic bacterial strains, Shigella dysen-
8.0 mm zone of growth inbibition followed by hot water (4.7 mm) teriae 1, S. dysenteriae 2, S. boydii 8, S. aureus ML267, S. aureus
and cold water (3.8 mm). Ciprofloxacin had the highest zone of inhi- NCTC 7447, Streptococcus pneumoniae 7465, E. coli ROW 7/12, E. coli
bition (9.0 mm) against S. typhi followed by ofloxacin (6.0 mm) and CD/99/1, B. subtilis CD/99/1, Salmonella typhimurium ATCC 6539,
amoxicillin (4.0 mm) (Oluwafemi and Debiri, 2008). Antimicrobial Vibrio cholerae 8531, P. aeruginosa 7241, and K. pneumoniae RM 8/98
effect of the plant extracts of P. amarus showed that the organic by disc diffusion and agar dilution method. Shigella spp. were found
and aqueous extracts of P. amarus were inhibitory to Streptococ- to be inhibited at the least concentration (25 mg/mL) and found to
cus faecalis, while the extracts were not inhibitory to C. albicans. show the maximum diameter of zone of inhibition at the largest
Agar-well determined minimum inhibitory concentration (MIC) tested concentration of 1000 mg/mL comparable with sparfloxacin.
values ranged between 3.125 and 6.25 mg/mL while the disc diffu- Methanolic extract posses significant antimicrobial activity against
sion determined MIC values ranged between 6.25 and 25.0 mg/mL. all the tested strains; maximum inhibitory effect was noted against
The agar-well determined MIC values for the ethanolic P. amarus Shigella spp., E. coli, V. cholerae, and S. aureus. The extract was found
extracts (3.12 mg/mL) were lower than the corresponding disc dif- to be moderately active against S. typhimurium, P. aeruginosa, B.
fusion MIC determined values (6.25–25.00 mg/mL). Bacteriocidal subtilis, and other pneumonia causing strains of Klebsiella and Strep-
and bacteriostatic effect varied with, solvent type of extract, con- tococcus spp. The antibacterial action was mainly due to the isolated
centration and method of the test adopted. The active components phyllanthin (Mazumder et al., 2006). The ethanolic extracts of nine
of the plant have no antifungal effect on the tested yeast, C. albi- Peruvian medicinal plants including P. amarus were screened for
cans (Okigbo and Igwe, 2007). The antimicrobial potential of the antimicrobial activity against Bacillus cereus ATCC 11,778, B. subtilis

Fig. 6. Structures of triterpenes isolated from P. amarus.


Author's personal copy

J.R. Patel et al. / Journal of Ethnopharmacology 138 (2011) 286–313 299

as determined by the MTT assay. Treatment of cell lines with


P. amarus produced cytotoxicity after 24 h. The maximum effect
was observed in DLA cell lines where treatment with P. amarus
at a concentration of 500 ␮g/mL produced cytotoxicity IC50 value
was 102 ± 1.38 ␮g/mL. It also induced the formation of apoptotic
bodies with characteristic features like plasma membrane invagi-
nation, elongation, fragmentation, and chromatin condensation.
P. amarus at concentrations of 100 and 200 ␮g/mL has shown to
induce DNA-fragmentation (Harikumar et al., 2009). Oral adminis-
tration of 75% methanolic extract of aerial parts (stem and leaves)
Fig. 7. Structures of steroids isolated from P. amarus.
of P. amarus was found to enhance the life span of leukaemia
harboring animals and decrease the incidence of anemia. Treat-
ment with P. amarus (750 and 250 mg/kg) induced the expression
of p53 and p45NFE2 and decreased the expression of Bcl-2 in
the spleen of infected mice. Histopathological evaluations of the
spleen demonstrated that administration of P. amarus decreased
the infiltration of leukemic cells into the sinusoidal space when
Fig. 8. Structures of volatile oils isolated form P. amarus.
compared with the vehicle treated group (Harikumar and Kuttan,
2009). A mixture of phyllanthin and hypophyllanthin (1:1), iso-
ATCC 6633, Bacteroides fragilis ATCC 25,285, E. faecalis ATCC 29,212, lated from P. amarus exhibited antitumor activities against EAC in
E. coli ATCC 25,922, P. aeruginosa ATCC 27,853, S. aureus ATCC Swiss albino mice. The decrement of tumor volume and packed
25,923, Staphylococcus epidermidis ATCC 12,228, and Streptococ- cell volume and viable cell count were observed in lignan treated
cus pyogenes ATCC 19,615. Among the plants tested, 80% ethanolic mice when compared only to EAC tumor bearing mice. Treat-
extract of aerial parts of P. amarus showed the most promising ment with test compounds increased the survival time and normal
antibacterial properties, inhibiting all of the strains tested with peritoneal cell count. Hematological parameters, PCV which were
minimum inhibitory concentrations (MICs) ranging from 0.25 to altered by tumor volume inoculation, were restored consider-
16 mg/mL (Kloucek et al., 2005). ably (Islam et al., 2008a). An alcoholic extract of aerial parts of
P. amarus was found to inhibit cytochrome P450 enzymes both
8.3. Anticancer activity in vivo as well as in vitro. It was studied using specific resorufin
derivatives, as substrate for isoenzymes in the P450 super family.
Cytotoxicity of the crude extracts (aqueous and methanolic) Concentration needed for 50% inhibition of 7-ethoxyresorufin-
and their two fractions of P. amarus, were screened using the O-deethylase (EROD), CYP1A1 was 4.6 ␮/mL while concentration
MTS (3-(4,5-dimethylthiazol-2-yl)-5(3-carboxymethoxyphenyl)- needed for 7-methoxyresorufin-O-demethylase (MROD) CYP1A2
2-(4-sulfophenyl)-2H-tetrazolium) reduction assay. It was shown was 7.725 ␮/mL and 7-pentoxyresorufin-O-depentylase (PROD),
to inhibit MCF-7 (breast carcinoma) and A549 (lung carcinoma) CYP2B1/2 was found to be 4.18 ␮/mL indicating that the extract
cells growth with IC50 values ranging from 56 to 126 ␮g/mL and inhibited the P450 enzymes at very low concentration. Extract also
150–240 ␮g/mL for methanolic and aqueous extracts respectively. inhibited the activity of aniline hydroxylase (an indicator of CYP
In comparison, they have lower toxicity on normal cells with cell 2E1 activity, IC50 50 ␮/mL) and aminopyrine demethylase (an indi-
viability of 50% when treated up to 1000 ␮g/mL for both aqueous cator of CYP 1A, 2A 2B, 2D and 3A activity, IC50 > 1000 ␮g/mL).
and methanolic extracts. After determining the non-toxic effective Oral administration of the extract was also found to reduce the
dose, several antimetastasis assays were carried out and P. amarus elevated P450 enzyme activities produced by phenobarbitone by
extracts were shown to effectively reduce invasion, migration, and 50% at 250 mg/kg body weight. Extracts of P. amarus has pre-
adhesion of both MCF-7 and A549 cells in a dose-dependent man- vented or stopped the cells from mutation with the existence of
ner. This was followed by an evaluation of the possible modes chemical agents (Hari Kumar and Kuttan, 2006). N-methyl N -
of cell death that occurred along with the antimetastatic activ- nitro-N-nitrosoguanidine (MNNG) induced stomach cancer in male
ity. P. amarus was shown to be capable of inducing apoptosis in Wistar rats was significantly inhibited by the administration of
conjunction with its antimetastastic action, with more than 3-fold 75% methanolic extract of aerial parts of P. amarus at a dose of
increase of caspases-3 and -7, the presence of DNA-fragmentation 150 and 750 mg/kg body weight. It also reduced the incidence of
and terminal deoxynucleotidyl transferase mediated dUTP nick end gastric neoplasms in rats (44%) as well as their numbers. The ele-
labeling assay (TUNEL)-positive cells. The ability of P. amarus to vated enzymes level in the stomach was also found to reduce by
exert antimetastatic activities is mostly associated to the presence P. amarus treatment (Raphael et al., 2006). 70% ethanolic extracts
of polyphenol compounds in its extracts (Lee et al., 2011). The of three Phyllanthus species, P. urinaria, P. amarus and P. debilis
methanolic extract of P. amarus hairy roots revealed potent antipro- at the concentration of 10 ␮g/mL significantly inhibited the pro-
liferative activity in the MCF-7 cells through induction of apoptosis liferation of the HepG2 cells. The extracts induced apoptosis by
mediated by increased intracellular reactive oxygen species (ROS) inducing caspase-3. Further confirmation of extract-induced apo-
in conjunction with decreased mitochondrial membrane poten- ptosis was obtained by demonstrating that the Bcl-2/Bax ratio
tial (Abhyankar et al., 2010). The effects of aqueous extract of decreased in response to treatment with the extracts detected the
whole plant of P. amarus against Cr(VI)-induced oxidative toxicity mechanism by which the Phyllanthus extracts induced apopto-
in vitro in MDA-MB-435S human breast carcinoma cells revealed sis. These findings demonstrated that Phyllanthus extracts induce
a distinct decline in Cr(VI)-induced cytotoxicity was noticed in TNF-␣ production from the hepatocellular carcinoma cells while
MDA-MB-435S cells with an increase in extract dosage. Its phenolic inhibiting production of potent anti-apoptotic genes IL-8 and COX-
constituents simultaneously may inhibit Cr(VI)-induced oxidative 2. Untreated cells (control) received only 0.01% DSMO (Sureban
toxicity to MDA-MB-435S cells (Guha et al., 2010). In vitro, apop- et al., 2006). Two human leukaemia cell lines were employed:
totic effect of 75% methanolic extract of aerial parts (stem and K-562 and its vincristine-resistant counterpart Lucena-1, and Pgp-
leaves) of P. amarus against Dalton’s Lymphoma Ascites (DLA) overexpressing subline to evaluate the possible cytotoxic effect and
cells in culture produced significant reduction in cell viability multidrug resistance (MDR) reversing properties of the extract and
Author's personal copy

300 J.R. Patel et al. / Journal of Ethnopharmacology 138 (2011) 286–313

compounds isolated from P. amarus. It was reported that Lucena- (2-AAF) and aflatoxin B1 at concentrations of 0.25–2 mg/plate.
1 was significantly more resistant to the cytotoxicity of P. amarus It was also found to inhibit mutagenicity induced by direct
derivatives: the hexane extract (100 ␮g/mL), the lignans-rich frac- acting mutagens sodium azide (NaN3 ), MNNG, and 4-nitro-0-
tion (LRF, 100 ␮g/mL) and the lignans nirtetralin (43.2 ␮g/mL), phenylenediamine (NPD), at concentrations of 1–0.25 mg/plate.
niranthin (43 ␮g/mL) or phyllanthin (43 ␮g/mL) exerted cytotoxic Urinary mutagenicity produced in rats by benzo[a] pyrene was
effects on K-562 cells with 40.3, 66.0, 62.0, 61.0 or 24.1% of cell found to be significantly inhibited by the oral administration of
death, respectively. The cellular toxicity observed on Lucena-1 was P. amarus extract (Raphael et al., 2002a). Administration of aque-
16.3, 40.4, 29.4, 30.2, or 24.8%, respectively. These results suggested ous extract of aerial parts of P. amarus increases the life span
a potential action of P. amarus derivatives as MDR reversing agents, of rats with hepatocellular carcinoma. The effect of P. amarus
mainly due to their ability to synergize with the action of conven- extract administration after induction of hepatocellular carcinoma
tional chemotherapeutics (Leite et al., 2006). Administration of 75% (HCC) by N-nitrosodiethylamine (NDEA) was studied in Wistar rats.
ethanolic extract of P. amarus at doses 250 and 750 mg/kg body Administration of an aqueous extract of P. amarus was found to
weight i.p. in mice for 14 days significantly reduced the myelosup- significantly increase the survival of hepatocellular carcinoma har-
pression and improved the WBC count, bone marrow cellularity boring animals. All the untreated rats died of tumor burden by
as well as the number of maturing monocytes. P. amarus admin- 33,791.6 weeks. Administration of P. amarus extract (150 mg/kg
istration was found to decrease the activity of phase I enzyme. body weight orally, 5 days weekly continuously for 54 weeks
Administration of P. amarus also increased the cellular glutathione or till they died) after tumor development, increased the sur-
(GSH) and glutathione-S-transferase (GST), thereby decreasing the vival of animals to an average of 52,292.3 weeks (Rajeshkumar
effect of toxic metabolites of cyclophosphamide (CTX) on the cells. and Kuttan, 2000). The P. amarus aqueous extract at the dose of
Administration of P. amarus did not reduce the tumor reducing 150 and 75 mg/kg body weight on male Wistar rats, 5 days prior
activity of CTX. In fact, there was a synergistic action of CTX and P. to NDEA treatment for 20 weeks signifcantly inhibit hepatocar-
amarus in reducing the solid tumors in mice. It was indicated that cinogenesis induced by NDEA in a dose-dependent manner. The
administration of P. amarus can significantly reduce the toxic side anticarcinogenic activity of the extract was evaluated by its effect
effects of CTX and is not interfering with the antitumor efficiency on tumor incidence, level of carcinogen-metabolizing enzymes,
of CTX. When the aqueous extract of P. amarus was administered level of liver cancer markers and liver injury markers. Animals
to cancer bearing mice, it lowered the tumor incidents, level of treated with NDEA alone showed 100% tumor incidence and sig-
carcinogen-metabolizing enzymes levels of liver cancer markers nifcantly elevated tissue level of drug metabolizing enzymes such
dose dependently (Kumar and Kuttan, 2005). The 75% methanolic as GST and aniline hydroxylase (AH). Treatment of extract signif-
extract of aerial parts of P. amarus has been administered orally (750 icantly reduced these levels. Level of ␥-glutamyl transpeptidase
and 250 mg/kg body weight) in the radiation (6 Gy) induced balb/c (GGT) were also found to be elevated both in serum and tissues
mice for its protective activity against carcinogenesis. The WBC of tumor bearing animals, while they were significantly reduced
count, bone marrow cellularity and ␣-esterase activity increased in the treated group. Similar reduction was seen in tissue level
significantly as compared to only radiation exposed mice. The of reduced glutathione. Serum level of lipid peroxide (LPO), alka-
antioxidant enzymes such as superoxided dismutase (SOD), cata- line phosphatase (ALP) and glutamate pyruvate transaminase (GPT)
lase (CAT), (glutathione-S-transferase) GST, glutathione peroxidase were also elevated. Morphology of liver tissue and level of marker
(GPX), and glutathione reductase, both in blood and tissue, which enzymes indicated that these extracts offered protection against
was reduced by radiation induced. P. amarus possesses the ability chemical carcinogen (Jeena et al., 1999). Aqueous extract of P.
to inhibit the unusual enzymatic pathways peculiar to cancer cells amarus was reported a potent inhibitor of the hepatocarcinogene-
proliferation and growth rather than a direct toxic effect of killing sis induced in rats by NDEA. None of the P. amarus extract treated
the different types of cancer cells (Kumar and Kuttan, 2004). The animals developed any tumor even 32 weeks after NDEA admin-
aqueous extract of P. amarus 150 and 750 mg/kg body weight thrice istration, whereas all of the control animals died due to tumor
weekly for 8 weeks treatment exhibited potent anticarcinogenic burden. Liver weight was increased in the NDEA-treated group,
activity against 20-methylcholanthrene (20-MC) induced sarcoma whereas it was not altered after treatment with NDEA and P.
development and increased the survival of tumor harboring balb/c amarus. The liver markers g-glutamyl transpeptidase, glutamyl-
mice. The extract administration (p.o.) was also found to prolong S-transferase, reduced glutathione and the aniline-4-hydroxylase
the life span of DLA and EAC bearing mice and reduced the vol- cytochrome P450 enzyme were elevated in NDEA-treated animals,
ume of transplanted solid tumors (Rajeshkumar et al., 2002). The whereas they were almost normal in animals treated with the car-
aqueous extract of the entire plant of P. amarus, at the concentra- cinogen plus P. amarus extract. Animals were elevated by NDEA
tion of 10, 25, 50 and 100 ␮g/plate showed an antimutagenic effect treatment, were also normal in the NDEA and P. amarus treated
against induction by 2-aminofluorene (AF2), 2-aminoanthracene group (Joy and Kuttan, 1998). Phyllanthus extract reduced the cyto-
(2AA) and 4-nitroquinolone-1-oxide (4-NQO) in S. typhimurium toxic action of lead nitrate and aluminum sulfate in bone marrow
strains TA98 and TA100, and in E. coli WP2 uvrA/pKM101. The cells of mice. Nickel clastogenicity was also found to be reduced
inhibition of N-ethyl-N-nitrosoguanidine (ENNG)-induced muta- in mice fed with Phyllanthus extract and ascorbic acid. These pos-
genesis was observed only with S. typhimurium TA100. The extract itive reports on the antigenotoxic efficacy of Phyllanthus extract
also exhibited activity against 2-nitrofluorene (2NF) and sodium was confirmed (Dhir et al., 1990). Lignans Phyllanthin, nirtetralin,
azide-induced mutagenesis with S. typhimurium TA98 and TA100, and niranthin were found to be very effective against cancer.
respectively. Based on the alkaline elution method, the plant extract
prevented in vivo DNA single-strand breaks caused by dimethyl- 8.4. Anti-diarrhoeal, gastroprotective and antiulcer activity
nitrosamine (DMN) in hamster liver cells. When the extract was
administered 30 min prior to the administration of DMN, the elu- Oral administration of absolute ethanol (1 mL/200 g body
tion rate constant decreased more than 2.5 times, compared to the weight) induced multiple, elongated, reddish bands of hemor-
control (Sripanidkulchai et al., 2002). Methanolic extract of stems rhagic erosions in rat gastric mucosa was studied. The ethanol
and leaves of P. amarus was tested for its antimutagenic activity control group had the highest ulcer index of 45.20 ± 2.39. Pre-
in S. typhimurium strains TA1535, TA100, and TA102 (Ames test). treatment with P. amarus leaves aqueous extract (500 mg/kg) and
P. amarus extract 500 mg/kg body weight for 12 days was able cimetidine (100 mg/kg) significantly inhibited (p < 0.001) ulcera-
to inhibit the activation and mutagenicity of 2-acetaminofluorene tion by 59.3 and 41.2% respectively. The acetone extract yielded a
Author's personal copy

J.R. Patel et al. / Journal of Ethnopharmacology 138 (2011) 286–313 301

dose-dependent percentage ulcer inhibition and the lower dosage amarus on different phases of inflammation were performed using
of the aqueous extract had a higher ulcer inhibition (Shokunbi and different phlogistic agents-induced paw oedema, carrageenan-
Odetola, 2008). Methanolic extract of leaves and stems of P. amarus induced air-pouch inflammation and cotton pellet granuloma in
at the dose of 50, 200, and 1000 mg/kg body weight p.o., on adult male Wistar rats. Methanolic extract at a dose of 250 mg/kg body
male Wistar rats significantly inhibited gastric lesions, induced weight p.o. significantly inhibited carrageenan, bradykinin, sero-
by intragastric administration of absolute ethanol (8 mL/kg body tonin and prostaglandin E1 -induced paw oedema, but failed to
weight). Mortality, increased stomach weight, ulcer index, and inhibit the histamine-induced paw oedema (Mahat and Patil, 2007).
intraluminal bleeding were reduced significantly by P. amarus. The local administration of nirtetralin, phyltetralin or niranthin
Biochemical analysis indicated that reduced GSH of gastric mucosa (30 nmol/paw), similar to WEB2170 (a PAF receptor antagonist,
produced by ethanol administration was significantly elevated by 30 nmol/paw), significantly inhibited PAF-induced paw oedema
treatment with P. amarus (Raphael and Khuttan, 2003). Graded formation in mice. The extracts of P. amarus (100 ␮g/mL) and niran-
doses of the aqueous extract of P. amarus (100–800 mg/kg) admin- thin (30 ␮M), but not nirtetralin or phyltetralin (30 ␮M), decreased
istered orally produced a dose related inhibition of gut meal travel the specific binding of [3H]-PAF in mouse cerebral cortex mem-
distance in normal mice. The highest intestinal transit inhibition branes. The mean IC50 values from these effects were 6.5 and
of 31.65% was obtained with 400 mg/kg. In castor oil induced 0.3 ␮M, respectively. Both niranthin and WEB2170 (30 nmol/paw)
diarrhoea in mice, P. amarus extract (400 mg/kg) delayed the onset inhibited the increase of myeloperoxidase activity induced by
of diarrhoea, reduced frequency of defecation and reduced gut PAF injection in the mouse paw (Kassuya et al., 2006). The hex-
meal travel distance significantly resulting in intestinal transit ane extract (HE), the lignan-rich fraction (LRF), or the lignans
inhibition of 79.94% compared to 86.92% produced by morphine phyltetralin, nirtetralin, niranthin, but not hypophyllanthin or
(100 mg/kg). In addition, the activities of some intestinal mucosa phyllanthin from P. amarus inhibited carrageenan-induced paw
enzymes (maltase, sucrase, lactase and alkaline phosphatase) oedema and neutrophil influx. The HE, the LRF or nirtetralin also
in mice pretreated with extract before castor oil were not as inhibited the increase of IL1-␤ tissue levels induced by Cg. Fur-
severely depressed as those in castor oil treated mice (control) thermore, bradykinin (BK)-, platelet activating factor (PAF) – and
(Odetola and Akojenu, 2000). endothelin-1 (ET-1)-induced paw oedema were significantly inhib-
ited by the HE or LRF while histamine- and substance P-induced
8.5. Antifungal activity paw oedema were unaffected. Finally, nirtetralin or phyltetralin
caused inhibition of paw oedema induced by PAF or ET-1 (Kassuya
The effect of nor-securinine, an alkaloid isolated from P. amarus et al., 2005). P. amarus EtOH, H2 O (0.25%) and hexane extracts
was studied against spore germination of some fungi (Alternaria (0.01%) showed an inhibition of LPS-induced production of NO and
brassicae, Alternaria solani, Curvularia pennisetti, Curvularia sp., PGE2 in KC and in RAW264.7. The extracts also attenuated the LPS-
Erysiphe pisi, Helminthosporium frumentacei) as well as pea pow- induced secretion of TNF-␣ in RAW264.7 as well as in human blood.
dery mildew (E. pisi) under glasshouse conditions. The sensitivity Both extracts reduced expression of iNOS and COX-2 and inhib-
of fungi to nor-securinine varied considerably. Nor-securinine was ited activation of NF-kappa ␤, but not of AP-1. P. amarus inhibited
effective against most of the fungi. H. frumentacei was more sen- induction of interleukin (IL)-1␤, IL-10, and interferon-␥ in human
sitive even at the lowest concentration (1000 ␮g/mL). Conidia of blood and reduced TNF-␣ production in vivo (Kiemer et al., 2003).
E. pisi were also inhibited in partially or completely appresso- The anti-allodynic and anti-oedematogenic effects of the hexane
rium formation. Pre-inoculation treatment showed greater efficacy extract, lignan-rich fraction and purified lignans from aerial parts
than post-inoculation in inhibiting powdery mildew development of P. amarus was studied in the inflammatory and neuropathic
on pea plants in a glasshouse. Maximum inhibition occurred at models of nociception. The hexane extract at a dose of 100 mg/kg
2000 ␮g/mL (Sahni et al., 2005). Antifungal bioassay in terms of body weight orally in male swiss albino rats inhibited the allodynia
reduction in weight, colony diameter and sporulation of the target and the oedema induced by the intraplantar injection of complete
fungal colony was carried out using Broth Dilution method. Root Freund’s adjuvant (CFA). The inhibition observed was 76 ± 7% (ipsi-
part of the P. amarus, extracted in various organic solvents have lateral paw), 64 ± 7% (contralateral paw), and 41 ± 2% (oedema).
not shown any noticeable antifungal activity. The percentage inhi- Otherwise, the lignan-rich fraction 100 mg/kg body p.o., or the pure
bition observed in different solvent extracts of aerial part was found lignans 30–100 mg/kg body weight i.p. did not affect CFA-induced
as reduction in weight. It was concluded that Chloroform fraction allodynia. Administered chronically, the lignan fraction reduced
of the aerial part of the plant P. amarus have showed significant CFA-induced paw oedema (39 ± 9%). When evaluated in the model
inhibitory effect against dermatophytic fungi M. gypseum (Agrawal of neuropathic pain caused by partial ligation of sciatic nerve, the
et al., 2004). hexane extract inhibited the mechanical allodynia (77 ± 7%), with a
similar efficacy to the gabapentin (71 ± 10%) (Kassuya et al., 2003).
8.6. Analgesic, anti-inflammatory, anti-allodynic and The aqueous extracts of leaves and stems of P. amarus at the dose of
anti-oedematogenic activity 500, 250 and 100 mg/kg body weight orally as a single dose, 1 h prior
to experiment was administered to female stains of Balb/c mice.
The aqueous leaves extract of P. amarus was investigated for Extracts produced an inhibition of 26, 33 and 39% respectively at
analgesic and anti-inflammatory activities using both thermal and 3 h while methanolic extract of P. amarus at a dose level of 100, 250
chemical models of pain assessment in rats. The extract caused and 500 mg/kg body weight produced an inhibition of 29, 37 and
a significant (p < 0.05) dose related increased inhibition of the 42% respectively at 3 h and significant inhibition of paw oedema
carrageenan-induced paw oedema in the rats. The inhibition pro- was observed throughout the course of experiment up to 8 h
duced by 200 mg/kg aqueous extract of P. amarus (70.20%) was (Raphael and Khuttan, 2003).
significantly higher than that of the reference drug (acetylsali-
cylic acid). The extract produced a marked analgesic activity by 8.7. Antinociceptic activity
inhibiting both early and late phases of pain stimulus in formalin-
induced paw licking rats and also a significant and dose related The hydroalcoholic extract of four new species of Phyllanthus,
increase in inhibition of the mean tail immersion duration at vary- given intraperitoneally, produced significant inhibition of acetic
ing water bath temperature (50, 55 and 60 ◦ C) (Iranloye et al., acid-induced abdominal constrictions. The hydroalcoholic extract
2011). The effects of 75% methanolic extract of whole plant of P. of the Phyllanthus species elicited significant inhibition of the
Author's personal copy

302 J.R. Patel et al. / Journal of Ethnopharmacology 138 (2011) 286–313

capsaicin-induced neurogenic pain. The extract of these Phyllan- Acetone extract produced a dose-dependent increase whereas
thus species when given intraperitoneally, produced dose related 500 mg/kg of the aqueous extract seems more effective (Shokunbi
and pronounced antinociception when assessed against chem- and Odetola, 2008). Different drying treatments led to significant
ical models of nociception, including acetic acid, formalin and reduction (p < 0.05) in antioxidant properties of P. amarus methano-
capsaicin-induced pain. Orally, these species were less potent and lic extracts, with microwave drying causing the highest decrease
efficacious than given by intraperitoneally (Santos et al., 2000). in total phenolic compound and antioxidant activity exhibited by
the reduction in both radical scavenging activity and FRAP. On
8.8. Antioxidant activity the other hand, boiling water extracts appeared to exhibit sig-
nificantly stronger antioxidant potentials (p < 0.05) even in dried
Antioxidant activity of aqueous extract of whole plant of P. plant materials due to greater solubility of compounds, breakdown
amarus at the dose of 200 mg/kg body weight/day was evalu- of cellular constituents as well as hydrolysis of tannins (Lim and
ated in streptozotocin (STZ)-induced diabetic male Wistar albino Murtijaya, 2007). The antioxidant activity of methanolic extracts of
rats. Antioxidant enzymes; GR, GPX and GST, CAT and SOD were five species including P. debilis, P. urinaria, P. virgatus, P. maderas-
also assayed. Diabetic treated animal group showed a signifi- patensis, P. amarus from the genus Phyllanthus was evaluated by
cant decrease in renal LPO, protein oxidation and a significant various antioxidant assays. All the extracts at the concentration of
increase in GSH content and GR, GPX and GST activities when com- 50 ␮g/mL showed strong antioxidant activity in all the tested meth-
pared with STZ-induced diabetic rats. The activities of SOD and ods. Among the five plants, P. debilis has been found to possess the
CAT decreased significantly in STZ-induced diabetic rats, but were highest activity and P. amarus posses the lowest activity in all tested
normalized in diabetic treated group (Karuna et al., 2011). The models (Kumaran and Karunakaran, 2007). Methanolic extract of
aqueous extract of whole plant of P. amarus showed significant leaves and stems of P. amarus was found to have potential antiox-
(p < 0.05) potential in scavenging free radicals, and in inhibiting idant activity as it could inhibit lipid peroxidation, and scavenge
lipid peroxidation. Furthermore, the extract proved to contain a hydroxyl and superoxide radicals in vitro. The amount required for
high content of phenolic compounds which were found to have 50% inhibition of lipid peroxide formation was 104 ␮g/mL and the
strong and significant (p < 0.05) positive correlations to free-radical concentrations needed to scavenge hydroxyl and superoxide rad-
scavenging potential, lipid peroxidation inhibition capacity and icals were 117 and 19 ␮g/mL respectively (Raphael et al., 2002a).
cyto-protective efficiency against Cr(VI)-induced oxidative cellu- Amariin, repandusinic acid, phyllanthusiin D, phyllanthin and phe-
lar damage (Guha et al., 2010). Aqueous extract of P. amarus at nolic compouds isolated from P. amarus showed remarkable high
a dose of 200 mg/kg body weight/day for 8 weeks treated male antioxidant activity.
albino Wistar rats showed a significant decrease in plasma LPO
and a significant increase in plasma vitamin C, uric acid, GSH 8.9. Antiplasmodial activity
level, GPX, CAT and SOD activities. Single cell gel electrophore-
sis (SCGE) experiment revealed that aqueous extract of P. amarus The aqueous and ethanolic extracts of the whole plant of
was devoid of genotoxicity and had a significant protective effect P. amarus were administered to Swiss albino mice at doses of
against H2 O2 , STZ and nitric oxide (NO) induced lymphocyte DNA 200, 400, 800 and 1600 mg/kg/day to investigate the prophylac-
damage (Karuna et al., 2009). Free-radical scavenging activity of tic and chemotherapeutic effect of the extract against Plasmodium
50% ethanolic extract of aerial parts of P. amarus extract and yoelii infection and compared with those of standard antimalarial
phyllanthin was examined using 2,2-diphenyl-2-picrylhydrazyl drugs pyrimethamine and chloroquine, artesunate/amodiaquine
(DPPH) assay. The DPPH free-radical scavenging activity was respectively. Results showed that extracts demonstrated a dose-
concentration-dependent in both cases and reaches a maximum at dependent prophylactic and chemotherapeutic activity. The
a concentration of 300 ␮g/mL for P. amarus extract and 20 ␮mol/mL aqueous extract showed slightly higher effect than the ethanolic
for phyllanthin. No difference in inhibition was noted with fur- extract. The antiplasmodial effects of extracts were comparable
ther increase in concentration of either of the compounds. Results to the standard prophylactic and chemotherapeutic drugs used in
indicated that phyllanthin exhibited very high antioxidative prop- chloroquine resistant plasmodium infection. The extracts showed
erty as compared to P. amarus extract which is clearly evident prophylactic effect by significant delay in the onset of infection with
by its low IC50 value of 7.4 ␮mol/mL (Krithika et al., 2009). The the suppression of 79% at a dose of 1600 mg/kg/day. The results
antioxidant activity of some of its principal constituents, namely indicate that the extracts of the whole plant of P. amarus possess
amariin, 1-galloyl-2,3-dehydrohexahydroxydiphenyl (DHHDP)- repository and chemotherapeutic effects against resistant strains
glucose, repandusinic acid, geraniin, corilagin, phyllanthusiin of P. yoelii in Swiss albino mice (Ajala et al., 2011). ‘Saye’, a tra-
D, rutin and quercetin 3-O-glucoside were examined for ditional medicine used in Burkina Faso, which consists of extracts
their ability to scavenge free radicals in a range of systems of Cochlospermum planchonii (rhizome), Cassia alata (leaves) and
including DPPH, 2,2-azobis-3-ethylbenzthiazoline-6-sulfonic acid P. amarus (whole plant). Mice treated with 50, 100, 150, 200,
(ABTS)/ferrylmyoglobin, ferric reducing antioxidant power (FRAP) 250 mg/kg body weight for 0–3 days, orally showed a significant
and pulse radiolysis. The compounds showed significant antiox- effect against P. falciparum and Plasmodium berghei parasites grown
idant activities with differing efficacy depending on the assays in vivo (IC50 = 80.11 ± 3.40 ␮g/mL; ED50 = 112.78 ± 32.32 mg/kg). In
employed. Amariin, repandusinic acid and phyllanthusiin D vitro the activity was lower at the concentration of 100, 50, 25 and
showed higher antioxidant activity among the ellagitannins and 12.5 ␮g/mL and compared with chloroquine (Traore et al., 2008).
were comparable to the flavonoids, rutin and quercetin 3-O- The aqueous extract of leaves and stem of P. amarus at doses of
glucoside (Londhe et al., 2008). Pretreatment with P. amarus leaves 108.33, 165 and 325 mg/kg in Swiss albino mice was found to cause
extracts on antioxidant enzymes in gastric mucosa homogenate a significant dose-dependent suppression of P. berghei parasites
was studied. Significant reductions (p < 0.05) in the gastric mucosa [p < 0.05] sulfadoxine/pyrimethamine caused a similar significant
catalase (CAT), super oxide dismutase (SOD) and glutathione-s- suppression of P. berghei parasites (Dapper et al., 2007). Ethano-
transferase (GST) activities were observed in the ethanol group lic, methanolic and methylene chloride extracts of entire plant
compared with the normal control group. P. amarus acetone of P. amarus showed significant activity against the chloroquine-
extracts (1000 mg/kg) and cimetidine (100 mg/kg) caused an ele- sensitive strain of P. falciparum 3D7. The IC50 of methanolic extract
vation by 53 and 52% for CAT, 8 and 14% for SOD and 33 and and methylene chloride extract was 5 and 14.53 ␮g/mL respectively
38% for GST respectively when compared with the ethanol group. (Adjobimey et al., 2004).
Author's personal copy

J.R. Patel et al. / Journal of Ethnopharmacology 138 (2011) 286–313 303

8.10. Antiviral activity were most active (0.24 ␮g/mL). HIV-1 replication was also blocked
in CD4+ lymphoid cells with comparable EC50 values. Applying
Inhibitory effect of methanolic extracts of root and leaves of P. a cell-based internalization assay, it was demonstrated 70–75%
amarus against the NS3 and NS5B enzymes of hepatitis-C virus were inhibition of virus uptake at concentrations of 2.5 ␮g/mL for the
screened by in vitro enzyme assays. Effect on viral RNA replication water–alcohol extract and geraniin. In addition, a concentration-
was investigated by using TaqMan Real time RT-PCR. P. amarus root dependent inhibition of HIV-1 reverse transcriptase (RT) was
extract showed significant inhibition of HCV-NS3 protease enzyme; demonstrated in vitro. The 50% inhibitory concentration (IC50 ) val-
whereas P. amarus leaves extract showed considerable inhibition of ues varied from 1.8 to 14.6 ␮g/mL (Notka et al., 2003). Study on
NS5B in the in vitro assays. Further, the P. amarus root and leaves 25 compounds isolated from P. amarus, P. multiflorus, P. tenel-
extracts significantly inhibited replication of HCV monocistronic lus and P. virgatus found that niranthin, nirtetralin, hinokinin and
replicon RNA and HCV H77S viral RNA in HCV cell culture system. geraniin at the non-cytotoxic concentration of 50 ␮m, suppressed
However, both P. amarus root and P. amarus leaves extracts did not effectively both HBsAg and hepatitis B effective antigen (HbeAg)
show cytotoxicity in HuH-7 cells in the MTT assay. Furthermore, expression, of these, niranthin showed the best anti-HBsAg activ-
P. amarus root extract with IFN-␣ showed additive effect in the ity, while the most potent anti-HBeAg activity was observed with
inhibition of HCV RNA replication. Results suggested the possible hinokinin (Huang et al., 2003). Analysis in HuH-7 cells (human hep-
molecular basis of the inhibitory activity of P. amarus extract against ato cellular carcinoma cell line) with transfected plasmids using a
HCV which would help in optimization and subsequent develop- luciferase reporter showed that DMSO solution of whole plant of
ment of specific antiviral agent using P. amarus as potent natural P. amarus at the concentration of 50, 100 or 200 ␮g/mL specifically
source (Ravikumar et al., 2011). The aqueous extract of P. amarus inhibited HBV enhancer I activity. To identify the mechanism of
showed partial antiviral activity against white spot syndrome virus this HBV enhancer I inhibition, liver-enriched cellular transcrip-
in shrimp at the concentration of 150 mg/kg of animal body weight tion factors were co-expressed in HuH-7 cells. The C/EBP ␣ and
for 30 days (Balasubramanian et al., 2007). The aqueous, butanol, ␤ as well as HNF-3␣ and ␤ (hepatocyte nuclear factor), transcrip-
and alcoholic extracts of P. amarus and P. maderaspatensis, was stud- tion factor significantly upregulated the HBV enhancer I activity.
ied for antiviral properties on duck hepatitis B virus infection. One In contrast, co-transfection of HNF-I ␣ or ␤ had no effect upon
hundred and fourteen ducks infected posthatch with the duck hep- the HBV enhancer I activity. Exposure to P. amarus inhibited C/EBP
atitis B virus (DHBV) were divided into groups at 3 months of age ␣ and ␤-mediated up-regulation of HBV enhancer I activity in a
and treated intraperitoneally with the aqueous, butanol, and alco- dose-dependent manner, whereas HNF-3␣- and ␤-mediated up-
holic extracts of these two plants at doses of 25, 50, or 200 mg/kg regulation of HBV enhancer I was unaffected. In vitro gel shifts
body weight. There was no definite antiviral property observed in showed that P. amarus inhibited complexing of C/EBP transcrip-
the treated ducks (Munshi et al., 1993a). The therapeutic potential tion factors to a consensus oligonucleotide sequence, whereas DNA
of plant extracts of P. amarus and P. maderaspatensis for postexpo- binding of AP-1 and SP-1 transcription factors was unaffected
sure prophylaxis against infection by Hepadnaviruses was studied (Ott et al., 1997). P. amarus inhibited hepatitis B virus polymerase
in ducklings infected by the DHBV. The observations suggested that activity, decreased episomal hepatitis B virus DNA content and sup-
aqueous extract of P. amarus (100 mg/kg body weight) and ethano- pressed virus release into culture medium. When DSMO solution of
lic extract of P. maderaspatensis (100 mg/kg body weight) are not whole plant of P. amarus 80 mg/kg body weight i.p. daily for 7 days
useful as therapeutic agents for postexposure prophylaxis against was administered to transgenic mice, hepatic HBsAg mRNA level
DHBV infection (Munshi et al., 1993b). The polyphenol containing was decreased, indicating transcriptional or post-transcriptional
extract of P. amarus and representatives of simple phenols (shikimic down-regulation of the transgene. Increase in hepatitis B virus
acid 3-and 5-O-gallate), flavan-3-ols (epigallocatechin 3-gallate), mRNA expression after stimulation of the glucocorticoid responsive
proanthocyanidins (a hexamer) and hydrolysable tannins (corila- element was also suppressed by P. amarus, suggesting involvement
gin, casuariin, geraniin) were studied in vitro for gene expressions of the hepatitis B virus enhancer in this response. Disruption by P.
(iNOS, IL-1, IL-10, IL-12, IL-18, TNF-␣, IFN-␣ and ␤) by RT-PCR. All amarus of hepatitis B virus polymerase activity, mRNA transcrip-
extracts and compounds at the concentration of 50 ␮g/mL were tion and replication supports its role as an antiviral agent (Lee et al.,
capable of enhancing the iNOS and cytokine mRNA levels in para- 1996). The effect of an aqueous extract of whole plant of P. amarus
sitised cells when compared with those in non-infected conditions at the concentration of 1 mg/mL on the cultured hepatoma cell
(Kolodziej et al., 2005). In vitro culture of hairy roots of P. amarus line HepA2 has been reported. The cell line had been transfected
induced by Agrobacterium rhizogenes was shown to possess 85% with tandemly arranged HBV DNA and continued to synthesize
inhibition (in contrast to 15% in the control) in binding of Hepatitis and secrete both HBsAg and HBeAg. Extract of P. amarus reversibly
B Surface Antigen (HBsAg) to its antibody (anti-HBs) after 24 h of inhibited cellular proliferation and suppressed HBsAg production
incubation with HbsAg-positive sera in vitro at 37 ◦ C (Bhattacharyya but not HBeAg production in HepA2 cells. P. amarus also suppressed
and Bhattacharya, 2004). Water–alcohol extract of leaves of P. HBsAg gene expression at mRNA level in a time-dependent manner,
amarus in vitro blocks HIV-1 attachment and the HIV-1 enzymes and selectively abolished the HBsAg gene promoter driven chlo-
integrase, reverse transcriptase and protease to different degrees. ramphenicol acetyltransferase activity (Yeh et al., 1993). Ethanolic
A gallotannin containing fraction and the isolated ellagitannins extract and subsequent fractions (hexane, chloroform, butanol and
geraniin and corilagin were shown to be the most potent mediators water) were tested for in vitro effects on HBsAg, HBeAg and HBV
of these antiviral activities. P. amarus derived preparations blocked DNA in serum samples positive for HBV antigens followed by the
the interaction of HIV-1 gp120 with its primary cellular recep- screening of respective antigens by ELISA. HBV DNA was deter-
tor CD4 at 50% inhibitory concentrations of 2.65 (water–alcohol mined by molecular hybridization. The extracts were effective
extract) to 0.48 ␮g/mL (geraniin). Inhibition was also evident for against HBV antigens, the butanol extract being the most potent.
the HIV-1 enzymes integrase (0.48–0.16 ␮g/mL), reverse transcrip- The chromatographic fractions showed an enhanced activity. The
tase (8.17–2.53 ␮g/mL) and protease (21.80–6.28 ␮g/mL) (Notka active fractions inhibited the interaction between HBsAg/HBeAg
et al., 2004). Aqueous as well as alcohol-based P. amarus extracts and their corresponding antibodies suggesting anti-HBs, anti-Hbe
potently inhibited HIV-1 replication in HeLa CD4+ cells with like activity and also an effect on HBV DNA (Mehrotra et al., 1991).
50% effective concentration (EC50 ) values ranging from 0.9 to Extracts of P. amarus have been shown to inhibit the DNA poly-
7.6 ␮g/mL. A gallotannin enriched fraction showed enhanced activ- merase of HBV and Woodchuck hepatitis virus (WHV) in vitro.
ity (0.4 ␮g/mL), and the purified gallotannins geraniin and corilagin Woodchuck carriers of WHV were treated intraperitoneally with
Author's personal copy

304 J.R. Patel et al. / Journal of Ethnopharmacology 138 (2011) 286–313

P. amarus extract. When three out of four recently infected WHV like IFN-␣, lamivudine, adefovir dipivoxil, thymosin, vidarabine, or
carriers were treated with aqueous extract of P. amarus at a dose conventional treatment with the same antiviral drug alone. Phyl-
of 0.9 mL (9 mg dry weight), i.p., twice weekly, they lost the WHV. lanthus significantly affected serum HBV DNA (RR 0.69; 95% CI
Animals infected for greater than or equal to 3 months showed 0.52–0.91, p = 0.008; I(2) = 71%), serum HBeAg (RR 0.70; 95% CI
a decrease in virus levels. Preliminary results in human carriers 0.60–0.81, p < 0.00001; I(2) = 68%), and HBeAg seroconversion (RR
treated orally with P. amarus for 1 month indicated that approxi- 0.77; 95% CI 0.63–0.92, p = 0.005; I(2) = 78%), but the heterogene-
mately 60% of the carriers lost HBV during the observation period ity was substantial. The result obtained regarding serum HBV DNA
(Blumberg et al., 1989, 1990). The aqueous, butanolic and alcoholic was not supported by trial sequential analysis. None of the trials
extract of P. amarus were described for the treatment of chronic reported mortality and hepatitis B related morbidity, quality of
hepatitis B virus infection on duck hepatitis B virus at the doses life, or liver histology. Only two trials reported adverse events with
of 25, 50, and 200 mg/kg body weight. Nine ducks congenitally numbers without significant differences. No serious adverse events
infected with the DHBV were treated either orally (four ducks for were reported (Xia et al., 2011). Volunteers received either 1200 mg
10 weeks) or intraperitoneally (five ducks for 12 weeks) with P. of water extract from leaves of P. amarus or 450 mg lamivudine,
amarus, compared to placebo-treated control ducks. These treat- and blood was collected before, and 1, 2 and 3 h after treatment.
ments did not result in a reduction of circulating viral DNA in the MAGI cells were inoculated with HIV-1 in the presence of pre-
serum or in the level of viral DNA replication in the liver. In two and post-treatment serum (1, 2 and 3 h post-administration, as
of the five intraperitoneal-treated ducks, a reduction in the level of indicated). Sera at a final concentration of 5% reduced HIV repli-
duck hepatitis B surface antigenaemia (DHBsAg) was observed. The cation by more than 30%. These results supported that P. amarus
data strongly suggested that P. amarus has no significant inhibitory has inhibitory effects on HIV in vitro and in vivo (Notka et al.,
effect on DHBV DNA replication and only a minor effect on DHB- 2004). Fifty-five patients with chronic viral hepatitis B were ran-
sAg P. amarus production (Niu et al., 1990). Alexander cell line, a domly divided into two groups. Thirty patients were treated with
human hepatocellular carcinoma derived cell line which has the PA compound capsule (Chief ingredients: P. amarus, Radix noto-
property of secreting HBsAg in the supernatant was used to study ginseng, etc. each capsule contains P. amarus 275 mg), orally, three
the antiviral property of P. amarus. Aquous extract of P. amarus times daily, four capsule each time for 3 months in the treatment
was evaluated for its in vitro ability to inhibit HBsAg secretion on group; another 25 patients were treated with domestic recombi-
a dose-dependent manner. Aquous extract of P. amarus at the con- nant human interferon alpha-1b (IFN-␣1␤) for 3 months as control.
centration of 1 mg/mL as a single dose inhibited the secretion of The total effective rate in the treatment group was 83.3%, showed
HBsAg for a period of 48 h. Observations revealed the anti hepatitis no significant difference from the control (p > 0.05). The normaliza-
B virus property of P. amarus at cellular level and further confirmed tion rates of ALT, A/G and SB in the treatment group were 73.3, 80.0
its beneficial use in the treatment of acute and chronic hepatitis B and 78.2% respectively, which were significantly higher than that
and healthy carriers of HBV (Jayaram and Thyagarajan, 1996). Lig- in the control (p < 0.05). The negative conversion rates of HbeAg
nans geraniin, corilagin, niranthin and hinokinin isolated from P. and HBV DNA in the treatment group were 42.3 and 47.8%, showed
amarus exhibited excellent antiviral activity. no significant difference from the control (p > 0.005) (Wang et al.,
2001). The efficacy and safety of genus Phyllanthus for chronic hep-
8.11. Clinical studies atitis B virus (HBV) infection was evaluated by a systematic review
of randomized clinical trials. Randomized trials comparing genus
Study focuses on effect of P. amarus therapy for protection of Phyllanthus vs. placebo, no intervention, general nonspecific treat-
liver in hepatitis-C through investigating liver profile enzymes; ment, other herbal medicine, or interferon treatment for chronic
antioxidant enzymes, antioxidant vitamins and lipid peroxidation. HBV infection were identified by electronic and manual searches.
The study consists of 50 clinical diagnosed hepatitis-C patients Trials of Phyllanthus herb plus interferon (IFN) vs. IFN alone were
ranging in between age group 25 and 60 years. The control group also included. No blinding and language limitations were applied.
includes 50 ages and sex matched normal healthy persons. Oxida- The methodological quality of trials was assessed by the Jadad
tive stress was assessed by estimating LPO. The parameters like scale plus allocation concealment. Twenty-two randomized trials
serum bilirubin, total proteins and activity of liver profile enzymes (n = 1947) were identified. The methodological quality was high in
were done. Activity of enzymatic antioxidants; SOD, GPX and lev- five double-blind trials and low in the 17 remaining trials. The com-
els of non-enzymatic antioxidant vitamins E and C was measured bined results showed that Phyllanthus species had positive effect
in plasma or erythrocytes. Methods used in the study are mainly on clearance of serum HBsAg (relative risk 5.64, 95% CI 1.85–17.21)
enzyme kinetics by autoanalyzer and by turbidimetry. Plasma LPO compared with placebo or no intervention. There was no signifi-
levels were significantly high but activity of SOD, GPX, catalase and cant difference on clearance of serum HBsAg, HBeAg and HBV DNA
levels of vitamins E and C were significantly lowered in hepatitis-C between Phyllanthus and IFN. Phyllanthus species were better than
in comparison with controls. After P. amarus therapy for 5 and 10 nonspecific treatment or other herbal medicines for the clearance of
weeks plasma LPO levels were significantly decreased and activ- serum HbsAg, HBeAg, HBV DNA, and liver enzyme normalization.
ity of SOD, GPX, catalase and vitamins E and C were significantly Analysis showed a better effect of the Phyllanthus plus IFN com-
increased in hepatitis-C. It was concluded that hepatitis-C increased bination on clearance of serum HBeAg (1.56, 1.06–2.32) and HBV
oxidative stress and might be playing an important role in hepatic DNA (1.52, 1.05–2.21) than IFN alone. The efficacy and safety of
cell damage and pathogenesis of hepatitis-C. It was suggested that genus Phyllanthus for chronic hepatitis B virus (HBV) infection has
the therapy with P. amarus increased antioxidants and reduced lipid been systematically reviewed the clinical trials (Liu et al., 2001). The
peroxidation of hepatic cellular and intracellular membranes and powder of the plant P. amarus was given in capsule form (300 mg
protected liver damage due to free radicals in hepatitis-C (Nikam capsules-3 capsules thrice daily) and an antacid powder in similar
et al., 2011). A total of 16 randomized trials with 1326 patients capsule was used as placebo. Fifty-seven patients were random-
were included. One trial with 42 participants compared phyllan- ized to receive either the placebo (28 cases) or the drug (28 cases).
thus with placebo. The trial found no significant difference in HBeAg The two groups were comparable at the time of entry. Two cases
seroconversion after the end of treatment [risk ratio (RR) 0.9; 95% from the placebo and one from the placebo and one from the drug
(confidence intervals) CI 0.73–1.25] or follow-up (RR 1.00; 95% group dropped out of the study. The duration of disease (time taken
CI 0.63–1.60). No other outcomes could be assessed. Fifteen tri- for bilirubin to come to below 2 mg %) was taken as the outcome
als were compared with phyllanthus plus and an antiviral drug measure. The duration of disease in the two groups was compared
Author's personal copy

J.R. Patel et al. / Journal of Ethnopharmacology 138 (2011) 286–313 305

by Cox’s proportional hazards analysis after adjusting for the vari- P. amarus (200 mg doses in gelatin capsule presterilized with ethy-
ables that influence the duration of jaundice. Only initial serum lene oxide, three times daily) for 30 days. 22 of 37 (59%) treated
bilirubin was an independent predictor of duration of jaundice. patients had lost hepatitis B surface antigen when tested 15–20
The analysis showed that P. amarus powder did not significantly days after the end of the treatment compared with only 1 of 23
reduce the duration of jaundice in persons with virus B hepati- (4%) placebo-treated controls. Some subjects have been followed
tis (Narendranathan et al., 1999). The efficacy of P. amarus to treat for up to 9 months. In no case has the surface antigen returned.
acute viral hepatitis (AVH) was evaluated in parallel to another drug Clinical observation revealed few side effects which include fatigue,
Essentiale (an essential phospholipid extracted from soybean oil) malaise, fever, chills, urticaria, anorexia, nausea, abdominal pain,
and compared with a group of patients who were treated symp- diarrhoea, headache, dizziness, disturbance of sleep and skin rash
tomatically with vitamins as the controls. Serological profile of 93 (Thyagarajan et al., 1988).
sporadic AVH cases of the study showed that 25.8% had an acute
infection due to hepatitis A virus (HAV), 52.6% suffered due to hep- 8.12. Aphrodisiac activity
atitis B viral (HBV) infection, while 19.3% of cases were classified
as non-A non-B hepatitis (NANB) by exclusion. On follow up of the The effect of methanolic extract of the leaves of P. amarus on the
patients at the end of treatment period of 4 weeks with respec- hormonal parameters of male guinea pigs was investigated. The
tive drug regimen, it was seen that both P. amarus and Essentiale hormonal parameters investigated were testosterone, leutinizing
brought about significant biochemical and clinical normalcy among and follicle stimulating hormone. Methanolic extract of P. amarus
the HAV infected patients compared to control group (p < 0.001). In leaves (50–800 mg/kg) caused a statistically significant increase in
acute HBV group, P. amarus treated patients recovered faster than the level of testosterone of the male guinea pigs, from 2.3 ± 0.06 to
the essentiale treated group and the controls (p < 0.001). Essentiale 3.9 ± 0.05, 4.3 ± 0.6 and 2.8 ± 0.6 after the 7th, 14th and 21st day of
was found to help the non-A non-B hepatitis patients to resume the administration of the extracts, respectively. Furthermore, the
earlier biochemical normalcy than by P. amarus and control treat- methanolic extract of P. amarus (800 mg/kg) caused an insignifi-
ments. P. amarus seemed to accelerate the clearance of HBsAg in cant change in the level of leutenizing (LH) and follicle stimulating
86.9% of convalescing AVH-B cases in 3 months time as against (FSH) hormones from 3.1 ± 0.22 and 1.6 ± 0.50 to 3.0 ± 0.08 and
48.0% in the Essentiale treated group and 50.0% in the controls 1.5 ± 0.13, respectively (Obianime and Uche, 2009).
(Jayaram et al., 1997). The role of P. amarus in eradication of the
virus in hepatitis B carriers was evaluated by administering it to
8.13. Contraceptive effect
30 asymptomatic carriers of HBsAg in a dosage of 250–500 mg
thrice daily for 4–8 weeks. It was found that none of the 30 sub-
Antifertility effect of an alcoholic extract of the whole plant of P.
jects cleared HBsAg. P. amarus was well tolerated, with no clinical
amarus at a dose of 100 mg/kg body weight for 30 days orally was
side effects or changes in the organ profiles for safety evaluation.
investigated in cyclic adult female mice. The results revealed no
It was found that P. amarus was not effective in clearing HBsAg
significant change in absolute body and organ weights in extract
in asymptomatic carriers of the antigen (Doshi et al., 1994). Sixty-
fed animals indicated no alteration in general metabolic status.
five adult asymptomatic chronic carriers of hepatitis B virus were
Cohabited females with normal male mice were unable to become
enrolled to the randomized controlled efficacy study of P. amarus.
pregnant as their cyclicity was affected. These factors are related
Thirty-four received P. amarus 600 mg per day for 30 days and
to a change in the hormonal milieu that governs female reproduc-
31 received placebo in identical capsules. The conversion rate of
tive function. Upon withdrawal of feeding for 45 days, these effects
HBsAg was 6% in the study group at day 30. When 20 subjects in
were reversible. Thus, this extract manifests a definite contracep-
the P. amarus group were given a further 30-day treatment and 22
tive effect in female mice (Rao and Alice, 2001).
placebo recipients given P. amarus 1200 mg per day for 30 days,
the conversion was observed in 1 (5%) in the higher dose group.
The results indicated that P. amarus, whole plant except root, given 8.14. Diuretic and antihypertentive activity
at the studied dosage and duration, had a very minimal effect on
eradication of HBsAg asymptomatic chronic carriers (Thamlikitkul 8.14.1. Clinical study
et al., 1991). A total of 79 human carriers of HBV, out of these, 40 Diuretic, hypotensive and hypoglycaemic effects of P. amarus
were given 200 mg of dried, powdered whole plant of P. amarus, on human subjects were assessed. Nine mild hypertensives (four
three times daily. The remaining 39 carriers were given lactose of them also suffering from diabetes mellitus) were treated with
placebo at the same dosage and frequency. Carriers were assigned a preparation of the whole plant of P. amarus for 10 days. Suit-
randomly to the treatment or control groups and neither the car- able parameters were studied in the blood and urine samples of
riers nor their physicians were told of the assignment. The drug the subjects along with physiological profile and dietary pattern
or placebo was administered for 30 days then stopped; the carri- before and after the treatment period. Significant increase in 24 h
ers were then tested monthly for up to 9 months after cessation of urine volume, urine and serum Na levels was observed. A significant
treatment or placebo. Thirty-seven of the treated carriers and 23 reduction in systolic blood pressure in non-diabetic hypertensives
of the controls returned for follow-up. Of the treated subjects 59%, and female subjects was noted. Blood glucose level was also signif-
but only 4% of 23 control subjects, became HBsAg-negative after icantly reduced in the treated group. Clinical observations revealed
30 days and remained so until the end of the follow-up period. no harmful side effects (Srividya and Periwal, 1995).
Thirteen out of 14 carriers with HBsAg but without HBeAg became
negative, while only five out of 17 HBsAg positive, HBeAg-positive 8.15. Hepatoprotective activity
subjects lost the surface antigen. HBsAg did not reappear in any
of these subjects. The individuals who remained HBsAg-positive The effect of aqueous leaves extract of P. amarus on matrix met-
are now being treated for longer periods to determine whether alloproteinases and tissue inhibitors of matrix metalloproteinases
active replication of virus requires longer duration of therapy. There was evaluated in alcohol and thermally oxidized polyunsaturated
was no evidence of toxicity in the treated individuals, but these fatty acid-induced hepatic fibrosis. The matrix metalloproteinase
observations were based primarily on clinical examination expression was found to be significantly decreased and the levels
(Blumberg et al., 1990). In a preliminary study, carriers of hep- of tissue inhibitors of matrix metalloproteinases and the colla-
atitis B virus were treated with a preparation of the plant gen were significantly increased in alcohol and thermally oxidized
Author's personal copy

306 J.R. Patel et al. / Journal of Ethnopharmacology 138 (2011) 286–313

polyunsaturated fatty acid treated male Wistar albino rats. Admin- acute toxicity study, single dose of P. amarus (25, 50 and 75 mg/kg,
istration of P. amarus extract 3 mL/day for 45 days significantly p.o.) or SL (Silymarin, 5 mg/kg), 24 h before ethanol (5 g/kg, p.o.) on
decreased the levels of collagen and tissue inhibitors of matrix met- male Wistar rats lowered the ethanol-induced levels of AST and/or
alloproteinases; and positively modulated the expression of matrix ALT. The 75 mg/kg P. amarus dose gave the best result similar to
metalloproteinases. It revealed that P. amarus effectively modified SL. Histopathological observations confirmed the beneficial roles
alcohol and thermally oxidized polyunsaturated fatty acid-induced of P. amarus and SL against ethanol-induced liver injury in rats
fibrosis (Surya Narayanan et al., 2011). The protective effect of (Pramyothin et al., 2007). The hepatoprotective effect of ethano-
phyllanthin, a known principal constituent of P. amarus on ethanol- lic extract of whole plant except root of P. amarus was evaluated
induced rat liver cell injury was evaluated. Primary cultures of on aflatoxin B1 -induced liver damage in mice using different bio-
rat hepatocytes (24 h culturing) were pretreated with phyllan- chemical parameters and histopathological studies. Albino mice
thin (1, 2, 3 and 4 ␮g/mL) for 24 h. After 24 h pretreatment, cells treatment with P. amarus extract (300 mg/kg body weight for 3
were treated with ethanol (80 ␮L/mL) for 2 h. Ethanol decreased months and animals were sacrified with the interval of 30 days
%MTT, increased the release of ALT and AST with increase in the till the completion of study) was found to show hepatoprotective
production of intracellular ROS and lipid peroxidation. Phyllan- effect by lowering down the content of thiobarbituric acid reactive
thin demonstrated its role in protection by antagonizing the above substances (TBARS) and enhancing the reduced glutathione level
effect induced by ethanol. Phyllanthin also restored the antioxidant and the activities of antioxidant enzymes, GPX, GST, SOD and CAT.
capability of rat hepatocytes including level of total glutathione, Histopathological analysis of liver samples also confirmed the hep-
and activities of SOD and glutathione reductase (GR) which were atoprotective value of the ethanolic extract of P. amarus (Naaz et al.,
reduced by ethanol (Chirdchupunseree and Pramyothin, 2010). The 2007). The plasma concentrations of the liver function parameters
hepatoprotective activity of 50% ethanolic extract of aerial parts in aqeous extract of whole plant of P. amarus treated Wistar rats
of P. amarus plant (100, 200, and 300 mg/kg body weight daily (4 mL/rat/day) showed that the ALT and AST activities decreased
for 30 days) against CCl4 -induced liver damage in Swiss strain significantly in the blood of the test animals (Igwe et al., 2007). The
female albino mice was determined. Carbon tetrachloride adminis- ␤-glucuronidase inhibitory action of 50% methanolic, and aqueous
tration caused a significant increase in liver and ALT, AST, ALP and extracts as well as isolated actives constituents such as corilagin,
acid phosphatase (ACP), while total protein content significantly brevifolin carboxylic acid, phyllanthin, and hypophyllanthin from P.
decreased as compared to vehicle control. The effect was dose- amarus was demonstrated. The results revealed that 50% methano-
dependent. Oral administration of aqueous extract of P. amarus lic and water extracts were highly active and corilagin, the phenolic
caused significant mitigation of CCl4 induced changes. P. amarus principle isolated from 50% methanolic extract of P. amarus was
attenuated the toxic effects of carbon tetrachloride (CCl4 ) and found to be more potent at the concentration of 200 ␮g/mL which
caused a subsequent recovery towards normalization. Adminis- is comparable with silymarin (Joshi and Priya, 2007). Hot water
tration of P. amarus at 300 mg/kg body weight offered maximum extracts of P. amarus (0.8, 1.6 or 3.2 g/kg) were orally administered
recovery (98–100%) against CCl4 (Krithika and Verma, 2009a,b). b.i.d. for 7 days prior, 2 days after, or 7 days prior and 2 days after
The protective effect of P. amarus extract and phyllanthin was single oral dose of paracetamol (3 g/kg). The results showed that the
studied on CCl4 -induced toxicity in human hepatoma HepG2 cell extract at 1.6 and 3.2 g/kg decreased the paracetamol-induced hep-
line. The results indicated that CCl4 treatment caused a significant atotoxicity as indicated by the decrease in SGOT and SGPT, bilirubin
decrease in cell viability. It was observed that phyllanthin (4.18, and histopathological score while the ALP did not change. It was evi-
8.36 and 12.54 ␮g/mL for 24 h) effectively alleviated the changes dent that the hepatoprotective mechanism of this plant was neither
induced by CCl4 in a concentration-dependent manner with much related to inhibition on cytochrome P450, nor induction on sulfate
smaller strengths as compared to (200, 400 and 600 ␮g/mL) P. and glucuronide conjugation pathways of paracetamol, but partly
amarus water ethanol extract (Krithika et al., 2009). Silymarin and due to the antioxidant activity and the protective effect on the
standardized extract obtained from the whole plant of P. amarus decrease of hepatic reduced glutathione (Wongnawa et al., 2005).
100 mg/kg body weight against CCl4 -induced hepatotoxicity in rats Silymarin, fresh leaves juice and ethanolic extracts of P. amarus
were found effective as hepatoprotective as evidenced by plasma (50 mg/kg, 1 mL/kg and 300 mg/kg body weight respectively) pre-
and liver biochemical parameters. The combination of silymarin vented the CCl4 -induced reduction of ascorbic acid excretion in
and P. amarus (50 mg + 50 mg/kg body weight for 6 days orally on urine. The results indicated that the measurement of ascorbic acid
male Rattus norvegicus strain showed synergistic effect for hep- excretion could be used as a non-invasive test for screening protec-
atoprotection and silymarin with ethanolic extract of P. amarus tive substances against CCl4 -induced hepatotoxicity in albino rats
showed better activity due to the higher concentration of phyl- (Visweswaram et al., 1994).
lanthin in ethanolic extract in comparison to aqueous extract
of the plant (Yadav et al., 2008). The hepatoprotective effect of 8.16. Hypoglycemic and hypocholesterolemic activities
methanolic extract of the leaves of P. amarus was evaluated against
ethanol-induced oxidative damage in adult male Wistar albino The hypoglycemic potential of aqueous extract of whole plant
rats. Methanolic extract of P. amarus (250 and 500 mg/kg body of P. amarus was investigated in alloxan-induced diabetic Wistar
weight/day and ethanol 5 g/kg body weight/day 20% w/v) were albino rats. The extract at a dose of 260 mg/kg produced a signifi-
administered orally to animals for 4 and 3 weeks respectively. cant (p < 0.05) reduction in blood glucose level by 112% at 24 h of
It was reported that the ethanol treatment markedly decreased oral administration. A significant reduction (p < 0.01) in blood glu-
the level of reduced GSH, SOD, and CAT in the liver, which were cose level of 81 and 61% (day 7) at doses of 130 and 260 mg/kg
significantly enhanced by P. amarus treatment. GST, which was of extract were observed respectively. The extract also showed a
increased after chronic ethanol administration, was significantly highly significant (p < 0.001) decrease in blood glucose level of 38
reduced by P. amarus treatment in the liver (Faremi et al., 2008). and 30% (day 14) at doses of 130 and 260 mg/kg respectively. On
In vitro study, P. amarus aqueous extract (1–4 mg/mL) increased the administration of 390 mg/kg dose of extract, significant reduc-
%MTT reduction assay and decreased the release of AST and ALT in tion (p < 0.001) in blood glucose level of 41% on day 7 and 16%
rat primary cultured hepatocytes being treated with ethanol. Hep- on day 14 was observed (Mbagwu et al., 2011). The antihyper-
atotoxic parameters studied in vivo included serum AST and ALT, glycemic and hypolipidemic activities of aqueous extract of whole
serum triglyceride (STG), hepatic triglyceride (HTG), TNF-␣, inter- plant of P. amarus were evaluated in streptozotocin (STZ)-induced
leukin 1␤ (IL-1␤), together with histopathological examination. In diabetic male Wistar albino rats. Aqueous extract of P. amarus was
Author's personal copy

J.R. Patel et al. / Journal of Ethnopharmacology 138 (2011) 286–313 307

administered at 200 mg/kg body weight/day to normal treated and taking 1000 mg/kg body weight (Raphael et al., 2002b). Aqueous
STZ-induced diabetic treated rats by gavage for 8 weeks. During extract of aerial parts of P. amarus, 0.1 and 1 g/kg body weight,
the experimental period, blood was collected from fasted rats at 10 significantly enhanced clearance of glucose from the blood as com-
days intervals and plasma glucose level was estimated. The plasma pared to controls during an oral glucose tolerance test (OGTT), using
lipid profile was estimated at the end of experimental period. After normal fasted albino rabbits. Both doses had no effect on blood glu-
the treatment period, kidney LPO, protein oxidation and GSH were cose in the unfed rabbits. A methanolic extract of the aerial parts,
estimated. The significant decrease in the body weight, hyper- 1 g/kg body weight, worsened glucose tolerance causing a signif-
glycemia and hyperlipidemia was observed in STZ-induced diabetic icant increase in area under the OGTT and fasting blood glucose
rats with the treatment of aqueous extract of P. amarus in dia- curves (Moshi et al., 1997).
betic treated group. STZ-induced diabetic rats showed increased
renal oxidative stress with increased LPO and protein oxidation 8.16.1. Clinical study
(Karuna et al., 2011). The ␣-amylase inhibitory activity of ethanol, The glycaemic response to 124.5 ± 9.3 (mean ± SD) g of pancakes
chloroform, and hexane extracts of P. amarus against porcine pan- was monitored in 21 non-insulin dependent diabetic (NIDDM)
creatic amylase in vitro was evaluated. Different concentrations patients while on oral hypoglycaemics, after a week washout period
(10, 20, 40, 60, 80, and 100 ␮g/mL) in DMSO were subjected and after a week twice daily treatment with 100 mL of an aque-
to ␣-amylase inhibitory assay using starch azure as a substrate. ous extract from 12.5 g of powdered aerial parts of P. amarus. After
The ethanol and hexane extracts of P. amarus exhibited apprecia- the week washout period, the fasting blood glucose (FBG) and
ble ␣-amylase inhibitory activity with an IC50 values 36.05 ± 4.01 postprandial blood glucose increased significantly compared with
and 48.92 ± 3.43 ␮g/mL, respectively, when compared with acar- treatment on oral hypoglycaemics (p < 0.05). After a week herbal
bose (IC50 value 83.33 ± 0.34 ␮g/mL). However, the chloroform treatment no hypoglycaemic activity was observed (Moshi et al.,
extract failed to inhibit ␣-amylase activity (Tamil et al., 2010). 2001).
Hydro-alcoholic extract of leaves of P. amarus (HAEPA) was stud-
ied for its in vivo anti-hyperlipidemic potential using cholesterol 8.17. Immunomodulatory activity
diet induced hyperlipidemia model in rats. Results indicated that
HAEPA possessed significant hypolipidemic activity at doses of 300 The methanolic extract of P. amarus was investigated for its
and 500 mg/kg (Umbare et al., 2009). Oral administration of aque- effects on the respiratory burst of human whole blood, iso-
ous extract of whole plant of P. amarus to Wistar albino rats at doses lated human polymorphonuclear leukocytes and isolated mice
of 50, 100 and 200 mg/kg body weight promotes glucose uptake. In macrophages using a luminol/lucigenin-based chemiluminescence
the oral glucose tolerance test P. amarus extract showed significant assay. The effect of the extract on chemotactic migration of poly-
(p < 0.05) reduction of serum glucose level based on the hypo- morphonuclear leukocytes at the concentration of 10, 5, 2.5, 1.25
glycemic effect in normal rats. Daily administration of the extracts and 0.625 ␮g/mL was also tested using the Boyden chamber tech-
for 14 days showed significantly (p < 0.05) reduced AST and ALT and nique. The extract of P. amarus produced the strongest oxidative
urea at 100 mg/kg body weight when compared with other concen- burst of polymorphonuclear leukocytes with luminol-based chemi-
tration doses and that of the control. However significant (p < 0.05) luminescence, with IC50 values of 0.7 ␮g/mL (Jantan et al., 2011).
elevation of AST was observed with animals treated 200 mg/kg Administration of 75% methanolic extract of P. amarus at doses 250
body weight extract. Creatinine of treated animal did not show any and 750 mg/kg body weight significantly reduced the myelosup-
significant (p > 0.05) difference. Significant (p < 0.05) reduction was pression and improved the WBC count, bone marrow cellularity
observed with animal treated 100 mg/kg body weight for urea. The as well as the number of maturing monocytes. CTX treatment also
extract did not produce significant (p > 0.05) effect on heamatolog- reduced the activity of glutathione system and increased the activ-
ical parameters except that the animals receiving the highest dose ity of phase I enzyme that metabolize CTX to its toxic side products.
(200 mg/kg body weight) of the extracts had significant (p < 0.05) P. amarus administration was found to decrease the activity of
lowered PCV and Hb. All the animals gained some weight, while for phase I enzyme. P. amarus also increased the cellular GSH and GST,
treated animal (50 and 100 mg), the weight gain are significantly thereby decreasing the effect of toxic metabolites of CTX on the
(p < 0.05) lower compared to the control (James et al., 2009). The cells. P. amarus did not reduce the tumor reducing activity of CTX. In
aqueous extracts of leave and seed of P. amarus at oral dose of 150, fact, there was a synergistic action of CTX and P. amarus in reducing
300 and 600 mg/kg produced a dose-dependent decrease in the the solid tumors in balb/c mice (Kumar and Kuttan, 2005).
fasting plasma glucose and cholesterol, and reduction in weights
in treated male Swiss mice. The results suggested that the extracts 8.18. Nephroprotective activity
could be enhancing the peripheral utilization of glucose (Adeneye
et al., 2006). Hexane extract of P. amarus had ␣-amylase inhibitory Single oral dose (100–400 mg/kg/day) of the leaves and seed
properties. Extraction and fractionation of P. amarus hexane extract aqueous extracts of P. amarus were studied for their protective
led to the isolation of dotriacontanyl docosanoate, triacontanol and effects in acetaminophen and gentamicin-induced nephrotoxic
a mixture of oleanolic acid and ursolic acid. The compounds were Wistar rats for 14 days. The acetaminophen nephrotoxic rats,
tested in the ␣-amylase inhibition assay and the results revealed 100–400 mg/kg/day significantly (p < 0.05, p < 0.01, p < 0.001) atten-
that the oleanolic acid and ursolic acid (2:1) mixture was a potent uated elevations in the serum creatinine and blood urea nitrogen
␣-amylase inhibitor with IC50 = 2.01 ␮g/mL (4.41 ␮M) and it con- levels in dose related fashion. Similar effects were also recorded in
tributed significantly to ␣-amylase inhibition activity of the extract. the gentamicin model of acute renal injury (Adeneye and Adokiye,
Three pure pentacyclic triterpenoids, oleanolic acid, ursolic acid 2008).
and lupeol isolated from P. amarus were also shown to inhibit ␣-
amylase (Ali et al., 2006). The methanolic extract of leaves and stem 8.19. Radioprotective effect
of P. amarus was found to reduce the blood sugar in alloxan diabetic
rats at 4th hour by 6% at a dose level of 200 mg/kg body weight and The radioprotective activity of pure compounds isolated from
18.7% at a concentration of 1000 mg/kg body weight. Continued the plant P. amarus was studied using rat liver mitochondria and
administration of extract for 15 days produced significant pBR322 plasmid DNA as an in vitro model system. Ellagitannins
(p = 0.001) reduction in blood sugar. On 18th day after alloxan (amariin, 1-galloyl-2, 3-dehydrohexahydroxydiphenyl (DHHDP)-
administration values were almost same to normal in the group glucose, repandusinic acid, geraniin, corilagin, phyllanthusiin D)
Author's personal copy

308 J.R. Patel et al. / Journal of Ethnopharmacology 138 (2011) 286–313

and flavonoids (rutin, and quercetin 3-O-glucoside) at the con- on the kidneys of adult Wistar rats (Andrew and Enogieru, 2011).
centration of 0.1, 0.2, 0.3 and 0.4 nM effectively prevented lipid The effects of chronic administration of aqueous extract of leaves of
peroxidation and protein oxidation in mitochondria. The com- P. amarus on histology of kidney of adult Wistar rats indicated that
pounds also prevented radiation induced single-strand breaks in rats in the treated groups showed some varying degree of distortion
pBR322 plasmid DNA (Londhe et al., 2009). 75% methanolic extract and disruption in microanatomy of the kidney including intersti-
of aerial parts of P. amarus at concentrations of 250 and 750 mg/kg tial oedema and tubular necrosis. It provides further evidence that
body weight on balb/c mice were found to elevate the antioxidant medicinal use of P. amarus has a potential adverse effect on kid-
enzymes in the intestine and decrease the lipid peroxidation levels. ney (Adjene and Nwose, 2010). The aqueous extract of whole plant
Histopathological evaluations of the intestine revealed decreased of P. amarus was found to be more cytotoxic than hydroalcoholic
damage to intestinal cells. P. amarus was found to protect the extract (IC50 being 89.6 ␮g/mL vs. 277 ␮g/mL). Acute and sub-acute
clastogenic effects of radiation as seen from decreased number toxicity of the extract in Swiss mice and Wistar rats respectively
of micronuclei. The administration of P. amarus was also found to showed that no significant differences were observed in body
decrease the percentage of chromosomal aberrations (Harikumar weight gain and blood glucose level between control and treated
and Kuttan, 2007). The radioprotective effect of 75% methanolic groups. Clinical biochemistry revealed no toxic effect. Neither gross
extract of aerial parts of P. amarus was investigated in adult balb/c abnormalities nor histopathological changes in liver, kidney and
mice. P. amarus extract (750 and 250 mg/kg body weight) signif- pancreas were observed. Extract of P. amarus could then be consid-
icantly increased the total WBC count, bone marrow cellularity, ered to be safe in animals by oral route (LD50 > 5 g/kg) even though it
and ␣-esterase activity as compared to untreated radiation exposed is slightly cytotoxic to the human adenocarcinoma cell line Caco-2
animals. P. amarus treatment also increased the activity of various (Lawson-Evi et al., 2008). Chemical and cytotoxicity examinations
antioxidant enzymes, such as SOD, CAT, GST, GPX, and GR, both of the crude methanol extract of the aerial parts of P. amarus
in blood and tissue, which were reduced by radiation treatment. were investigated. The cytotoxicity property of the P. amarus was
There was also a significant increase in GSH levels of blood and tis- evaluated in vitro, using the human ovarian A2780 cancer cell.
sue. Lipid peroxidation levels, which were increased after radiation, Bioassay-guided fraction of the crude extract of the P. amarus (IC50
were significantly reduced by P. amarus treatment, both in serum value of 31.2 ␮g/mL), showed that the dichloromethane fraction
and liver (Kumar and Kuttan, 2004). was most toxic with an IC50 value of 22.7 ␮g/mL, whereas the
polar methanol fraction was least cytotoxic with an IC50 value of
8.20. Spasmolytic activity 31.2 ␮g/mL. This led to the isolation of a new chroman derivative
from the dichloromethane fraction. The compound exhibited very
Potential spasmolytic activity of the extracts of P. amarus was little or no in vitro cytotoxicity with an IC50 value of 16.2 ␮g/mL, rel-
judged by their ability to reduce forces of smooth muscle con- ative to actinomycin, the reference compound, with an IC50 value
traction of a 2 cm long piece of guinea pig ileum induced by of 2.0 ␮g/mL (Ajaiyeoba and Kingston, 2006). Acute oral admin-
EC50 acetylcholine (27 ± 5 ␮g/L) or EC50 histamine (102 ± 13 ␮g/L) istration of P. amarus leaves extract is non-toxic to the rat liver,
(Mans et al., 2004). even at a dose of 5 g/kg body weight. The chronic toxicity stud-
ies of P. amarus extracts administration (of 100–800 mg/kg body
8.21. Effect on reproductive organs weight) showed the absence of cumulative toxicity as reflected by
the non-significant change in the parameters studied as well as
The effect of a carbamate insecticide, carbofuran was studied from the results of the histological studies (Sirajudeen et al., 2006).
on estrous cycle and follicular growth in virgin female Wistar rats Chromatographic fractions obtained from fresh leaves extract of
as well as recovering from the damaged estrous cycle with treat- P. amarus were tested for toxicity on the serum biochemistry of
ment of P. amarus lignans viz. phyllanthin and hypophyllanthin. rats. The six fractions were administered orally to albino rats at the
Since, phyllanthin and hypophyllanthin at the dose of 100 mg/kg doses of 400, 800 and 1600 mg/kg body weight for 14 days. The
body weight have been found to be systemically transformed into animals in the controlled experiment received only distilled water
enterolignan(s), which is known to be responsible for augmenting for the same number of days. The results revealed that some frac-
estrous cycle in rats (Islam et al., 2008b). The aqueous crude extracts tions of P. amarus had potentially deleterious effects on the blood
of P. amarus were administered to 38-week old sexually mature and caused a significant increase in level of ALT, ALP, total bilirubi-
male albino to determine the effect of extract on the male repro- nand blood urea nitrogen when compared with control (Adedapo
ductive organs of these animals. The results from the study revealed et al., 2005a). The aqueous crude extract was administered orally
that the aqueous crude extracts of P. amarus caused varying degrees to male rats of the Sprague Dawley strain in three groups receiv-
of testicular degeneration as well as reduction in the mean semi- ing doses of 400, 800 and 1000 mg/kg but the fourth group served
niferous tubular diameter (STD) in the treated rats (Adedapo et al., as a control and received distilled water only. The pathological
2003). changes by the aqueous crude extract of the leaves of P. amarus
caused a decrease in the red blood cell (RBC) count, packed cell
9. Toxicological assessment and contraindications volume (PCV), haemoglobin concentration (Hb), but an increase in
the white blood cell (WBC) count. The extract also resulted in an
Histological studies to know the effects of oral administration of increase in the levels of aspartate amino transferase (AST), total and
aqueous extract of P. amarus on the kidney of adult Wistar albino conjugated bilirubin, total protein and albumin. The study, how-
rats were carried out at the doses of 500 and 1000 mg/kg body ever, caused a decrease in the level of alanine amino transferase
weight respectively for 28 days, while the control rats received (ALT). Histopathologically, there were cases of protein casts in the
equal volume of distilled water. The rats were sacrificed on day kidney tubules with tubular nephrosis, foci of lymphocytic infil-
29 of the experiment and kidneys were dissected and quickly fixed tration at the portal areas of the liver as well as marked testicular
in 10% formal saline for routine histological study. The histolog- degeneration with severe disorganization of seminiferous tubules,
ical findings indicated that the treated sections of the kidneys which were devoid of spermatic cells. A reduction in the weight of
showed hypertrophy of blood vessels, mild-severe infiltrate of the experimental animals was also recorded. The results revealed
chronic inflammatory cells and varying degrees of tubular necrosis that P. amarus has potential toxic properties (Adedapo et al., 2005b).
when compared to the control sections. The findings indicated that P. amarus has demonstrated hypotensive effects in animals and
the administration of P. amarus extract has some adverse effects humans. People with a heart condition and/or taking prescription
Author's personal copy

J.R. Patel et al. / Journal of Ethnopharmacology 138 (2011) 286–313 309

of heart medications should consult their doctor before taking this (1:1) exhibited antitumor activity against EAC in Swiss albino
plant. It may potentiate insulin and antidiabetic drugs. It contains mice. Lignans nirtetralin, niranthin or phyllanthin exerted cyto-
a naturally occurring phytochemical geraniin. This chemical has toxic effects on K-562 cells. Phyllanthin demonstrated its role in
been documented with negative chronotropic, negative inotropic, protection by antagonizing rat liver cell injury induced by ethanol.
hypotensive and angiotensin-converting enzyme inhibitor effects It effectively alleviated the changes induced by CCl4 in a concen-
in animal studies with frogs, mice and rats (Ueno et al., 1988). As tration dependence manner. Three pure pentacyclic triterpenoids,
such, this plant may potentiate antihypertensive drugs, ␤-blockers oleanolic acid, ursolic acid and lupeol isolated from hexane extract
and other heart medications (including chronotropic and inotropic of P. amarus were shown to inhibit ␣-amylase. More importantly,
drugs). It has been considered in herbal medicine to be abortive (at there have been no side effects or toxicity reports from many years
high dosages) as well as an emmenagogue. Although it is not stud- of research on this herb. Thus activity guided phytochemical and
ied in humans but animal studies do indicated that it has uterine phytoanalytical information appears to be very useful and might
relaxant effects. It is therefore contraindicated during pregnancy. It led to development of novel agents for various disorders and could
has been documented with female antifertility effects in mice (the be explored further for commercial purposes. However, there are
effect was reversed in 45 days after cessation of dosing) (Rao and many aspects, which need to be explored like well-controlled clin-
Alice, 2001). While this effect has not been documented in humans, ical trials using large sample size (large number of patients) for the
the use of the plant is probably contraindicated in women seek- efficacy and toxicity, the mechanism of biological activity of active
ing pregnancy or taking fertility drugs. Chronic and acute use of constituents present in the plant. On the basis of biological activities
this plant may be contraindicated in various other medical con- of P. amarus, crude extract and derived phytochemicals and their
ditions where diuretics are not advised. Chronic long-term use of uses as pharmacological agents in traditional and modern research
any diuretic can cause electrolyte and mineral imbalances; how- are possible but will first require more clinical trials and product
ever, human studies with P. amarus (for 3 months of chronic use) development. The current evidence is large limited to correlation
have not reported any side effects. between identified phytochemicals and mode of action for any
pharmacological activity. Mechanism of action studies are expected
to lead the way in the discovery of new agents with improved
10. Conclusion and intriguing pharmacological properties. This could be achieved
by molecular modeling studies involving interaction of bioactive
The scientific research on P. amarus suggests a huge biolog- phytochemicals from P. amarus with their respective molecular tar-
ical potential of this plant. It is strongly believed that detailed gets and the extract of P. amarus could be further explored in the
information as presented in this review on the phytochemical and future as a source of useful phytochemicals for the pharmaceutical
various biological properties of the plant might provide detailed industry.
evidence for the use of this plant in different diseases. It has
various traditional uses that differ from one country to another References
whereas some important uses for the treatment of jaundice, dia-
betes, dysentery, fever, gonorrhea, syphilis and stomachache and Abhyankar, G., Suprasanna, P., Pandey, B.N., Mishra, K.P., Rao, K.V., Reddy, V.D., 2010.
skin diseases are almost common. P. amarus, a potent herbal Hairy root extract of Phyllanthus amarus induces apoptotic cell death in human
breast cancer cells. Innovative Food Science and Emerging Technologies 11,
medicine is attracting researchers since many decades due to its 526–532.
high therapeutic value. There is a demand to standardize the prop- Abo, K.A., Fred-Jaiyesimi, A.A., Jaiyesimi, A.E.A., 2008. Ethnobotanical studies of
erties of P. amarus and their detailed clinical trials. Pharmacological medicinal plants used in the management of diabetes mellitus in South Western
Nigeria. Journal of Ethnopharmacology 115, 67–71.
and chemical studies have demonstrated that the extracts of the Adedapo, A.A., Abatan, M.O., Akinloye, A.K., Idowu, S.O., Olorunsogo, O.O., 2003. Mor-
plant possess various pharmacological actions viz. antiviral, anti- phometric and histopathological studies on the effects of some chromatographic
inflammatory antimalarial, antimicrobial, anticancer, antidiabetic, fractions of Phyllanthus amarus and Euphorbia hirta on the male reproductive
organs of rats. Journal of Veterinary Science 4, 181–185.
hypolipidemic, hepatoprotective and nephroprotective. Owing to
Adedapo, A.A., Abatan, M.O., Idowu, S.O., Olorunshogo, O.O., 2005a. Toxic effects of
the impressive preclinical therapeutic potential, the plant extracts chromatographic fractions of Phyllanthus amarus on the serum biochemistry of
have been evaluated in human trials for the treatment of HIV, jaun- rats. Phytotherapy Research 19, 812–815.
Adedapo, A.A., Adegbayibi, A.Y., Emikpe, B.O., 2005b. Clinicopathological changes
dice, hypertension and diabetes.
associated with the aqueous extract of the leaves of Phyllanthus amarus in rats.
P. amarus is reported to contain lignans, flavonoids, hydrolysable Phytotherapy Research 19, 971–976.
tannins (ellagitannins), polyphenols, triterpenes, sterols and Adeneye, A.A., Adokiye, S.B., 2008. Protective effect of aqueous leaf and seed extract
alkaloids. The phytochemicals exhibited different structural char- of Phyllanthus amarus on gentamicin and acetaminophen-induced nephrotoxic
rats. Journal of Ethnopharmacology 118, 318–323.
acteristics with various pharmacological actions. The lignans Adeneye, A.A., Amole, O.O., Adeneye, A.K., 2006. Hypoglycemic and hypercholes-
nirtetralin, phyltetralin or niranthin isolated from P. amarus sig- terolemia activities of aqueous leaf and seed extract of Phyllanthus amarus in
nificantly inhibited PAF-induced paw oedema formation in mice. mice. Fitoterapia 77, 511–514.
Adjanohoun, E.J., Ahyi, M.R.A., Ake Assi, L., Akpagana, K., Chibon, P., El-Haji, A.,
Niranthin decreased the specific binding of 3[H]-PAF in mouse Eyme, J., Garba, M., Gassita, J.N., Gbeassor, M., Goudote, E., Guinko, S., Hodouto,
cerebral cortex membranes. Phyltetralin, nirtetralin and niranthin K.K., Houngnon, P., Keita, A., Keoula, Y., Kluga-Ocloo, W.P., Lo, I., Siamevi, K.M.,
also inhibited carrageenan-induced paw oedema and neutrophil Taffame, K.K., 1986. Contribution aux etudes ethnobotaniques et floristiques au
Togo, first ed. Agence de Coopération Culturelle et Technique, première edition
influx. Niranthin also showed the best anti-HBsAg activity while (ACCT), Paris.
the most potent anti-HBeAg activity was observed with henokinin. Adjene, J.O., Nwose, E.U., 2010. Histological effects of chronic administration of Phyl-
The presence of high contents of phenolic compounds in the aque- lanthus amarus on the kidney of adult Wistar rat. North American Journal of
Medical Sciences 2, 193–195.
ous extract of P. amarus was found to have strong and significant
Adjobimey, T., Edaye, I., Lagnika, L., Gbenou, J., Moudachirou, M., Sanni, A., 2004.
antioxidant activity. Phyllanthin isolated from aqueous extract In vitro antiplasmodial activity of some antimalarial plants of Beninese pharma-
of leaves of P. amarus exhibited very high antioxidant activity. copoeia. Comptes Rendus Chimie 7, 1023–1027.
Agrawal, A., Srivastava, S., Srivastava, J.N., Srivasava, M.M., 2004. Evaluation of
Amariin, repandusinic acid and phyllanthusiin D showed higher
inhibitory effect of the plant Phyllanthus amarus against dermatophytic fungi
antioxidant activity. A gallotanin containing fraction and the iso- Microsporum gypseum. Biomedical and Environmental Sciences 17, 359–365.
lated elllagitanins geraniin and corilagin were shown to be most Ahirrao, Y.A., Patil, D.A., 2010. Indigenous healthcare practices in Buldhana district
potent mediators of antiviral activities. The purified gallatanins (Maharashtra). Indian Journal of Natural Products and Resources 1, 85–88.
Ahmad, B., Alam, T., 2003. Components from whole plant of Phyllanthus amarus Linn.
geraniin and corilagin were most active to inhibit HIV-1 replica- Indian Journal of Chemistry, Section B: Organic Chemistry including Medicinal
tion in Hela CD4+ cells. Mixture of phyllanthin and hypophyllanthin Chemistry 42, 1786–1790.
Author's personal copy

310 J.R. Patel et al. / Journal of Ethnopharmacology 138 (2011) 286–313

Ajaiyeoba, E., Kingston, D., 2006. Cytotoxicity evaluation and isolation of a chroman Doshi, J.C., Vaidya, A.B., Antarkar, D.S., Deolalikar, R., Antani, D.H., 1994. A two-stage
derivative from Phyllanthus amarus aerial part extract. Pharmaceutical Biology clinical trial of Phyllanthus amarus in hepatitis B carriers: failure to eradicate the
44, 668–671. surface antigen. Indian Journal of Gastroenterology 13, 7–8.
Ajala, T.O., Igwilo, C.I., Oreagba, I.A., Odeku, O.A., 2011. The antiplasmodial effect of Dymock, W., 1886. The Vegetable Materia Medica of Western India. Education Soci-
the extracts and formulated capsules of P. amarus on Plasmodium yoelii infection ety’s Press, Bombay, pp. 701–702.
in mice. Asian Pacific Journal of Tropical Medicine 4, 283–287. Dymock, W., Warden, C.J.H., Hooper, D., 1893. Pharpacographta Indica, Vol. 3. Kegan
Ajibesin, K.K., Ekpoa, B.A., Bala, D.N., Essien, E.E., Adesanya, S.A., 2008. Ethnobotan- Paul, Trench, Trubner and Co. Inc, London, pp. 265–268.
ical survey of Akwa Ibom State of Nigeria. Journal of Ethnopharmacology 115, Eldeen, I.M.S., Seow, E.M., Abdullah, R., Sulaiman, S.F., 2011. In vitro antibacterial,
387–408. antioxidant, total phenolic contents and anti-HIV-1 reverse activities of extracts
Akinjogunla, O.J., Eghafona, N.O., Enabulele, I.O., Mboto, C.I., Ogbemudia, F.O., of seven Phyllanthus sp. South African Journal of Botany 77, 75–79.
2010. Antibacterial activity of ethanolic extracts of Phyllanthus amarus against Faremi, T.Y, Suru, S.M., Fafunso, M.A., Obioha, U.E., 2008. Hepatoprotective potentials
extended spectrum ␤-lactamase producing Escherichia coli isolated from stool of Phyllanthus amarus against ethanol induced oxidative stress in rats. Food and
samples of HIV sero-positive patients with or without diarrhea. African Journal Chemical Toxicology 46, 2658–2664.
of Pharmacy and Pharmacology 4, 402–407. Foo, L.Y., 1993a. Amarulone, a novel cyclic hydrolyzable tannin from Phyllanthus
Ali, H., Houghton, P.J., Soumyanath, A., 2006. ␣-Amylase inhibitory activity of some amarus. Natural Product Letters 3, 45–52.
Malaysian plants used to treat diabetes; with particular reference to Phyllanthus Foo, L.Y., 1995. Amarinic acid and related ellagitannins from Phyllanthus amarus.
amarus. Journal of Ethnopharmacology 107, 449–455. Phytochemistry 39, 217–224.
Alli, A.I., Ehinmidu, J.O., Ibrahim, Y.K.E., 2011. Preliminary phytochemical screening Foo, L.Y., Wong, H., 1992. Phyllanthusiin D, an unusual hydrolysable tannin from
and antimicrobial activities of some medicinal plants used in Ebiraland. Bayero Phyllanthus amarus. Phytochemistry 31, 711–713.
Journal of Pure and Applied Sciences 4, 10–16. Foo, L.Y., 1993b. Amariin, a di-dehydro hexahydroxy diphenoyl hydrolysable tannin
Alvari, A., Rafsanjani, M.S.O., Ahmed, F.J., Hejazi, M.S., Abdin, M.Z., 2011. Rapid from Phyllanthus amarus. Phytochemistry 33, 487–491.
RP-HPLC technique for the determination of phyllanthin as bulk and its quan- George, V., Muthukrishnan, M., Chojnacki, T., 2001. The occurrence of polyphenols
tification in Phyllanthus amarus extract. International Journal of Phytomedicine in Euphorbiaceae. Acta Societatis Botanicorum Poloniae 70, 39–41.
3, 115–119. Guha, G., Rajkumar, V., Ashok, K.R., Mathew, L., 2010. Aqueous extract of Phyllanthus
Andrew, E.O., Enogieru, A., 2011. Effects of oral administration of Phyllanthus amarus inhibits chromium (VI)-induced toxicity in MDA-MB-435S cells. Food
amarus leaf extract on the kidneys of adult Wistar rats – a histological study. and Chemical Toxicology 48, 396–401.
African Journal of Traditional Complementary and Alternative Medicines 8, Guo, Z.-H., Ni, L., Jiang, S.-H., 2007. Determination of gallic acid in Phyllanthus amarus
307–311. capsules by HPLC. Huaxi Yaoxue Zazhi 22, 81–82.
Annamalai, A., Laxmi, P.T.V., 2009. HPLC and HPTLC analysis of bioactive phyllanthin Hamrapurkar, P., Phale, M., Pawar, S., 2009. Extraction, isolation and characteriza-
from different organs of Phyllanthus amarus. Asian Journal of Biotechnology 1, tion of phyllanthin from Phyllanthus amarus with preliminary phytochemical
154–162. evaluation of the crude extract. Natural Products: An Indian Journal 5,
Asase, A., Akwetey, G.A., Achel, D.G., 2010. Ethnopharmacological use of herbal reme- 120–124.
dies for the treatment of malaria in the Dangme West District of Ghana. Journal Hamrapurkar, P., Pawar, S., Phale, M., 2010. Quantitative HPTLC analysis of phyllan-
of Ethnopharmacology 129, 367–376. thin in Phyllanthus amarus. Journal of Planar Chromatography 23, 112–115.
Balakrishnan, V., Prema, P., Ravindran, K.C., Philip Robinson, J., 2009. Ethnobotani- Handa, K.L., Kapoor, L.D., Chopra, I.C., Nath, S., 1951. The present position of the crude
cal studies among villagers from Dharapuram Taluk, Tamil Nadu, India. Global drugs used in the indigenous medicine. Indian Journal of Pharmacy 13, 29–48.
Journal of Pharmacology 3, 8–14. Hari Kumar, K.B., Kuttan, R., 2006. Inhibition of drug metabolizing enzymes
Balasubramanian, G., Sarathi, M., Rajesh Kumar, S., Sahul Hameed, A.S., 2007. Screen- (cytochrome P450) in vitro as well as in vivo by Phyllanthus amarus Schum &
ing the antiviral activity of Indian medicinal plants against white spot syndrome Thonn. Biological and Pharmaceutical Bulletin 29, 1310–1313.
virus in shrimp. Aquaculture 263, 15–19. Harikumar, K.B, Kuttan, G., Kuttan, R., 2009. Phyllanthus amarus inhibits cell growth
Bhattacharyya, R., Bhattacharya, S., 2004. Development of a potent in-vitro source and induces apoptosis in Dalton’s Lymphoma Ascites Cells through activa-
of Phyllanthus amarus roots with pronounced activity against surface antigen of tion of caspase-3 and downregulation of Bcl-2. Integrative Cancer Therapies 8,
the hepatitis B virus. In-vitro Cellular & Development Biology. Plant 40, 504–508. 190–194.
Blumberg, B.S., Millman, I., Venkateswaran, P.S., Thyagarajan, S.P., 1990. Hepatitis Harikumar, K.B., Kuttan, G., 2009. Inhibition of viral carcinogenesis by Phyllanthus
B virus and primary hepatocellular carcinoma: treatment of HBV carriers with amarus. Integrative Cancer Therapies 8, 254–260.
Phyllanthus amarus. Vaccine 8, 86–92. Harikumar, K.B., Kuttan, R., 2007. An extract of Phyllanthus amarus protects mouse
Blumberg, B.S., Millman, I., Venkateswaran, P.S., Thyagarajan, S.P., 1989. Hepatitis B chromosomes and intestine from radiation induced damages. Journal of Radia-
virus and hepatocellular carcinoma-treatment of HBV carriers with Phyllanthus tion Research 48, 469–476.
amarus. Cancer Detection Prevention 14, 195–201. Heyde, H., 1968. Surinaamse planten als volksmedicijn. Surinamese plants as folk
Cabieses, F., 1993. Apuntes de medicina traditional. La racionalizacion de lo irra- medicine. GRANMA-MK, Paramaribo-Suriname, 33 p.
cional. Notes of traditional medicine. Consejo Nacional de Ciencia Technologia Heyde, H., 1990. Medicijn planten in Suriname (Den dresi wiwiri foe Sranan). Medic-
Concytec Lima-Peru, pp. 414. inal Plants in Suriname. Uitg. Stichting Gezondheidsplanten Informaite (SGI)
Cartaxo, S.L., de Almeida Souza, M.M., de Albuquerque, U.P., 2010. Medicinal plants Paramaribo, 157 p.
with bioprospecting potential used in semi-arid Northeastern Brazil. Journal of Houghton, P.J., Woldemariama, T.Z., Siobhan, O.S., Thyagarajan, S.P., 1996. Two
Ethnopharmacology 131, 326–342. securinega type alkaloids from Phyllanthus amarus. Phytochemistry 43,
Chaudhuri, P.K., Bagchi, G., Srivastava, R., Kumar, S., 2001. Procedure for the extrac- 715–717.
tion and isolation of phyllanthin from the plant Phyllanthus amarus. Ger. Offen. Huang, R.L., Huang, Y.L., Ou, J.C., Chen, C.C., Hsu, F.L., Chang, C., 2003. Screening of 25
6 pp. CODEN: GWXXBX DE 10014674 A1 20011031. compounds isolated from Phyllanthus Species for anti-human hepatitis B virus
Chevallier, A., 2000. Encyclopedia of Herbal Medicine: Natural Health, Second ed. in vitro. Phytotherapy Research 17, 449–453.
Dorling Kindersley Book, USA, 336 p. Idu, M., Ejale, A., Timothy, O., Comor, A.O., 2008b. Common medicinal plants sold in
Chirdchupunseree, H., Pramyothin, P., 2010. Protective activity of phyllanthin in some local markets in Warri, Delta State, Nigeria. Plant Archives 8, 557–561.
ethanol treated primary culture of rat hepatocytes. Journal of Ethnopharmacol- Idu, M., Timothy, O., Oghinan, O.M., 2008a. Plants used for ethnomedicine in Esan
ogy 128, 172–176. North East local government area of Edo State in Nigeria. Ethnobotany 20,
Chopra, R.N., Chopra, I.C., Handa, K.L., Kapur, L.D., 1958. Indigenous Drugs of India, 85–90.
2nd ed. UN Dhar and Sons Pvt. Ltd., Calcutta, pp. 12–23, 519, 520, 598, 605, 609, Ignacimuthu, S., Ayyanar, M., Sankarasivaraman, K., 2008. Ethnobotanical study of
660. medicinal plants used by Paliyar tribals in Theni district of Tamil Nadu, India.
Chowdhury, L.B., Rao, R.R., 2002. Taxonomic study of herbaceous species of Phyllan- Fitoterapia 79, 562–568.
thus L. (Euphorbiaceae) in India. Phytotaxonomy 2, 143–162. Igwe, C.U., Nwaogu, L.A., Ujuwondu, C.O., 2007. Assessment of the hepatic effects,
Dapper, D.V., Aziagba, B.N., Ebong, O.O., 2007. Antiplasmodial effects of the aqueous phytochemical and proximate compositions of Phyllanthus amarus. African Jour-
extract of Phyllanthus amarus Schum. and Thonn. against Plasmodium berghei in nal of Biotechnology 6, 728–731.
Swiss albino mice. Nigerian Journal of Physiological Sciences 22, 19–25. Inta, A., Shengji, P., Balslev, H., Wangpakapattanawong, P., Trisonthi, C., 2008. A
De, B., Datta, P.C., 1990. Pharmacognostic evaluation of Phyllanthus amarus. Interna- comparative study on medicinal plants used in Akha’s traditional medicine
tional Journal of Crude Drug Research 28, 81–88. in China and Thailand, cultural coherence or ecological divergence? Journal of
Deb, S., Mandal, S.K., 1996. TLC-densitometric determination of phyllanthin and Ethnopharmacology 116, 508–517.
hypophyllanthin in Phyllanthus amarus (bhumiamalaki) and in polyherbal for- Iranloye, B.O., Owoyele, V.B., Kelani, O.R., Olaleye, S.B., 2011. Analgesic activity of
mulation. Indian Drugs 33, 415–416. aqueous leaf extracts of Phyllanthus amarus. African Journal of Medicine and
Dhalwal, K., Birandar, Y.S., Rajani, M., 2006. High performance thin layer chromatog- Medical Sciences 40, 47–50.
raphy densitometric method for simultaneous quantitation of phyllanthin, Islam, A., Naskar, S., Mazumder, U.K., Gupta, M., Ghosal, S., 2008b. Estrogenic prop-
hypophyllanthin, gallic acid and ellagic acid in Phyllanthus amarus. Journal of erties of phyllanthin and hypophyllanthin from Phyllanthus amarus against
A.O.A.C. International 89, 619–623. carbofuran induced toxicity in female rats. Pharmacologyonline 3, 1006–1016.
Dhir, H., Roy, A.K., Sharma, A., Talukder, G., 1990. Protection afforded by aque- Islam, A., Selvan, T., Mazumder, U.K., Gupta, M., Ghosal, S., 2008a. Antitumour effect
ous extracts of Phyllanthus species against cytotoxicity induced by lead and of phyllanthin and hypophyllanthin from Phyllanthus amarus against Ehrlich
aluminium salts. Phytotherapy Research 4, 172–176. ascites carcinoma in mice. Pharmacologyonline 2, 796–807.
Dhooghe, L., Meert, H., Cimanga, R.K., Vlietinck, A.J., Pieters, L., Apers, S., 2011. The Jain, A., Katewa, S.S., Galav, P.K., Sharma, P., 2005. Medicinal plant diversity of Sita-
quantification of ellagic acid in the crude extracts of Phyllanthus amarus Schum. mata wildlife sanctuary, Rajasthan, India. Journal of Ethnopharmacology 102,
& Thonn. (Euphorbiaceae). Phytochemical Analysis 22, 361–366. 143–157.
Author's personal copy

J.R. Patel et al. / Journal of Ethnopharmacology 138 (2011) 286–313 311

Jain, N., Shasany, A.K., Sundaresan, V., Rajkumar, S., Darokar, M.P., Bagchi, G.D., Kumar, K.B.H., Kuttan, R., 2004. Protective effect of an extract of Phyllanthus amarus
Gupta, A.K., Kumar, S., Khanuja, S.P.S., 2003. Molecular diversity in Phyllan- against radiation induced damage in mice. Journal of Radiation Research 45,
thus amarus assessed through RAPD analysis. Current Science (Bangalore) 85, 133–139.
1454–1458. Kumar, K.B., Kuttan, R., 2005. Chemoprotective activity of an extract of Phyllanthus
James, D.B., Owolabi, O.A.N., Hassan Elebo, S., Odemene, L., 2009. Glucose toler- amarus against cyclophosphamide induced toxicity in mice. Phytomedicine 12,
ance test and some biochemical effect of Phyllanthus amarus aquoeus extacts on 494–500.
normaglycemic albino rats. African Journal of Biotechnology 8, 1637–1642. Kumaran, A., Karunakaran, R.J., 2007. In vitro antioxidant activities of methanol
Jantan, I., Harun, N.H., Septama, A.W., Murad, S., Mesaik, M.A., 2011. Inhibition extract of five Phyllanthus species from India. LWT – Food Science and Tech-
of chemiluminescence and chemotactic activitis of phagocytes in vitro by nology 40, 344–352.
the extracts of selected medicinal plants. Journal of Natural Medicine 65, Kuppusamy, C., Murugan, K., 2010. In vitro antimalarial activity of traditionally
400–405. used Western Ghats plants from India and their interactions with chloroquine
Jayaram, S., Thyagarajan, S.P., 1996. Inhibition of HbsAg secrection from Alexander against chloroquine-resistant Plasmodium falciparum. Parasitology Research
cell line by Phyllanthus amarus. Indian Journal of Pathology and Microbiology 107, 1351–1364.
39, 211–215. Lawson-Evi, P., Eklu-Gadegbeku, K., Agbonon, A., Aklikokou, K., Moukha, S.E., Creppy,
Jayaram, S., Thyagarajan, S.P., Sumathi, S., Manjula, S., Malathi, S., Madanagopalan, E., Gbéassor, M., 2008. Toxicological assessment on extracts of Phyllanthus
N., 1997. Efficiency of Phyllanthus amarus treatment in acute viral hepatitis A. amarus Schum & Thonn. Scientific Research and Essay 3, 410–415.
B and non A and non B: an open clinical trial. Indian Journal of Viorology 13, Lee, C.D., Ott, M., Thyagarajan, S.P., Shafritz, D.A., Burk, R.D., Gupta, S., 1996.
59–64. Phyllanthus amarus down-regulates hepatitis B virus mRNA transcription and
Jeena, K.J., Joy, K.L., Kuttan, R., 1999. Effect of Emblica officinalis. Phyllanthus amarus replication. European Journal of Clinical Investigation 26, 1069–1076.
and Picrorrhiza kurroa on N-nitrosodiethylamine induced hepatocarcinogenesis. Lee, S.H., Jaganath, I.B., Wang, S.M., Sekaran, S.D., 2011. Antimetastatic effects of
Cancer Letters 136, 11–16. Phyllanthus on human lung (A549) and breast (MCF-7) cancer cell lines. PLoS
Joshi, C.S., Priya, E.S., 2007. ␤-Glucuronidase inhibitory effect of phenolic con- ONE 6, e20994, doi:10.1371/journal.pone.0020994.
stituents from Phyllanthus amarus. Pharmaceutical Biology 45, 363–365. Leite, D.F., Kassuya, C.A., Mazzuco, T.L., Silvestre, A., de Melo, L.V., Rehder, V.L.,
Joshi, H., Parle, M., 2007. Pharmacological evidences for antiamnesic potentials Rumjanek, V.M., Calixto, J.B., 2006. The cytotoxic effect and the multi-drug resis-
of Phyllanthus amarus in mice. African Journal of Biomedical Research 10, tance reversing action of lignans from Phyllanthus amarus. Planta Medica 72,
165–173. 1353–1358.
Joshi, H., Parle, M., 2006. Evaluation of antiamnesic potentials of [6]-gingerol and Lim, Y.Y., Murtijaya, J., 2007. Antioxidant properties of Phyllanthus amarus extracts
phyllanthin in mice. Natural Products 2, 109–117. as affected by different drying methods. LWT – Food Science and Technology 40,
Joy, K.L., Kuttan, R., 1998. Inhibition by Phyllanthus amarus of hepatocarcinogen- 1664–1669.
esis induced by N-nitrosodiethylamine. Journal of Clinical Biochemistry and Linnaeus, C., 1738. Hotus Cliffortianus Amstel, p. 438.
Nutrition 24, 133–139. Liu, J., Lin, H., McIntosh, H., 2001. Genus Phyllanthus for chronic Hepatitis B virus
Kala, C.P., Dhyani, P.P., Sajwan, B.S., 2006. Developing the medicinal plants sector infection: a systematic review. Journal of Viral Hepatology 8, 358–366.
in Northern India: challenges and opportunities. Journal of Ethnobiology and Londhe, J.S., Devasagayam, T.P., Foo, L.Y., Ghaskadbi, S.S., 2008. Antioxidant activity
Ethnomedicine 2, 1–15. of some polyphenol constituents of the medicinal plant Phyllanthus amarus Linn.
Karuna, R., Bharathi, V.G., Reddy, S.S., Ramesh, B., Saralakumari, D., 2011. Protec- Redox Report 13, 199–207.
tive effects of Phyllanthus amarus aqueous extract against renal oxidative stress Londhe, J.S., Devasagayam, T.P., Foo, L.Y., Ghaskadbi, S.S., 2009. Radioprotective
in Streptozotocin-induced diabetic rats. Indian Journal of Pharmacology 43, properties of polyphenols from Phyllanthus amarus Linn. Journal of Radiation
414–418. Research 50, 303–309.
Karuna, R., Reddy, S.S., Baskar, R., Saralakumari, D., 2009. Antioxidant potential of Maciel, M.A.M., Cunha, A., Dantas, F.T.N.C., Kaiser, C.R., 2007. NMR characterization of
aqueous extract of Phyllanthus amarus in rats. Indian Journal of Pharmacology bioactive lignans from Phyllanthus amarus Schum & Thonn. Journal of Magnetic
41, 64–67. Resonance Imaging 6, 76–82.
Kassuya, C.A.L., Leite, D.F.P., Melo, L.V., de Rehder, V.L.G., Calixto, J.B., 2005. Mahat, M.A., Patil, B.M., 2007. Evaluation of antiinflammatory activity of methanol
Anti-inflammatory properties of extracts, fractions and lignans isolated from extract of Phyllanthus amarus in experimental animal models. Indian Journal of
Phyllanthus amarus. Planta Medica 71, 721–726. Pharmaceutical Science 69, 33–36.
Kassuya, C.A.L., Silvestre, A.A., Rehder, V.L.G., Calixto, J.B., 2003. Anti-allodynic and Mahishi, P., Srinivasa, B.H., Shivanna, M.B., 2005. Medicinal plant wealth of local
anti-oedematogenic properties of the extract from Phyllanthus amarus in models communities in some villages in Shimoga District of Karnataka, India. Journal of
of persistent inflammatory and neuropathic pain. European Journal of Pharma- Ethnopharmacology 98, 307–312.
cology 478, 145–153. Mans, D., Toelsie, J., Jagernath, Z., Ramjiawan, K., Brussel, A.V., Jhanjan, N., Orie, S.,
Kassuya, C.A., Silvestre, A., Menezes-de-Lima Jr., O., Marotta, D.M., Rehder, V.L., Muringen, M., Elliot, U., Jurgens, S., Macnack, R., Rigters, F., Mohan, S., Chigharoe,
Calixto, J.B., 2006. Antiinflammatory and antiallodynic actions of the lignan V., Illes, S., Bipat, R., 2004. Assessment of eight popularly used plant-derived
niranthin isolated from Phyllanthus amarus. Evidence for interaction with preparations for their spasmolytic potential using the isolated guinea pig ileum.
platelet activating factor receptor. European Journal of Pharmacology 546, Pharmaceutical Biology 42, 422–429.
182–188. May, A.I., 1982. Surinaams Kruidenboek. Sranan Oso Dresi. Surinamese Book of
Khatoon, S., Rai, V., Singh Rawat, A.K., Mehrotra, S., 2006. Comparative pharmacog- Herbs. Uitgeverij Vaco, Paramaribo-Suriname, 80 p.
nostic studies of three Phyllanthus species. Journal of Ethnopharmacology 104, Mazumder, A., Mahato, A., Mazumder, R., 2006. Antimicrobial potentiality of Phyl-
79–86. lanthus amarus against drug resistant pathogens. Natural Product Research 20,
Kiemer, A.K., Hartung, T., Huber, C., Vollmar, A.M., 2003. Phyllanthus amarus has 323–326.
anti-inflammatory potential by inhibition of iNOS, COX-2, and cytokines via the Mbagwu, H.O.C., Jackson, C., Jackson, I., Ekpe, G., Eyaekop, U., Essien, G., 2011. Eval-
NF-kappa ␤ pathway. Journal of Hepatology 38, 289–297. uation of the hypoglycemic effect of aqueous extract of Phyllanthus amarus in
Kirtikar, K.R., Basu, B.D., 1975. Indian Medicinal Plants, 2nd ed. Bishen Singh Mahen- alloxan-induced diabetic albino rats. International Journal of Pharmaceutical
dra Pal Singh, New Delhi, pp. 2225. and Biomedical Research 2, 158–160.
Kloucek, P., Polesny, Z., Svobodova, B., Vlkova, E., Kokoska, L., 2005. Antibacterial Medicinal Plants of India, 1987. CSIR Publication, New Delhi.
screening of some Peruvian medicinal plants used in Calleria District. Journal of Mehrotra, R., Rawat, S., Kulshreshtha, D.K., Goyal, P., Patnaik, G.K., Dhawan, B.N.,
Ethnopharmacology 99, 309–312. 1991. In-vitro effect of Phyllanthus amarus on hepatitis B virus. Indian Journal of
Koday, N.K., Rangaiah, G.S., Bobbrala, V., Cherukuri, S., Duggirala, V., 2009. In-vitro Medical Research 93, 71–73.
control of some ocular infectious bacteria by using medicinal plants Adhatoda Mitra, R.L., Jain, S.K., 1985. Concept of Phyllanthus niruri (Euphorbiaceae) in Indian
vasica, Cassia occidentalis and Phyllanthus amarus. Journal of Pharmacy Research Floras. Bulletin of Botanical Survey of India 27, 161–176.
2, 1869–1872. Moronkola, D.O., Ogunwande, I.A., Oyewole, I.O., Baser, K.H.C., Ozek, T., Ozek, G.,
Kolodziej, H., Burmeister, A., Trun, W., Radtke, O.A., Kiderlen, A.F., Ito, H., Hatano, 2009. Studies on the volatile oils of Momordica charantia L. (Cucurbitaceae)
T., Yoshida, T., Foo, L.Y., 2005. Tannins and related compounds induce nitric and Phyllanthus amarus Sch. et Thonn (Euphorbiaceae). Journal of Essential Oil
oxide synthase and cytokines gene expressions in Leishmania major-infected Research 21, 393–399.
macrophage-like RAW 264.7 cells. Bioorganic and Medicinal Chemistry 13, Morton, J.F., 1981. Atlas of Medicinal Plants of Middle America. Library of Congress
6470–6476. Cataloging in Publication Data. Thomas books, 1420 p.
Komuraiah, A., Bolla, K., Rao, K.N., Ragan, A., Raju, V.S., Singara Charya, M.A., 2009. Moshi, M.J., Lutale, J.J., Rimoy, G.H., Abass, Z.G., Josiah, R.M., Swai, A.B., 2001. The
Antibacterial studies and phytochemical constituents of South Indian Phyllan- effect of Phyllanthus amarus aqueous extract on blood glucose in non-insulin
thus species. African Journal of Biotechnology 8, 4991–4995. dependent diabetic patients. Phytotherapy Research 15, 577–580.
Krithika, A., Verma, R.R.J., 2009b. Mitigation of carbon tetrachloride induced dam- Moshi, M.J., Uiso, F.C., Mahmunnah, R.L.A., Malele, S.R., Swai, A.B.M., 1997. A study
age by Phyllanthus amarus in liver of mice. Acta Poloniae Pharmaceutica – Drug of the effect of Phyllanthus amarus extracts on blood glucose in rabbits. Interna-
Research 66, 439–445. tional Journal of Pharmacognosy 35, 167–173.
Krithika, A.R., Verma, R.J., 2009a. Ameliorative potential of Phyllanthus amarus Mukherjee, P.K., Wahile, A., Kumar, V., Rai, S., Mukherjee, K., Saha, B.P., 2006. Marker
against carbon tetrachloride induced hepatotoxicity. Acta Poloniae Pharmaceu- profiling of botanicals used for hepatoprotection in Indian system of medicine.
tica – Drug Research 66, 579–583. Drug Information Journal 40, 131–139.
Krithika, R., Mohankumar, R., Verma, R.J., Shrivastav, P.S., Mohamad, I.L., Munshi, A., Mehrotra, R., Panda, S.K., 1993b. Evaluation of Phyllanthus amarus
Gunasekaran, P., Narasimhan, S., 2009. Isolation, characterization and antiox- and Phyllanthus maderaspatensis as agents for postexposure prophylaxis in
idative effect of phyllanthin against CCl4 -induced toxicity in HepG2 cell line. neonatal duck hepatitis B virus infection. Journal of Medical Virology 40,
Chemico Biological Interaction 181, 351–358. 53–58.
Author's personal copy

312 J.R. Patel et al. / Journal of Ethnopharmacology 138 (2011) 286–313

Munshi, A., Mehrotra, R., Ramesh, R., Panda, S.K., 1993a. Evaluation of anti- Row, L.R., Satyanarayana, P., Subba Rao, G.S.R., 1967. Crystalline constituents of
hepadnavirus activity of Phyllanthus amarus and Phyllanthus maderaspatensis Euphorbeaceae – the synthesis and absolute configuration of phyllanthin. Tetra-
in duck hepatitis B virus carrier Pekin ducks. Journal of Medical Virology 41, hedron 23, 1915.
275–281. Rumphius, G.E., 1750. Herbarium Amboinense, vol. 6. Meinard Uyrwerf, Amsterdam,
Murali, B., Amit, A., Anand, M.S., Dinesh, T.K., Samiulla, D.S., 2001. An improved pp. 41–43.
HPLC method for estimation of phyllanthin and hypophyllanthin in Phyllanthus Sahni, S., Maurya, S., Singh, U.P., Singh, A.K., Singh, V.P., Pandey, V.B., 2005. Antifungal
amarus. Journal of Natural Remedies 1, 55–59. activity of nor-securinine against some phytopathogenic fungi. Mycobiology 33,
Muthu, C., Ayyanar, M., Raja, N., Ignacimuthu, S., 2006. Medicinal plants used by 97–103.
traditional healers in Kancheepuram District of Tamil Nadu, India. Journal of Samuel, J.K., Andrews, B., 2010. Traditional medicinal plant wealth of Pachalur and
Ethnobiology and Ethnomedicine 2, 1–10. Periyur hamlets Dindigul District, Tamil Nadu. Indian Journal of Traditional
Naaz, F., Javed, S., Abdin, M.Z., 2007. Hepatoprotective effect of ethanolic extract Knowledge 9, 264–270.
of Phyllanthus amarus Schum.et.Thonn. on afltoxin B1 -induced liver damage in Samy, R.P., Pushparaj, P.N., Gopalakrishnakone, P., 2008. A compilation of bioactive
mice. Journal of Ethnopharmacology 113, 503–509. compounds from Ayurveda. Bioinformation 3, 100–110.
Nanden, A.T., 1998. Medicinale planten: tips en simpele recepten voor eengoede Sane, R.T., Chawla, J.L., Kuber, V.V., 1997. Studies on Phyllanthus amarus, Part I. Indian
gezondheid. In: Medicinal Plants and Simple Recipes for a Good Health. Drugs 34, 580–584.
Paramaribo-Suriname, p. 18. Santos, A.R., De Campos, R.O., Miguel, O.G., Cechinel Filho, V., Siani, A.C., Yunes, R.A.,
Narendranathan, M., Remla, A., Mini, P.C., Satheesh, P., 1999. A trial of Phyllanthus Calixto, J.B., 2000. Antinociceptive properties of extracts of new species of plants
amarus in acute viral hepatitis. Tropical Gastroenterology 20, 164–166. of the genus Phyllanthus (Euphorbiaceae). Journal of Ethnopharmacology 72,
Nikam, P.S., Nikam, S.V., Sontakke, A.V., Khanwelkar, C.C., 2011. Role of Phyllanthus 229–238.
amarus treatment in Hepatitis-C. Biomedical Research 22, 319–322. Sedoc, N., 1992. Afro-Surinaamse natuurgeneeswijzen. Bevattende meer dan twee-
Niu, J., Wang, Y., Qiao, M., Gowans, E., Edwards, P., Thyagarajan, S.P., Gust, I., honderd meest gebruikelijke geneeskrachtige kruiden. African–Surinamese
Locarnini, S., 1990. Effect of Phyllanthus amarus on duck hepatitis-B virus repli- natural cures, containing more than two hundred most useful healing/curative
cation in vivo. Journal of Medical Virology 32, 212–218. herbs. 224 p.
Notka, F., Meier, G.R., Wagner, R., 2003. Inhibition of wild type huma immuno- Shanker, K., Singh, M., Srivastava, V., Verma, R.K., Gupta, A.K., Gupta, M.M., 2011.
deficiency virus and reverse transcriptase inhibitor-resistant variants by Simultaneous analysis of six bioactive lignans in Phyllanthus species by reversed
Phyllanthus amarus. Antiviral Research 58, 175–186. phase hyphenated high performance liquid chromatographic technique. Acta
Notka, F., Meier, G.R., Wagner, R., 2004. Concerted inhibitory activities of Phyllanthus Chromatographica 23, 321–337.
amarus on HIV replication in vitro and ex vivo. Journal of Antiviral Research 64, Shanmugam, S., Manikandan, K., Rajendran, K., 2009. Ethnomedicinal survey of
93–102. medicinal plants used for the treatment of diabetes and jaundice among
Obianime, A.W., Uche, F.I., 2009. The Phytochemical constituents and the effects the villagers of Sivagangai District, Tamilnadu. Ethnobotanical Leaflets 13,
of methanol extracts of Phyllanthus amarus leaves (kidney stone plant) on the 189–194.
hormonal parameters of male guinea pigs. Journal of Applied Sciences and Envi- Sharma, A., Singh, R.T., Anand, S., 1993. Estimation of phyllanthin and hypophyl-
ronmental Management 13, 5–9. lanthin by high performance liquid chromatography in Phyllanthus amarus.
Odetola, A.A., Akojenu, S.M., 2000. Anti-diarrhoeal and gastro-intestinal potentials Photochemical Analysis 4, 226–229.
of the aqueous extract of Phyllanthus amarus (Euphorbiaceae). African Journal Shokunbi, O.S., Odetola, A.A., 2008. Gastroprotective and antioxidant activities of
of Medicine and Medical Sciences 29, 119–122. Phyllanthus amarus extracts on absolute ethanol-induced ulcer in albino rats.
Ogunlesi, M., Okiei, W., Osibote, E.A.S., Muotoh, C., 2009. Antimicrobial activity of Journal of Medicinal Plants Research 2, 261–267.
the essential oil and the fractional samples obtained from the leaves and seeds Singh, M., Tiwari, N., Shanker, K., Verma, R.K., Gupta, A.K., Gupta, M.M., 2009. Two
of Phyllanthus amarus (Euphorbiaceae). Research Journal of Phytochemistry 3, new lignans from Phyllanthus amarus. Journal of Asian Natural Products Research
77–84. 11, 562–568.
Okigbo, R.N., Igwe, D.I., 2007. Antimicrobial effects of Piper guineense ‘Uziza’ and Sirajudeen, K.N.S., Sulaiman, S.A., Madhavan, M., Ismail, Z., Swamy, M., Ismail, M.L.,
Phyllanthus amarus ‘Ebe-Benizo’ on Candida albicans and Streptococcus faecalis. Yaacob, M., 2006. Safety evaluation of aqueous extract of leaves of a plant Phyl-
Acta Microbiologica et Immunologica Hungarica 54, 353–366. lanthus amarus, in rat liver. African Journal of Traditional Complementary and
Okoli, R.I., Turay, A.A., Mensah, J.K., Aigbe, A.O., 2009. Phytochemical and antimi- Alternative Medicines 3, 78–93.
crobial properties of four herbs from Edo State, Nigeria. Report and Opinion 1, Sripanidkulchai, B., Tattawasart, U., Laupatarakasem, P., Vinitketkumneun, U.,
67–73. Sripanidkulchai, K., Furihata, C., Matsushima, T., 2002. Antimutagenic and anti-
Oluwafemi, F., Debiri, F., 2008. Antimicrobial effect of Phyllanthus amarus and Par- carcinogenic effects of Phyllanthus amarus. Phytomedicine 9, 26–32.
quetina nigrescens on Salmonella typhi. African Journal of Biomedical Research Srirama, R., Senthilkumar, U., Sreejayan, N., Ravikanth, G., Gurumurthy, B.R., Shiv-
11, 215–219. anna, M.B., Sanjappa, M., Ganeshaiah, K.N., Uma Shaanker, R., 2010. Assessing
Ott, M., Thyagarajan, S.P., Gupta, S., 1997. Phyllanthus amarus suppresses hep- species admixtures in raw drug trade of Phyllanthus, a hepato-protective plant
atitis B virus by interrupting interactions between HBV enhancer I and using molecular tools. Journal of Ethnopharmacology 130, 208–215.
cellular transcription factors. European Journal of Clinical Investigation 27, Srivastava, V., Singh, M., Malasoni, R., Shanker, K., Verma, R.K., Gupta, M.M., Gupta,
908–915. A.K., Khanuja, S.P.S., 2008. Separation and quantification of lignans in Phyllanthus
Pramyothin, P., Ngamtin, C., Poungshompoo, S., Chaichantipyuth, C., 2007. Hepato- species by a simple chiral densitometric method. Journal of Separation Science
protective activity of Phyllanthus amarus Schum & Thonn. extract in ethanol 31, 2338.
treated rats: in vitro and in vivo studies. Journal of Ethnopharmacology 114, Srividya, N., Periwal, S., 1995. Diuretic, hypotensive and hypoglycaemic effect of
169–173. Phyllanthus amarus. Indian Journal of Experimental Biology 33, 861–864.
Prasad, P.R., Reddy, C.S., Raja, S.H., Dutt, C.B.S., 2008. Folklore medicinal plants of Sureban, S.M., Subramaniam, D., Rajendran, P., Ramanujam, R.P., Dieckgraefe, B.K.,
North Andaman Islian, India. Fitoterapia 79, 458–464. Houchen, C.W., Anant, S., 2006. Therapeutic effects of Phyllanthus species:
Rajakumar, N., Shivanna, M.B., 2009. Ethno-medicinal application of plants in the induction of TNF-␣-mediated apoptosis in HepG2 hepatocellular carcinoma
eastern region of Shimoga district, Karnataka, India. Journal of Ethnopharma- cells. American Journal of Pharmacology and Toxicology 1, 65–71.
cology 126, 64–73. Surya Narayanan, B., Latha, P., Rukkumani, R., 2011. Protective effects of Phyl-
Rajeshkumar, N.V., Joy, K.L., Kuttan, G., Ramsewak, R.S., Nair, M.G., Kuttan, R., 2002. lanthus amarus on fibrotic markers during alcohol and polyunsaturated fatty
Antitumor and anticarcinogen activity of Phyllanthus amarus extract. Journal of acid-induced toxicity. Toxicology Mechanisms and Methods 21, 48–52.
Ethnopharmacology 81, 17–22. Tamil, B., Iniyan, G., Dineshkumar, B., Nandhakumar, M., Senthilkumar, M., Mitra,
Rajeshkumar, N.V., Kuttan, R., 2000. Phyllanthus amarus extract administration A., 2010. In vitro study on ␣-amylase inhibitory activity of an Indian medicinal
increases the life span of rats with hepatocellular carcinoma. Journal of plant, Phyllanthus amarus. Indian Journal of Pharmacology 42, 280–282.
Ethnopharmacology 73, 215–219. Thamlikitkul, V., Wasuwat, S., Kanchanapee, P., 1991. Efficacy of Phyllanthus amarus
Rao, M.V., Alice, K.M., 2001. Contraceptive effects of Phyllanthus amarus in female for eradication of hepatitis B virus in chronic carriers. Journal of the Medical
mice. Phytotherapy Research 15, 265–267. Association of Thailand 74, 381–385.
Raphael, K.R., Ajith, T.A., Joseph, S., Kuttan, R., 2002a. Anti-mutagenic activity of Thyagarajan, S.P., Subramanian, S., Thirunalasundari, T., Venkateswaran, P.S., Blum-
Phyllanthus amarus Schum & Thonn. in vitro as well as in vivo. Teratogenesis, berg, B.S., 1988. Effect of Phyllanthus amarus on chronic carriers of hepatitis B
Carcinogenesis and Mutagenesis 22, 285–291. virus. Lancet 2, 764–766.
Raphael, K.R., Khuttan, R., 2003. Inhibition of experimental gastric lesion and Tirimana, A.S.L., 1987. Medicinal Plants of Suriname. Uses and Chemical Con-
inflammation by Phyllanthus amarus extract. Journal of Ethnopharmacology 87, stituents. Chemical Laboratory, Ministry of Agriculture, Animal Husbandry and
193–197. Fisheries, Suriname, 92 p.
Raphael, K.R., Sabu, M.C., Kuttan, R., 2002b. Hypoglycemic effect of methanol extract Titjari, 1985. Famiri-encyclopedia Foe da Natoera Dresi-Fasi. Gezinskruidenboek van
of Phyllanthus amarus Schum & Thonn. on alloxan induced diabetes mellitus in de Natuurgeneeswijzen. Natuurgeneeswijzen uit het zonnige Suriname. Family
rats and its relation with antioxidant potential. Indian Journal of Experimental herb book of natural cures. Natural cures of sunny Suriname, Amsterdam, 419
Biology 40, 905–909. p.
Raphael, K.R., Sabu, M., Kumar, K.H., Kuttan, R., 2006. Inhibition of N-Methyl N - Tjong, A., Young, G., 1989. Het gebruik van medicinale planten door de Javaanse
nitro-N-nitrosoguanidine (MNNG) induced gastric carcinogenesis by Phyllan- bevolkingsgroep in Suriname. The Use of Medicinal Plants by the Javanese Com-
thus amarus extract. Asian Pacific Journal of Cancer Prevention 7, 299–302. munity in Suriname. Instituut voor de Opleiding van Leraren, Paramaribo, p.
Ravikumar, Y.S., Ray, U., Nandhitha, M., Perween, A., Naika, H.R., Khanna, N., Das, S., 196.
2011. Inhibition of hepatitis C virus replication by herbal extract: Phyllanthus Traore, M., Diallo, A., Nikiema, J.B., Tinto, H., Dakuyo, Z.P., Ouedraogo, J.B., Guis-
amarus as potent natural source. Virus Research 158, 89–97. sou, I.P., Guiguemde, T.R., 2008. In Vitro and in vivo antiplasmodial activity of
Author's personal copy

J.R. Patel et al. / Journal of Ethnopharmacology 138 (2011) 286–313 313

‘Saye’, an herbal remedy used in Burkina Faso traditional medicine. Phytother- Van Andel, T., Westers, P., 2010. Why Surinamese migrants in the Netherlands
apy Research 22, 550–551. continue to use medicinal herbs from their home country. Journal of Ethnophar-
Tripathi, A.K., Verma, R.K., Gupta, A.K., Gupta, M.M., Khanuja, S.P.S., 2006. Quanti- macology 127, 694–701.
tative determination of phyllanthin and hypophyllanthin in Phyllanthus species Visweswaram, D., Rao, P.R., Satyanarayana, S., 1994. A noninvasive method for
by high-performance thin layer chromatography. Phytochemical Analysis 17, screening hepatoprotective drugs against carbon tetrachloride-induced hepa-
394–397. totoxicity. Indian Journal of Pharmacology 26, 301–303.
Udayan, P.S., Tushar, K.V., George, S., Balachandran, I., 2007. Ethnomedicinal infor- Wang, X.H., Li, C.Q., Guo, X.B., Fu, L.C., 2001. A comparative study of Phyllanthus
mation from Kattunaykas tribes of Mudumalai Wildlife sanctuary, Nilgiris amarus compound and interferon in the treatment of chronic viral hepati-
district Tamil Nadu, Indian. Journal of Traditional Knowledge 6, 574–578. tis B. Southeast Asian Journal of Tropical Medicine and Public Health 32,
Ueno, H., Horie, S., Nishi, Y., Shogawa, H., Kawasaki, M., Suzuki, S., Hayashi, T., 140–142.
Arisawa, M., Shimizu, M., Yoshizaki, M., Morita, N., Berganza, L.H., Ferro, E., Webster, G.L., 1957. A monographic study of the West Indian species of Phyl-
Basualdo, I., 1988. Chemical and pharmaceutical studies on medicinal plants in lanthus. Journal of the Arnold Arboretum 38, 51–80, pp. 170–178 and 295–
Paraguay. Geraniin, an angiotensinconverting enzyme inhibitor from ‘paraparai 373.
mi’, Phyllanthus niruri. Journal of Natural Products 51, 357–359. Webster, G.L., 1994. Synopsis of the genus and suprageneric taxa of Euphorbiaceae.
Umbare, R.P., Mate, G.S., Jawalkar, D.V., Patil, S.M., Dongare, S.S., 2009. Quality evalu- Annals of the Missouri Botanical Garde 81, 33–144.
ation of Phyllanthus amarus Schum & Thonn. leaves extract for its hypolipidemic Wessels Boer, J.G., Hekking, W.H.A., Schulz, J.P., 1976. Fa joe kan tak’mi no moi.
activity. Biology and Medicine 1, 28–33. Inleiding in de flora en vegetatie van Suriname; Deel I en II. Why do say that I
Unander, D.W., Herbert, H.B., Connete, J.L., Robert, T.M., 1993. Cultivation of Phyl- am not beautiful. Introduction to the Flora and Vegetation of Suriname; Part I
lanthus amarus and evaluation of variable potentially affecting yield and the and II. Natuurgids serie B No. 4, Stinasu, Paramaribo.
inhibition of viral DNA polymerase. Economic Botany 47, 79–88. Wongnawa, M., Thaina, P., Bumrungwong, N., Nitiruangjarat, A., Muso, A., Prasart-
Unander, D.W., Webster, G.L., Blumberg, B.S., 1991. Uses and bioassays in Phyllan- thong, V., 2005. Congress on Medicinal and Aromatic Plants – Vol. 6: Traditional
thus (Euphorbiaceae): a compilation II. The sub genus Phyllanthus. Journal of Medicine and Nutraceuticals. ISHS Acta Horticulturae 680: III WOCMAP.
Ethanopharmacology 34, 97–133. Xia, Y., Luo, H., Liu, J.P., Gluud, C., 2011. Phyllanthus species for chronic hepatitis B
Unander, D.W., Webster, G.L., Blumberg, B.S., 1995. Usage and bioassays in Phyllan- virus infection (Review). The Cochrone Library 4, 1–65.
thus (Euphorbiaceae) IV. Clustering of antiviral uses and other effects. Journal of Yadav, N.P., Pal, A., Shanker, K., Bawankule, D.U., Gupta, A.K., Darokarm, M.P.S.,
Ethnopharmacology 45, 1–18. Khanuja, S.P., 2008. Synergistic effect of silymarin and standardized extract of
Upadhyay, B., Parveen, A.K., Dhaker, A.K., 2010. Ethnomedicinal and ethnopharmaco Phyllanthus amarus against CCl4 -induced hepatotoxicity in Rattus norvegicus.
statistical studies of Eastern Rajasthan, India. Journal of Ethnopharmacology Phytomedicine 15, 1053–1061.
129, 64–86. Yeh, S.F., Hong, C.Y., Huang, Y.L., Liu, T.Y., Choo, K.B., Chou, C.K., 1993. Effect of an
Van Rhede, A., 1690. Horti Malabarici Pars Decima de Herbis et Diversis Warum extract from Phyllanthus amarus on hepatitis B surface antigen gene expression
Specibus, vol. 10. Someren Amsterdem, pp. 29–31. in human hepatoma cells. Antiviral Research 20, 185–192.

Das könnte Ihnen auch gefallen