Equisetum Arvense - Estudo de Revisão

Download as pdf or txt
Download as pdf or txt
You are on page 1of 9

Pharmacogn Rev.

2019;13(26):50-58
A multifaceted peer reviewed journal in the field of Pharmacognosy and Natural Products Review Article
www.phcogrev.com | www.phcog.net

Equisetum arvense: New Evidences Supports Medical


use in Daily Clinic
Danilo Maciel Carneiro1,2, Thiago Veiga Jardim1, Ymara Cássia Luciana Araújo1, Ana Carolina Arantes1,
Andrea Cristina de Sousa1, Weimar Kunz Sebba Barroso1, Ana Luiza Lima Sousa1, Luiz Carlos da Cunha3,*,
Hérica Núbia Cardoso Cirilo3, Maria Teresa Freitas Bara3, Paulo César Brandão Veiga Jardim1,4

ABSTRACT
Relevant aspects to the clinical use of Equisetum arvense L. (common horsetail; EA) were
reviewed and a search was conducted in the databases PubMed, LILACS, SciELO, Virtual
Health Library, Cochrane and Scopus using the keyword “Equisetum arvense” for articles
Danilo Maciel Carneiro1,2, Thiago published from 2013 through 2017. So, eighty-eight articles that addressed the pharmacog-
Veiga Jardim1, Ymara Cássia nostic aspects and in vitro and in vivo biological activity, clinical trials with Equisetum arvense
Luciana Araújo1, Ana Carolina (EA) combined with other plants or alone, case reports concerning possible drug interactions
Arantes1, Andrea Cristina de and review studies were selected. The most relevant reported pharmacological effects in-
Sousa1, Weimar Kunz Sebba cluded antioxidant, anticarcinogenic, antimicrobial, vascular and ileum smooth muscle relax-
ant, analgesic, anti-inflammatory, antinociceptive, cytotoxic, antidiabetic, hepatoprotective,
Barroso1, Ana Luiza Lima Sousa1, antilithiatic, dermatologic, wound healing, antileishmanial, diuretic, immunizing, platelet ag-
Luiz Carlos da Cunha3,*, Hérica gregation inhibitory, osteoblast response-promoting, remineralizing, anxiolytic, sedative, anti-
Núbia Cardoso Cirilo3, Maria convulsant and cognitive performance-stimulating activities. Five clinical trials demonstrated
Teresa Freitas Bara3, Paulo César the efficacy of phytotherapeutic complexes containing EA, with three for treating benign
Brandão Veiga Jardim1,4 prostate hyperplasia, one for chronic musculoskeletal pain and one describing the effects of a
topical preparation on brittle nail syndrome. Three clinical trials tested EA alone; one analyzed
1
Hypertension League, School of its pharmacokinetics; another addressed the wound-healing effect of 3% EA ointment and a
Medicine, Federal University of Goiás randomized double-blind clinical trial found that the diuretic effect of EA was superior com-
(UFG). Goiânia - Goiás, BRAZIL. pared to the negative control and equivalent to treatment with hydrochlorothiazide (25 mg/
2
Reference Center in Integrative and day) without changes in electrolyte excretion. Considering its long history of traditional use in
Complementary Medicine of the State several countries, in vivo and in vitro research and more recent clinical studies, EA meets the
Health Department of Goiás, Goiânia – requisites for having well-defined medical applications with proven efficacy and acceptable
Goiás, BRAZIL. safety.
3
NEPET-UFG – Toxicopharmacological Key words: Common horsetail, Diuretic, Equisetum arvense, Medicinal plants, Phytotherapy.
Studies and Research Group (School
of Pharmacy-UFG). Goiânia – Goiás,
BRAZIL.
4
School of Medicine of UFG. FM/HC/UFG INTRODUCTION
Hypertension League. Goiânia – Goiás, Surveillance Agency (ANVISA, for its initials in
Equisetum arvense L. (EA) is in the family Equisetaceae.
BRAZIL. Portuguese) addresses the reporting of drugs of plant
It is an herbaceous perennial plant native to the
Correspondence origin and regulates the commercialization of EA as
northern hemisphere that has long been used for
Prof. Luiz Carlos da Cunha a plant-derived drug indicated for “swelling (edema)
medicinal purposes. The various species in this family
due to fluid retention” in the form of an infusion or
NEPET-UFG – Toxicopharmacological are widely distributed all across Canada, the United
Studies and Research Group (School a decoction.[6] A dry EA extract-based pharmaceutical
States (except for the southeast), Europe, Africa and
of Pharmacy-UFG). Goiânia – Goiás, preparation has been registered by ANVISA as a
southern Asia, including Turkey, Iran, the Himalayas,
BRAZIL. phytotherapeutic drug.[7]
China (except for the southeast), Korea and Japan.[1,2]
Phone no : +55 62 32096329 According to the European Medicines Agency
EA is not native to Brazil, where common horsetail is
E-mail: [email protected] (EMA), the Evaluation of Medicines for Human Use
popularly known as “cavalinha,” and it was brought
Committee on Herbal Medicinal Products reported
DOI : 10.5530/phrev.2019.2.4 to the country to be grown for medicinal use.[3,4]
that the clinical data for the absorption, distribution
Although the genus Equisetum is not yet included in
and pharmacokinetics of EA are still insufficient.[2]
Article Available online the Brazilian pharmacopoeia, several species, including
http://www.phcogrev.com/v13/i26
EA, are described in other official or non-official The aim of the present study was to review the
pharmacopoeias.[4] EA is listed in the Phytotherapeutic literature on EA (common horsetail) to serve as a
Copyright
© 2019 Phcog.Net. This is an open- Memento of the 1st edition of Brazilian Pharmacopoeia reference for phytotherapy researchers and legislators
access article distributed under the terms and also in the Brazilian National List of Medicinal interested in data on this important medicinal plant.
of the Creative Commons Attribution 4.0 Plants of Interest to the Unified Health System
International license.
(RENISUS, for its initials in Portuguese) as published
MATERIALS AND METHODS
by the Ministry of Health.[5] The Collegiate Board A literature search was performed in the databases
Regulation (RDC) nº 10 (2010) of the National Health PubMed, LILACS (Latin American and Carib-

Cite this article: Carneiro DM, Jardim TV, Araújo YCL, Arantes AC, Sousa AC, et al. Equisetum
arvense: New Evidences Supports Medical use in Daily Clinic. Pharmacog Rev. 2019;13(26):50-8.

50  Pharmacognosy Reviews, Vol 13, Issue 26, Jul-Dec, 2019


Carneiro, et al.: Equisetum arvense: New Medical Evidences

bean Health Sciences Literature), SciELO (Scientific Electronic Library The name Equisetum derives from Latin (equi = horse and setum = tail)
Online), Virtual Health Library (BVS, for its initials in Portuguese), and may be explained by the botanical description of the plant. The
Cochrane and Scopus for the period from July 2013 to August 2017 using leaf-free stems resemble asparaguses and when desiccated, they become
the keywords “Equisetum arvense, phytotherapy, medicinal plants, com- hollow, rough and strong; their many fine branches with small, scale-like
mon horsetail and diuretic”. Articles published in Portuguese, English, leaves resemble a horsetail (Figure 2).[9] The term “arvense” in turn comes
French or Spanish that addressed pharmacognostic, pharmacological, from the overall tree-like appearance of the plant. EA is a perennial plant
pharmacokinetic or in vivo toxicological aspects or those that reported with a peculiar appearance that reaches up to 20 to 65 cm high. It is
clinical trials with EA or its extracts as a plant-derived medicine as the characterized by primary branched aerial stems with whorls.[10] The
primary subject or together with other species were considered. All the stems have two types, sporiferous (fertile) and sterile. Fertile stems are
available articles were evaluated and the ones that analyzed issues related simple and have a reddish hue, loose brown sheaths and an oblong spike
to the clinical use of EA were selected for review. Studies on agronomic that disappears in the summer. The reproductive structure, which is
aspects or those that were focused on the analysis of the chemical struc- known as a strobilus and is where spore production occurs, is located on
ture of substances isolated from EA were excluded. the terminal end and the branches. Sterile stems are green, furrowed and
hollow; the branches are thin and arranged in pairs. These simple, light
RESULTS green branches exhibit four angles and they are rough and articulate. The
deep rhizome reaches down to 2 meters deep.[10,11]
The literature search retrieved 181 articles. Eighty-six articles that
The parts used for medicinal purposes are the shoots, which are primarily
analyzed several aspects of EA were included for systematic review, in
desiccated sterile stems.[2,12]
addition to the ANVISA and EMA documents. The articles were
categorized according to their primary topics as follows: pharmacognostic The sterile stems are composed of the cortex, parenchyma, stomata and
aspects (8), in vitro (36) and in vivo (28) biological activity, clinical trials silica granules. The parameters that characterize the plant-derived drug
of EA combined with other plants (6) or alone (3), case reports relative to are its water content (15.45%), total ash (22%), acid insoluble ash (11%),
possible drug interactions (1) and review studies (5) (Figure 1). water soluble ash (8%) w/w and foaming index (100).[12]

Pharmacognostic aspects Chemical components


Equisetum arvense L. belongs to the family Equisetaceae, genus A broad review study reported the presence of alkaloids, carbohydrates,
proteins, amino acids, phytosterols, saponins, sterols, ascorbic acid,
Equisetum L. and subgenus Equisetum. The genus Equisetum, which
silicic acid, phenol, tannin, flavonoids, triterpenoids and volatile oils,
belongs to the phylum Sphenophyta, appeared at the end of the
among many other biologically active components.[13] EA is quite rich
Paleozoic Era, approximately 300 million years ago. It appears that this is
in minerals and it stands out for its silicon (Si) content, especially in the
the single extant genus from class Equisetopsida and it is considered the
oldest extant genus on Earth. It comprises more than 30 species, most of
which are small plants that ordinarily do not reach one meter high. The
fact that these plants are uniformly distributed across the world might
be due to having appeared prior to the continental drift, as indicated by
the dating of fossil records, which might explain their easy adaptation
to the most disparate climates. They are considered the terrestrial plants
with the highest silica content, which might account for their many
medical applications.[4,8]

Figure 2: Illustration of the botanical characteristics of Equisetum


arvense.
Figure 1: Flowchart representing the search of articles for review. Source: Tropicos.org[9]

Pharmacognosy Reviews, Vol 13, Issue 26, Jul-Dec, 2019 51


Carneiro, et al.: Equisetum arvense: New Medical Evidences

form of SiO2 (5% to 10%) and a small portion as water-soluble silicates.[2,14] Table 1: List of phytochemicals present in E. arvense*.
Si is known to be beneficial for plants; its absorption by transporters Sn Groups Compounds
has a protective effect against biotic and abiotic stresses. EA is a valuable No.
model for studying the absorption and deposition of this important 1 Flavonoids Kaempferol-3-O-sophoroside-7-O-glucoside,
mineral.[15,16] Phenolic acids, such as di-E-caffeoyl-meso-tartaric acid kaempferol-3-O-(6’’-O malonylglucoside)-7-
(0.008%) and methyl esters of protocatechuic and caffeic acid were O-glucoside, kaempferol-3-O-sophoroside,
detected, in addition to silicic acids, silicates, polyenoic acids, dicar- quercetin-3-O-glucoside, apigenin, apigenin-5-
boxylic acids and styryl-pyrone.[12] Potassium, calcium and phosphorus O-glucoside, luteolin, luteolin-5-O-glucoside,
genkwanin-5-O-glucoside and isoquercitrin
ions were identified, as were sodium, magnesium, zinc, aluminum and
manganese, albeit in lower proportions.[12] Flavonoids including luteolin, 2 Triterpenoids Isobauerenol, taraxerol, germanicol, ursolic acid,
oleanolic acid and betulinic acid
quercetin, apigenin, isorhamnetin and kaempferol[17] were found at
percentages ranging from 0.2% to 0.09%; quercetin and kaempferol 3 Phenolic Equisetumoside A, equisetumoside B,
Glycosides equisetumoside C, onitin and onitin-9-O-
primarily appear as glucosides (quercetin 3-glucoside and its malonyl
glucoside
esters).[12] The steroid fraction is essentially composed of beta-sitosterol
(60%), campesterol (32,9%), isofucosterol (5.9%) and cholesterol (traces).[18] 4 Styrylpyrone Equisetumpyrone, 3’-deoxyequisetumpyrone
and 4’-O-methylequisetumpyrone
Isoprenoid flavor precursors, i.e., 3-oxo-alpha-ionol (spicy odor) and
(E,E)-pseudoionone (balsamic odor) as well as odorous benzenic deriva- 5 Alkaloids Nicotine, palustrine and palustrinine
tives, i.e., phenylethanal (hyacinth and lilac notes) and 2-phenylethanol 6 Phytosterols Cholesterol, epicholestanol,
(rose odor) contribute to the odor of EA(Fons, 2013 #30;Fons, 24-methylenecholesterol, isofucosterol,
campesterol and β-Sitosterol
2013 #30).[19] Traces of alkaloids (nicotine, palustrine and palustrinine)
were detected.[12,20] 7 Minerals Silicic acid, silicates, potassium, calcium,
aluminium, sulphur, magnesium and manganese
A complex that was initially categorized as having a saponin nature
(equisetonine) was later identified as a sugar and flavonoid mixture.[2] *Source: Badole and Kotwal, 2014.[43]
With regards to the dry weight of the EA extract, a study published in
2013 reported a total phenol content of 18.67%, 125 mg gallic acid/g, a
Table 2: Summary of the in vitro pharmacological activities of EA.
total antioxidant capacity of 1608 µM TEAC/mg and 0.0049 mg silicic
Activities References
acid/mg.[21]
Antioxidant Myagmar and Aniya, 2000;[48] Nagai et al. 2005;[49]
Considering the significant growth of the global market for phytothera-
Štajner et al. 2006;[50] Milovanović et al. 2007;[51]
peutic products in recent years, their regulation is crucial from a public Mimica-Dukic et al. 2008;[52] Da Silva and Do Carmo,
health viewpoint. EA is used in countless plant-based products, but it 2009;[53] Štajner et al. 2009;[54] Četojević-Simin et al.
might be adulterated with the closely related species E. palustre L., which 2010;[55] Huh and Han, 2015[56]
might contain toxic alkaloids. Because morphology-based identification is Antimicrobial Husson et al. 1986;[57] Suganda et al. 1983;[58] Guerin
often difficult, if not impossible, molecular techniques might be useful for and Reveillere, 1984;[59] Heisey and Gorham, 1992;[60]
the certification of processed materials. A study analyzed two molecular Radulović et al. 2006;[61] Milovanović et al. 2007;[51]
identification techniques as methods for testing the purity of these Brune et al. 2008;[25] Geetha et al. 2011;[62] Pereira et al.
products. One method is based on thin-layer chromatography (TLC), 2012;[63] Wojnicz et al. 2012;[64] De Oliveira et al. 2013;[65]
in accordance with European Pharmacopoeia and the other is based Garcia et al. 2013[66]
on DNA barcoding, which has been used in recent years to identify Anti-platelet Mekhfi et al. 2004;[67] Goun et al. 2002[68]
materials in plant-derived products. Although TLC is more cost- and aggregation
time-effective, DNA barcoding is more powerful for determining the Cytotoxic and Yoshinobu, 1992;[69] Goun et al. 2002;[68] Alexandru et al.
identity of adulterant species.[22] Regarding quality control, Gallo et al. anticarcinogenic 2007;[70] Četojević-Simin et al. 2010;[55] Al Mohammed
et al. 2017[71]
applied two chromatography methods (HPTLC and HPLC) to finger-
print EA and related species. For EA, HPLC might be useful for identi- Vasorelaxant Sakurai et al. 2003[72]
fying and evaluating the quality of plant materials derived from related Hepatoprotective Oh et al. 2004[73]
species and to investigate the presence of individual components.[23] The Remineralization Ferraz et al. 2008;[74] Raczuk et al. 2008;[75] Bye et al.
specific markers for EA identified by TLC were rutin, chlorogenic acid, 2010;[76] Law and Exley, 2011;[77] Pereira et al. 2012[63]
kaempferol and caffeic acid.[24] Brune et al. described the applicability Anti-inflammatory Gründemann et al. 2014;[30] Saeed et al. 2014[78]
of “inter-simple sequence repeats” (ISSR)-PCR for the differentiation
Antileishmanial Saeed et al. 2014[78]
of Equisetum species with a special focus on the detection of hybrids.
According to these authors, the ISSR banding patterns were highly typical
for each of the most common species in the Equisetum genus.[25] Table 1 In vivo pharmacological activities
shows de list of phytochemicals present in E. arvense. Literature evidences on anxiolytic, antidiabetic, analgesic and anti-
In vitro pharmacological activities inflammatory, benign prostate hyperplasia, wound healing, remineral-
izing, antilithiatic, bladder myorelaxant and diuretic in vivo activities of
References on the antioxidant, anticarcinogenic, antimicrobial, vascular
EA are summarized in Table 3.
and ileum smooth muscle relaxant, anticonvulsant, sedative, anxiolytic,
dermatologic, immune, antinociceptive, anti-inflammatory, antidiabetic, Review studies
antileishmanial, diuretic, platelet aggregation inhibitory and osteoblast References and scopes of review studies on EA found in the literature are
response-promoting activities of EA are shown in Table 2. shown in Table 4.

52 Pharmacognosy Reviews, Vol 13, Issue 26, Jul-Dec, 2019


Carneiro, et al.: Equisetum arvense: New Medical Evidences

Table 3: In vivo activities of EA. d) Tincture (1:5) in 96% hydroethanolic solution (v/v): 30 to 40 drops
Activities References e) Dry extract (4-7:1) via aqueous extraction: 185 mg
Anxiolytic Dos Santos Junior et al. 2005;[42] Rezaie et al. 2011;[20] f) Dry extract (7.5-10.5:1) via extraction with 70% hydroethanolic solution
Singh et al. 2011;[79] Sarris et al. 2013[80] (v/v): 200-225 mg.[2]
Antidiabetic Safiyeh et al. 2007;[81] Soleimani et al. 2007[82] The average dose is three single doses per day; the maximum daily dose
Analgesic and anti- Monte et al. 2004;[83] Sandhu et al. 2010[12] is four single doses.[2]
inflammatory Uses based on folk medicine
Benign prostate Oka et al. 2007;[34] Tamaki et al. 2008[36] For generations, EA has been used in folk medicine for the treatment
hyperplasia of several conditions, including tuberculosis, kidney and bladder infec-
Wound healing Ozay et al. 2010;[84] Hayat et al. 2011;[85] Ozay et al. tions; as a hemostatic for profuse menstruation, nose, lung and gastric
2013[86] bleeding; rheumatic conditions; gout; wounds and ulcers; swelling, bone
Remineralizing Kotwal and Badole, 2016[29] fractures and burns; brittle nails; and hair loss.[1] EA is traditionally
Antilithiatic Grases et al. 1994;[87] Crescenti et al. 2015[88] indicated primarily as a mild diuretic, for swelling, inflammation and
remineralization.[2,29] There is a long tradition of EA use in Europe for
Bladder myorelaxant Zhang et al. 2015[89]
treating inflammatory problems.[30]
Diuretic Rebuelta et al. 1978;[90] Franck Bakke and Hillestad,
1980[91] Uses based on traditional medicine
In Ayurveda, EA is traditionally used for treating prostate inflammation
and hypertrophy, urinary incontinence and nocturnal enuresis in
Table 4: Summary of the review studies activities of EA. children.[31] Nicholas Culpepper, a 19th century English botanist, used EA
juice or decoctions to make bleeding stop as well as for treating skin
Scope of review References
ulcers, wounds and inflammation, kidney stones and cystitis.[14]
Systematic review on EA Mello and Budel, 2013[4]
Uses based on preclinical studies
Pharmacological aspects of EA Al-Snafi, 2017[13]
In many countries, the corresponding legislation is based on the long
Ethanopharmacological and Phytochemical Badole and Kotwal, 2014[43]
history of use in traditional medicine, ethnopharmacological studies,
review on EA with reference to osteoporosis
phytochemistry research and pharmacological studies.[14]
Pharmacology and phytochemistry of EA Sandhu et al. 2010[12]
A report published by the EMA Committee on Herbal Medicinal Products
(CHMP)[2] lends support to the use of EA by oral route to promote the
kidney excretory function, the treatment of post-traumatic and stasis
Case Report edema, irrigation therapy in bacterial and low urinary tract inflamma-
A recent study described a possible interaction between EA and antiret- tory diseases and kidney and bladder stones. The dose recommended
roviral therapy in two HIV-positive patients. This interaction might have by EMA is 220 to 225 mg of dry extract 3 or 4 times per day.[2] The
triggered a virological breakthrough in the patients, who maintained the maximum daily dose used in clinical studies was 900 mg of dry extract.[32]
same regimen for many years that included lamivudine, zidovudine, German Commission E approved the use of EA as an herbal medicine
efavirenz, emtricitabine and tenofovir. Given the scarce data on the (in the form of powder or tea) at an average daily dose of 3 to 6 g or
pharmacological properties of EA regarding potential drug-herb interac- the equivalent amount of other preparations in 2 or 3 daily intakes for
tions, the authors concluded that these interactions might occur when treating post-traumatic or stasis swelling and as a diuretic in cases of
these agents are taken simultaneously and thus advise clinicians to avoid bacterial and inflammatory lower urinary tract diseases with the presence
this combination until further data become available.[26] of urinary sediment. EA was also approved for topical use as a decoction
(50 g/L) in compresses or baths or as a liquid extract at a dose of 50 drops
Summary for medical use in daily clinic diluted in water as an adjuvant for treating hard-to-heal wounds.[2,14,33]
Dosage form In Brazil, ANVISA’s RDC no. 10 recommends using EA in the form of
According to the Phytotherapeutic Memento in the 1st edition of an infusion or a decoction at 3 g/150 mL of water, 2 to 4 times per day to
Brazilian Pharmacopoeia, the primary pharmaceutical preparations are treat edema (swelling) due to fluid retention.[6]
as follows:[27] Uses based on clinical studies
–  Tincture: (1:4-5) in 31.5% hydroethanolic solution (w/w) Three studies evaluated the usefulness of Eviprostat®, a commercial
–  Tincture: (1:4-5) in 96% hydroethanolic solution (v/v) preparation containing Chimaphila umbellata, Populus tremula, Pulsatilla
– Capsules and tablets containing dry extract (4-7:1) via aqueous pratensis and EA extracts and wheat germ oil for treating Benign Prostate
extraction Hyperplasia (BPH). The effects of each individual component on the
Reactive Oxygen Species (ROS), namely superoxide anion (O2-) and
– Capsules and tablets containing dry extract (7.5-10.5:1) via extraction
hydroxyl radical (OH*), were assessed. The results suggest that ROS
with 70% hydroethanolic solution (v/v)[1]
suppression might partially contribute to the anti-inflammatory action
–  Medicinal tea (infusion) of Eviprostat®, which might be involved in its therapeutic effect in BPH.
Dose
[34]
The clinical efficacy of Eviprostat® was assessed in two studies. One
Single doses by oral route are used by adults as follows:[27,28] was an open-label multicenter clinical trial in which 100 patients with
BPH took 2 tablets of Eviprostat® 3 times per day over 12 weeks. The
a) Infusion of leaves or shoots: 2-3 g in 250 mL of boiling water (teacup)
primary parameters for efficacy were the International Prostate
b) Coarsely fragmented plant matter: 570 mg Symptom Score (IPSS), maximum urinary flow rate (Qmax), Residual
c) Tincture (1:4-5) in 31.5% hydroethanolic solution (w/w): 20 drops Urine (Ru) and prostate Volume (V). All the parameters exhibited

Pharmacognosy Reviews, Vol 13, Issue 26, Jul-Dec, 2019 53


Carneiro, et al.: Equisetum arvense: New Medical Evidences

significant improvement. The rate of clinical adverse events was 1%.[35] part in the study. At week 8, urinary day frequency was significantly
The other study compared the usefulness of Eviprostat® and EVI-F, a new lower in response to treatment compared to placebo. Similarly, episodes
formulation of Eviprostat® containing two times more active ingredients. of nocturia were significantly fewer after 8 weeks of treatment versus
Patients with BPH were randomly allocated to the Eviprostat® group placebo. Symptoms of urgency and total incontinence were also lower
(6 tablets/day) or the EVI-F group (3 tablets/day). The clinical efficacy of (all p<0.01) in the treatment group. Significant improvements in quality
these two drugs was evaluated by means of the IPSS and the Quality of of life were reported after treatment in comparison to placebo. No
Life (QOL) score and their safety was evaluated based on the incidence significant side effects were observed resulting in withdrawal from
of side effects. Based on guidelines for clinical studies on dysuria, the treatment.[40]
changes in the total IPSS and the QOL score were comparable to the In filling a considerable gap in the clinical studies on the diuretic effect
previously reported data for classical BPH treatment agents and these of EA, a double-blind, randomized clinical trial randomly distributed
parameters showed gradual improvements over the course of the treat- 36 healthy male volunteers into 6 groups (n=6) that underwent a three-
ment period. Both treatments were well tolerated. The clinical usefulness step intervention. EA dry extract (900 mg/day), placebo (900 mg/day)
of monotherapy with EVI-F or Eviprostat® was reasonably demonstrated or hydrochlorothiazide (25 mg/day) were alternately administered for
in this study. Furthermore, both treatments reduced nocturia, which has 4 consecutive days and separated by a 10-day washout period. The
an impact on the QOL of patients with BPH.[36] diuretic effect was assessed based on the 24-h water balance and serum
A clinical trial assessed the efficacy of EA, Urtica dioica, Boswellia serrata, electrolytes. Each volunteer served as his own control and the results
Allium sativum and Apium graveolens for treating chronic musculoskeletal were compared between the 3 groups. EA exhibited a diuretic effect that
pain. Combined with Vitamin B1, a mixture of the five plants promoted was stronger than that of the negative control and was equivalent to that
a significant reduction in the score on the pain scale and significantly of hydrochlorothiazide (25 mg) without causing significant changes in
improved the functional mobility of body sites affected by chronic joint, the excretion of electrolytes. There was no significant increase in the
back and muscle pain. No adverse effect was reported.[37] urinary elimination of the catabolites. Rare minor adverse events were
The efficacy of topical EA ointment application was tested for wound reported. All the laboratory parameters remained within the corresponding
healing and the reduction of inflammation and pain relief after episi- normal range. The clinical, electrocardiographic and laboratory data
otomies in nulliparous mothers. A double-blind clinical trial analyzed did not change before and after the experiment, which suggests that the
54 women in the EA group and 54 women in the placebo group. Five drug is safe for acute use. Further research is needed to better clarify the
and 10 days after childbirth, the primary outcomes (wound healing and mechanism of the diuretic action and the other possible pharmacological
pain intensity) were assessed based on the Redness, Edema, Ecchymosis, actions of this phytomedicine.[32]
Discharge and Approximation of the edges (REEDA) scale and a Visual
Analog Scale (VAS). In addition, the number of painkillers used and Toxicity, contraindications, adverse effects and interactions
adverse events experienced during the 10-day period of the study were In assessment of acute hepatotoxicity in rats, no anatomical pathology
analyzed. The mean scores were significantly lower in the treated group changes were found in the liver tissue or in the liver enzymes.[3]
compared to the controls at both evaluation times. The difference in the No toxic effect was detected with respect to clinical, hematological, urinary
pain score was significantly lower in the treated group and the mean or serum biochemical parameters, body weight or internal organ weight
number of acetaminophen tablets used over the 10-day period was higher when 0.03% to 3% EA hydroalcoholic extract was added to the food
for the control group. The 3% EA ointment promoted wound healing and given to male and female rats for 13 days.[41]
relieved pain during the 10-day period after the episiotomy.[22] A hydroalcoholic EA extract in doses of 2 and 5 g/Kg (IP) was associated
To assess the topical effect of EA on brittle nails, a clinical, blind, cross- with 12% and 37.5% mortality in rats, respectively. As the LD50 was
over and controlled study randomly allocated 38 female patients to two > 5 g/Kg, the extract was rated practically non-toxic.[42]
groups. The groups alternately received treatment with a cosmetic nail EA is contraindicated during pregnancy and breastfeeding as well as for
polish that contained EA, methyl sulfonyl methane and Hydroxypropyl children under 12 years old due to a lack of specific studies[2] and the
Chitosan (HPCH) that was indicated for treating brittle nail syndrome. presence of nicotine.[43]
The tested preparation was more effective at controlling the signs of the
Individuals with kidney or heart failure should avoid EA because its
syndrome compared to not using it.[38]
diuretic effect might cause potassium loss, which might particularly
The effect of a water-soluble nail polish containing EA on nail psoriasis affect patients with heart failure who were treated with digitalis medicines
was tested. A randomized, double-blind, placebo-controlled, parallel- or other drugs that reduce serum potassium.[6]
group trial was performed to evaluate the efficacy and tolerability of a
According to considerations based on pharmacological inferences, EA
nail polish containing HPCH, EA and methyl sulfonyl methane on nail
might cause a Vitamin B1 (thiamine) deficiency due to the presence of
psoriasis. The test product or a placebo was applied once daily for 24 weeks.
the enzyme thiaminase.[44]
The clinical cure rate showed the statistically significant superiority of the
tested nail polish compared to the placebo (Fisher’s exact test, P=0.0445). Luengo does not recommend EA for patients with active gastroduodenal
This superiority was already present after 16 weeks of treatment. In ulcers because the presence of tannins and silicic salts and acids might
addition, an analysis of the Nail Psoriasis Severity Index showed statis- irritate the gastric mucosa.[45]
tically significant clinical improvement after 12 weeks of treatment in Rare cases of allergies might develop among patients sensitive to EA’s
comparison to the results obtained after 8 weeks.[39] chemical components. Prolonged use and abuse might cause exudative
The efficacy of Urox®, a proprietary combination of phytomedicine edema, dysphagia, headache, tenesmus and loss of appetite and when
extracts including, Crataeva nurvala stem bark, Equisetem arvense stem high doses are used, gastric and urinary irritation may occur due to the
and Lindera aggregata root, in reducing a variety of bladder symptoms, presence of nicotine.[6]
compared to an identical placebo, was documented in a randomised, Pharmacological interactions with lithium and digitalis medicines have
double-blind, placebo-controlled trial. Data were collected at baseline, been described.[14] Since a possible interaction between EA and anti­
2, 4 and 8 weeks, with the primary outcome being self-reported urinary retroviral therapy might have triggered a virological breakthrough in
frequency. One hundred and fifty participants (59% female, aged) took two patients and given the scarce data on the pharmacological properties

54 Pharmacognosy Reviews, Vol 13, Issue 26, Jul-Dec, 2019


Carneiro, et al.: Equisetum arvense: New Medical Evidences

of EA regarding potential drug-herb interactions, clinicians should avoid incontinence were lower in the treatment group. EA is a genito-urinary
this combination until further data become available.[26] astringent for urinary incontinence and enuresis in children. Its silica
Although nicotine in high doses might cause tolerance and dependence, content likely contributes to this effect, besides its anti-inflammatory,
it might aid in smoking cessation in low doses.[46] anti-bacterial and anti-lithogenic effects.[40]
In a randomized, double-blind clinical trial, the diuretic effect of EA was
Precautions during the use of Equisetum arvense
superior compared to that of the negative control and equivalent to that
Individuals taking EA are recommended to take Vitamin B complex
of hydrochlorothiazide (25 mg), without significant changes in electrolyte
supplements. Chronic alcoholics should avoid EA due to a propensity
excretion, signs of toxicity or significant adverse effects, suggesting that
for Vitamin B1 deficiency. The concomitant use of nicotine-containing
EA is an effective diuretic and safe for acute use.[32] The flavonoids and
substances, diuretics or lithium medicines should be avoided. Supervision
the high potassium content may contribute to this effects.[2]
by a healthcare specialist is recommended while taking EA.[43]
A topical EA 3% ointment application promoted wound healing and
Absorption, distribution, metabolism, elimination relieved pain during the 10-day period after the episiotomy.[22] The
A clinical study on EA alone analyzed the excretion of urinary metabolites authors of the studies discussed that the positive effects on wound
of flavonoids and hydroxycinnamic acids, which are polyphenolic contraction may have resulted from silicea, silicic acid, silicon and
compounds present in our daily diet in the form of tea and vegetables saponin content in the extracts. The results support the traditional topical
as well as in herbal remedies used in phytomedicines. In vivo data on use of the water decoct in wound healing.[2]
absorption, bioavailability and metabolism after the oral intake of these The topical effect of EA on brittle nails was more effective at controlling
compounds are scarce and contradictory. To examine their metabolism the signs of the syndrome compared to not using it.[38] In another clinical
and renal excretion, a standardized EA extract was administered to trial, the effect of a water-soluble nail polish containing EA on nail
11 volunteers following a flavonoid-free diet for 8 days. Twenty-four-h psoriasis showed superiority, when compared to the placebo.[39] EA is a
urine samples were collected and analyzed by HPLC-diode array detection source of organic silicon and since nails contain 16 mg of silicon dioxide
(DAD). The putative quercetin metabolites (3,4-dihydroxyphenylacetic
per 100 g, it seems to contribute to their strength and hardness while
acid or 3,4-dihydroxytoluene) were not detected in any of the urine
maintaining stability among keratin fibrils.[38]
samples. The endogenous amount of homovanillic acid, which is
generally regarded as one of the primary quercetin metabolites, did Recent clinical evidences and the testing of preparations and products
not increase significantly. However, hippuric acid (the glycine conjugate containing EA for therapeutic purposes at a large scale are paving the
of benzoic acid) increased twofold after drug intake. Thus, the degrada- way for new research (including phase IV and drug surveillance studies).
tion to benzoic acid derivatives rather than phenylacetic acid derivatives The scientific studies that support the validation of EA by EMA,
seems to be a predominant metabolic route.[47] Commission E and the Brazilian Ministry of Health are based on its
traditional use, preclinical pharmacological studies and more recently,
DISCUSSION AND CONCLUSION on clinical studies. The applied criteria also serve to assess the toxicity
EA is a medicinal plant that has been used for millennia in the traditional and safety of the use of this plant; however, they do not rule out the
medicine of India, China and Greece and in the folk medicine of many possibility of still unknown toxicity-related factors. For this reason,
countries. The traditional knowledge about EA and scientific studies EA should not be used by children under 12 years old or by pregnant
conducted since the beginning of the 20th century led to its approval as a or breastfeeding women. Aqueous preparations should be avoided by
medicine in several countries, including Brazil. patients with health problems that demand fluid restriction (heart or
kidney failure).
In this systematic review, unlikely other review studies, we have compiled
all the clinical studies that include EA alone or in associations, with the The final report of the EMA/CHMP concludes that the toxicology
aim of supporting its safe clinical use in various situations at daily medical data on EA are still insufficient and thus the committee-maintained EA’s
clinic. In addition, we showed its main pharmacological actions and classification as a traditional medicinal product in Europe. However,
indicate the safe doses, along with the care that should be taken regarding considerable advances in research indicate that the status of EA as a
toxicity issues, adverse effects and medicinal interactions. Three studies phytotherapeutic agent is becoming more likely.
showed the usefulness of a preparation containing Chimaphila umbellata, Due to its age-long and almost cosmopolite use and the available studies
Populus tremula, Pulsatilla pratensis and EA  extracts and wheat germ in addition to its use as a traditional and folk medicinal agent, EA is
oil for treating benign prostate hyperplasia.[34-36] The results suggest that emerging as a phytotherapeutic drug that is potentially useful in several
ROS suppression might partially contribute to the anti-inflammatory medical fields. For this reason, new studies on its pharmacokinetics,
action. The flavonoid isoquercitrin (quercetin-3-O-glucoside) present pharmacodynamics and toxicology are needed, as is evidence on its
in the extracts of EA may interfere with the polyfunctionality of immu- therapeutic effects, with special emphasis on its potential as an effective
nocompetent cells, thereby providing an anti-inflammatory mode-of- and safe diuretic.
action.[30] One might thus conclude that as a function of its long history of tradi-
A clinical trial demonstrated the efficacy of EA, Urtica dioica, Boswellia tional use in several countries, in vivo and in vitro research and more
serrata, Allium sativum and Apium graveolens for treating chronic recent clinical studies, EA meets the requisites for well-defined medical
musculoskeletal pain, combined with Vitamin B1. The authors claims application with proven efficacy and acceptable safety. However, large-
that EA contains β-sitosterol, campesterol and isofucosterol, which scale clinical trials are needed to confirm this hypothesis.
provide anti-inflammatory steroidal effects and, thus, may explain the
success of the therapeutic effect.[37]
The efficacy a combination of phytomedicine extracts including
ACKNOWLEDGEMENT
Crataeva nurvala, EA and Lindera aggregata in reducing a variety of The authors are grateful for financial support provided by Fundação de
bladder symptoms, was documented in a randomised, double-blind, Amparo à Pesquisa do Estado de Goiás (FAPEG), a public foundation to
placebo-controlled trial. Symptoms of nocturia, urgency and total support research in the state of Goias, Brazil.

Pharmacognosy Reviews, Vol 13, Issue 26, Jul-Dec, 2019 55


Carneiro, et al.: Equisetum arvense: New Medical Evidences

CONFLICT OF INTEREST 18.  D’Agostino M, Dini A, Pizza C, Senatore F, Aquino R. Sterols from Equisetum
arvense. Boll Soc Ital Biol Sper. 1984;60(12):2241-5.
Due to the strictly academic nature of this publicly funded scientific 19.  Fons F, Froissard D, Bessiere JM, Fruchier A, Buatois B, Rapior S. Volatile
composition of six horsetails: Prospects and perspectives. Nat Prod Commun.
review, the authors declare no conflicts of interest. 2013;8(4):509-12.
20.  Rezaie A, Jafari B, Mousavi G, Nazeri M, Ebadi A, Ahmadeh C, et al. Comparative
ABBREVIATIONS study of sedative, pre-anesthetic and anti-anxiety effect of Origanum majorana
extract with diazepam on rats. Res J Biol Sci. 2011;6:611-4.
ANVISA: National Health Surveillance Agency of Brasil; BPH: Benign 21.  Uslu ME, Erdogan I, Bayraktar O, Ates M. Optimization of extraction conditions
Prostate Hyperplasia; BVS: Virtual Health Library, Brasil; CHMP: Com- for active components in Equisetum arvense extract. Rom Biotechnol Lett.
2013;18:8115-31.
mittee on Herbal Medicinal Products; EA: Equisetum arvense; EMA:
22.  Asgharikhatooni A, Bani S, Hasanpoor S, Alizade SM, Javadzadeh Y. The effect
European Medicines Agency; FAPEG: Fundação de Amparo à Pesquisa of Equisetum arvense (horse tail) ointment on wound healing and pain intensity
do Estado de Goiás; HPCH: Hydroxypropyl chitosan; HPLC: High per- after episiotomy: A randomized placebo-controlled trial. Iran Red Crescent Med
J. 2015;17(3):e25637.
formance liquid chromatography; HPLC-DAD: HPLC coupreld winth
23.  Gallo FR, Multari G, Federici E, Palazzino G, Giambenedetti M, Petitto V, et al.
diode array detector; HPTLC: High performance thin-layer chromatog- Chemical fingerprinting of Equisetum arvense L. using HPTLC densitometry
raphy; IP: Intraperitoneal administration route; IPSS: International Pros- and HPLC. Nat Prod Res. 2011;25(13):1261-70.
tate Symptom Score; ISSR-PCR: Inter-simple sequence repeats; LD50: 24.  Durón RR, Almaguer LC, Garza-Juárez AJ, Luz M, Cavazos S, Waksman-de-
Torres N. Development and validation of thin-layer chromatographic methods
Lethal median dose; LILACS: Latin American and Caribbean Health for quality control of herbal products. Acta Chromatogr. 2009;21(2):203-15.
Sciences Literature; Qmax: Maximum urinary flow rate; QOL: Quality of 25.  Brune T, Thiv M, Haas K. Equisetum (Equisetaceae) species or hybrids? ISSR
life; RDC: Collegiate Board Regulation of Brasil of the National Health fingerprinting profiles help improve diagnoses based on morphology and anatomy.
Plant Syst Evol. 2008;274(1-2):67-81.
Surveillance Agency; REEDA: Redness, Edema, Ecchymosis, Discharge
26.  Cordova E, Morganti L, Rodriguez C. Possible drug–herb interaction between
and Approximation of the Edges Scale; RENISUS: Brazilian National herbal supplement containing horsetail (Equisetum arvense) and antiretroviral
List of Medicinal Plants of Interest to the Unified Health System; ROS: drugs: Report of 2 cases. J Int Assoc Provid AIDS Care. 2017;16(1):11-3.
Reactive Oxygen Species; Ru: Residual Urine; SciELO: Scientific Elec- 27.  European commission. Pilot project: Proposal for approbation of basic substances,
in the context of regulation (EC) Nº 1107/2009 Equisetum arvense basic
tronic Library Online; TEAC: Total antioxidant capacity; TLC: Thin- substance application. 2012. [cited 2018 Dec 20]. Available from: http://www.
layer chromatography; V: Prostate volume; VAS: Visual Analog Scale. itab.asso.fr/downloads/com-intrants/4096_dar-4p_rapport-technique_final_an-
nexes.pdf.
REFERENCES 28.  Agência Nacional de Vigilância Sanitária-ANVISA. Memento Fitoterápico da Farma-
copéia Brasileira. Brasília (Brasil): Agência Nacional de Vigilância Sanitária. 2016.
1. Asgarpanah J, Roohi E. Phytochemistry and pharmacological properties of 29.  29. Kotwal SD, Badole SR. Anabolic therapy with Equisetum arvense along with
Equisetum arvense L. J Med Plant Res. 2012;6(21):3689-93. bone mineralising nutrients in ovariectomized rat model of osteoporosis. Indian
2.  European Medicines Agency. Assessment report on Equisetum arvense L. J Pharmacol. 2016;48(3):312-5.
herb. [updated 2016 Feb 2; cited 2019 Jan 10]. 2016;1-36. Available from: https:// 30.  Gründemann C, Lengen K, Sauer B, Garcia-Käufer M, Zehl M, Huber R.
www.ema.europa.eu/en/documents/herbal-report/final-assessment-report- Equisetum arvense (common horsetail) modulates the function of inflammatory
equisetum-arvense-l-herba_en.pdf immunocompetent cells. BMC Complement Altern Med. 2014;14:283.
3.  Baracho NC, Vicente BB, Arruda GD, Sanches BC, Brito J. Study of acute 31.  Jain R, Kosta S, Tiwari A. Ayurveda and urinary tract infections. J Young Pharm.
hepatotoxicity of Equisetum arvense L. in rats. Acta Cir Bras. 2009;24(6):449-53. 2010;2(3):337.
4.  Mello M, Budel JM. Equisetum L.(Equisetaceae): uma revisão. Cad Esc de Sau. 32.  Carneiro DM, Freire RC, Honório TCD, Zoghaib I, Cardoso FFSS, Tresvenzol
2013;1:1-15. LMF, et al. Randomized, double-blind clinical trial to assess the acute diuretic
5.  MS elabora Relação de Plantas Medicinais de Interesse ao SUS. Brasília: effect of Equisetum arvense (field horsetail) in healthy volunteers. Evid Based
Ministério da Saúde do Brasil. 2009. [udated 2009 Mar 3; cited 2019 Feb 15]. Complement Alternat Med. 2014;2014:760683.
Available from: http://bvsms.saude.gov.br/bvs/sus/pdf/marco/ms_relacao_plan- 33.  Aswal BS, Bhakuni DS, Goel AK, Kar K, Mehrotra BN. Screening of indian plants
tas_medicinais_sus_0603.pdf. for biological activity: Part XI. Indian J Exp Biol. 1984;22(6):487-504.
6.  Agência Nacional de Vigilância Sanitária. Resolução RDC nº. 26, de 13 de maio 34.  Oka M, Tachibana M, Noda K, Inoue N, Tanaka M, Kuwabara K. Relevance of
de 2014. Dispõe sobre o registro de medicamentos fitoterápicos e o registro e anti-reactive oxygen species activity to anti-inflammatory activity of components
a notificação de produtos tradicionais fitoterápicos. Diário Oficial da União 14 of Eviprostat®, a phytotherapeutic agent for benign prostatic hyperplasia.
mai. Seção 1. 2014. Phytomedicine. 2007;14(7-8):465-72.
7. Brasil. Agência Nacional de Vigilância Sanitária (ANVISA). Bulário Eletrônico. 2018. 35.  Song Y, Li NC, Wang XF, Ma LL, Wan B, Hong BF, et al. Clinical study of Eviprostat
[cited 2018 Jun 20]. Available from:http://portal.anvisa.gov.br/bulario-eletronico1. for the treatment of benign prostatic hyperplasia. Zhonghua Nan Ke Xue.
8.  Weberling F, Schwantes HO. Taxonomia Vegetal. São Paulo: Editora Pedagógica 2005;11(9):674-6.
Universitária. 1986. 36.  Tamaki M, Nakashima M, Nishiyama R, Ikeda H, Hiura M, Kanaoka T, et al.
9.  Tropicos.org. Missouri Botanical Garden. 1988. [update 1988 Apr 16; cited 2019 Assessment of clinical usefulness of Eviprostat for benign prostatic hyperplasia-
Feb 15]. Available from: http://www.tropicos.org/Image/88001. comparison of Eviprostat tablet with a formulation containing two-times more
active ingredients. Hinyokika Kiyo. 2008;54(6):435-45.
10.  Jermy AC, Page CN, Acock PJ. Equisetum. Plant Crib. London: Botanical
Society of the British Isles. 1998;1-8. 37.  Hedaya R. Five herbs plus thiamine reduce pain and improve functional mobility
in patients with pain: A pilot study. Altern Ther Health Med. 2017;23(1):14-9.
11. Alves IC, Freitas TMB, Luna AF, Luz EWM. Atividade antioxidante do chá da
Equisetum arvense L. In: IV CONNEPI; Pará, Brasil. 2009. 38.  DiChiacchio N, Restrepo MVS. Eficácia e segurança de uma formulação tópica
em pacientes com síndrome das unhas frágeis. Estudo randomizado, cego
12.  Sandhu NS, Kaur S, Chopra D. Equisetum arvense: Pharmacology and
simples, cruzado e controlado. Surg Cosmet Dermatol. 2015;7(1):26-32.
phytochemistry–a review. Asian J Pharm Clin Res. 2010;3(3):146-50.
39.  Cantoresi F, Caserini M, Bidoli A, Maggio F, Marino R, Carnevale C, et al.
13.  Al-Snafi AE. The pharmacology of Equisetum arvense-A review. IOSR J Pharm. Randomized controlled trial of a water-soluble nail lacquer based on hydroxypro-
2017;7(2):31-42. pyl-chitosan (HPCH), in the management of nail psoriasis. Clin Cosmet Investig
14.  Gallardo-Pérez JC, Esparza-Aguilar ML, Gómez-Campos A. Importancia etnobo- Dermatol. 2014;7:185-90.
tánica de una planta vascular sin semilla en México: Equisetum. Polibotanica. 40.  Schoendorfer N, Sharp N, Seipel T, Schauss AG, Ahuja KDK. Urox containing
2006;21:61-74. concentrated extracts of Crataeva nurvala stem bark, Equisetum arvense stem
15.  Vivancos J, Deshmukh R, Grégoire C, Rémus-Borel W, Belzile F, Bélanger RR. and Lindera aggregata root, in the treatment of symptoms of overactive bladder
Identification and characterization of silicon efflux transporters in horsetail and urinary incontinence: A phase 2, randomised, double-blind placebo
(Equisetum arvense). J Plant Physiol. 2016;200:82-9. controlled trial. BMC Complement Altern Med. 2018;18(1):42. 
16.  Sola-Rabada A, Rinck J, Belton DJ, Powell AK, Perry CC. Isolation of a wide 41.  DosSantos JJG, DoMonte FHM, Blanco MM, Lanziotti VMNB, Maia FD,
range of minerals from a thermally treated plant: Equisetum arvense, a Mare’s Leal LKA. Cognitive enhancement in aged rats after chronic administration
tale. J Biol Inorg Chem. 2016;21(1):101-12. of Equisetum arvense L. with demonstrated antioxidant properties in vitro.
17. Qureshi MN, Stecher G, Bonn GK. Quantification of polyphenolic compounds Pharmacol Biochem Behav. 2005;81(3):593-600.
and flavonoids in Achillea millefolium and Equisetum arvense. Pak J Pharm Sci. 42.  DosSantos JJG, Blanco MM, DoMonte FHM, Russi M, Lanziotti VMNB, Leal
2016;29(5):1519-23. LKAM, et al. Sedative and anticonvulsant effects of hydroalcoholic extract of

56 Pharmacognosy Reviews, Vol 13, Issue 26, Jul-Dec, 2019


Carneiro, et al.: Equisetum arvense: New Medical Evidences

Equisetum arvense. Fitoterapia. 2005;76(6):508-13. extract: phenolic composition and antifungal and antimycotoxigenic effect
43.  Badole S, Kotwal S. Equisetum arvense: Ethanopharmacological and phyto- against Aspergillus flavus and Fusarium verticillioides in stored maize. J Sci
chemical review with reference to osteoporosis. Int J Pharm Sci Health Care. Food Agric. 2013;93(9):2248-53.
2014;1:131-41. 67.  Mekhfi H, El Haouari M, Legssyer A, Bnouham M, Aziz M, Atmani F, et al.
44.  Secretaria de Estado de Saúde do Rio de Janeiro. Resolução SES/RJ no. 1757, Platelet anti-aggregant property of some Moroccan medicinal plants. J Ethno-
de 18 de fevereiro de 2002. Contra-indica o uso de plantas medicinais no pharmacol. 2004;94(2-3):317-22.
âmbito do estado do Rio de Janeiro e dá outras providências. Diário Oficial do 68.  Goun EA, Petrichenko VM, Solodnikov SU, Suhinina TV, Kline MA, Cunningham G,
Estado do Rio de Janeiro 20 fev. Parte I. 2002. et al. Anticancer and antithrombin activity of Russian plants. J Ethnopharmacol.
45.  Luengo MTL. Plantas medicinales con acción diurética. Offarm: Farmacia y 2002;81(3):337-42.
Sociedad. 2001;20(1):116-20. 69.  Yoshinobu Y. Antitumor activity of crude protein extracted from Equisetum
46.  Al-Badri HB, Al-Ani WMK, Naser AMAG. Detection of nicotine in Equisetum arvense Linn’e. Journal of Analytical Bio-Science. 1992;22:421-4.
arvense grown naturally in Iraq. Al-Mustansiriyah Journal for Pharmaceutical 70.  Alexandru V, Petrusca DN, Gille E. Investigation of pro-apoptotic activity of
Sciences. 2016;16(2):40-4. Equisetum arvense L. water extract on human leukemia U 937 cells. Rom
47.  Graefe EU, Veit M. Urinary metabolites of flavonoids and hydroxycinnamic acids Biotechnol Lett. 2007;12(2):3139-48.
in humans after application of a crude extract from Equisetum arvense. 71.  AlMohammed HI, Paray BA, Rather IA. Anticancer activity of EA1 extracted
Phytomedicine. 1999;6(4):239-46. from Equisetum arvense. Pak J Pharm Sci. 2017;30(5(Supplementary)):1947-50.
48.  Myagmar BE, Aniya Y. Free radical scavenging action of medicinal herbs from 72.  Sakurai N, Iizuka T, Nakayama S, Funayama H, Noguchi M, Nagai M. [Vasorelaxant
Mongolia. Phytomedicine. 2000;7(3):221-9. activity of caffeic acid derivatives from Cichorium intybus and Equisetum
49.  Nagai T, Myoda T, Nagashima T. Antioxidative activities of water extract and ethanol arvense]. Yakugaku Zasshi. 2003;123(7):593-8.
extract from field horsetail (tsukushi) Equisetum arvense L. Food Chem. 73.  Oh H, Kim DH, Cho JH, Kim YC. Hepatoprotective and free radical scavenging
2005;91(3):389-94. activities of phenolic petrosins and flavonoids isolated from Equisetum arvense.
50.  Stajner D, Popovic BM, Canadanovic-Brunet J, Boza P. Free radical scavenging J Ethnopharmacol. 2004;95(2-3):421-4.
activity of three Equisetum species from Fruska gora mountain. Fitoterapia. 74.  Ferraz M, Pereira A, Lopes M, Fernandes M. Equisetum arvense: avaliação das
2006;77(7-8):601-4. possibilidades de aplicação na regeneração óssea. Revista Da Faculdade De
51.  Milovanovic V, Radulovic N, Todorovic Z, Stankovic M, Stojanovic G. Antioxidant, Ciências Da Saúde. 2008;5:136-45.
antimicrobial and genotoxicity screening of hydro-alcoholic extracts of five 75.  Raczuk J, Biardzka E, Daruk J. Zawartosc Ca, Mg, Fe i Cu w wybranych
serbian Equisetum species. Plant Foods Hum Nutr. 2007;62(3):113-9. gatunkach ziół i ich naparach. Rocz Panstw Zakl Hig. 2008;59(1):33-40.
52.  Mimica-Dukic N, Simin N, Cvejic J, Jovin E, Orcic D, Bozin B. Phenolic 76.  Bye R, Thingstad SF, Paulsen BS. Horsetail (Equisetum spp,) as a source of
compounds in field horsetail (Equisetum arvense L.) as natural antioxidants. silicon supplement in human nutrition—a myth?. J Herbs Spices Med Plants.
Molecules. 2008;13(7):1455-64. 2010;16(2):119-25.
53.  DaSilva WS, DoCarmo DR. Comportamento voltamétrico do ácido ascórbico 77.  Law C, Exley C. New insight into silica deposition in horsetail (Equisetum
em presença da erva Equisetum arvense. 2009. Available from: https:// arvense). BMC Plant Biol. 2011;11:112.
pt.scribd.com/document/59485585/acido-ascorbico.
78.  Saeed BQ, Hassan HF, Arteen HI. Effect of some medical plant extracts on
54.  Stajner D, Popovic BM, Canadanovic-Brunet J, Anackov G. Exploring Equisetum metabolism of Leishmania tropica promastigotes in vitro. J Med Microbiol
arvense L., Equisetum ramosissimum L. and Equisetum telmateia L. as sources Diagn. 2014;3(4):165.
of natural antioxidants. Phytother Res. 2009;23(4):546-50.
79.  Singh N, Kaur S, Bedi PMS, Kaur D. Anxiolytic effects of Equisetum arvense
55.  Cetojevic-Simin DD, Canadanovic-Brunet JM, Bogdanovic GM, Djilas SM, Linn. extracts in mice. Indian J Exp Biol. 2011;49(5):352-6.
Cetkovic GS, Tumbas VT, et al. Antioxidative and antiproliferative activities of
different horsetail (Equisetum arvense L.) extracts. J Med Food. 2010;13(2):452-9. 80.  Sarris J, McIntyre E, Camfield DA. Plant-based medicines for anxiety disorders,
part 1. CNS Drugs. 2013;27(3):207-19.
56.  Huh MK, Han MD. Inhibitory effect of hyaluronidase and DPPH radical scavening
activity using extraction of Equisetum arvens. Eur J Adv Res Biol Life Sci. 81.  Safiyeh S, Fathallah FB, Vahid N, Hossine N, Habib SS. Antidiabetic effect of
2015;3(2):47-51. Equisetum arvense L. (Equisetaceae) in streptozotocin-induced diabetes in
male rats. Pak J Biol Sci. 2007;10(10):1661-6.
57.  Husson GP, Vilagines R, Delaveau P. Research into antiviral properties of a few
natural extracts [Geranium robertsianum, Morus alba, Adianthum capillus 82.  Soleimani S, Fathiazarbaijani F, Nejati V, Shojaei S, Nanghshbandi N. Effect of
veneris, Scrophularia aquatica, Juglans regia, Rauwolfia serpentina, Equisetum Equisetum arvense L.(Equisetaceae) in microalbuminuria and creatinine
arvense, Ranunculus repens, Ailanthus glandulosa, Polypodium aureum, excretion in streptozotocin-induced diabetes in male rats. Int J Pharmacol.
Polypodium vulgare, Dryopteris filix max]. Ann Pharm Fr. 1986;44:41-8. 2007;3:155-9.
58.  Suganda AG, Amoros M, Girre L, Fauconnier B. Effets inhibiteurs de quelques 83.  DoMonte FHM, DosSantos JJG, Russi M, Lanziotti VMNB, Leal LKAM,
extraites bruts et semi purifiés de plantes indigènes françaises sur la multiplica- Cunha GMA. Antinociceptive and anti-inflammatory properties of the hydroal-
tion de l’herpesvirus humain 1 et du poliovirus humain 2 en culture cellulaire. J coholic extract of stems from Equisetum arvense L. in mice. Pharmacol Res.
Nat Prod. 1983;46(5):626-32. 2004;49(3):239-43.
59.  Guerin J, Reveillere H. Antifungal activity of plant extracts used in therapy. 1: 84.  Ozay Y, Ozyurt S, Guzel S, Cimbiz A, Olgun EG, Cayci MK. Effects of Equisetum
Study of 41 plant extracts against 9 fungi species [Saccharomyces pastorianus, arvense ointment on dermal wound healing in rats. Wounds. 2010;22(10):261-7.
Candida albicans, Rhizopus nigricans, Aspergillus niger, Aspergillus fumigatus, 85.  Hayat A, Temamogullari F, Yilmaz R, Karabulut O. Effect of Equisetum arvense
Botrytis cinerea, Penicillium digitatum, Fusarium oxysporum, Trichophyton on wound contraction of full-thicnes skin wounds in rabbits. Journal of Animal
mentagrophytes]. Ann Pharm Fr. 1984;42:553-9. and Veterinary Advances. 2011;10(1):81-3.
60.  Heisey R, Gorham BK. Antimicrobial effects of plant extracts on Streptococcus 86.  Ozay Y, Kasim CM, Guzel-Ozay S, Cimbiz A, Gurlek-Olgun E, Sabri OM. Effects
mutans, Candida albicans, Trichophyton rubrum and other micro‐organisms. of Equisetum arvense ointment on diabetic wound healing in rats. Wounds.
Lett Appl Microbiol. 1992;14(4):136-9. 2013;25(9):234-41.
61.  Radulovic N, Stojanovic G, Palic R. Composition and antimicrobial activity of 87.  Grases F, Melero G, Costa-Bauza A, Prieto R, March JG. Urolithiasis and
Equisetum arvense L. essential oil. Phytother Res. 2006;20(1):85-8. phytotherapy. Int Urol Nephrol. 1994;26(5):507-11.
62.  Geetha RV, Lakshmi T, Roy A. In vitro evaluation of antibacterial activity of 88.  Crescenti A, Puiggros F, Colome A, Poch JA, Caimari A, Bas JM, et al. Efecto
Equisetum arvense Linn on urinary tract pathogens. Int J Pharm Pharm Sci. antiurolitiásico de una formulación de las plantas Herniaria glabra, Agropyron
2011;3(4):323-5. repens, Equisetum arvense y Sambucus nigra (Herbensurina®) en un modelo
63.  Pereira CB, Gomes PS, Costa‐Rodrigues J, Palmas RA, Vieira L, Ferraz MP, et al. experimental de nefrolitiasis en ratas. Arch Esp Urol. 2015;68(10):739-49.
Equisetum arvense hydromethanolic extracts in bone tissue regeneration: in vitro 89.  Zhang H, Li N, Li K, Li P. Effect of ethanol root extract of Equisetum arvense (L)
osteoblastic modulation and antibacterial activity. Cell Prolif. 2012;45(4):386-96. on urinary bladder activity in rats and analysis of principal plant constituents.
64.  Wojnicz D, Kucharska AZ, Sokół-Łetowska A, Kicia M, Tichaczek-Goska D. Trop J Pharm Res. 2015;14(8):1451-8.
Medicinal plants extracts affect virulence factors expression and biofilm formation 90.  Rebuelta M, Fernández MGS, DelBarrio LSR. Estudio del efecto diurético de
by the uropathogenic Escherichia coli. Urol Res. 2012;40(6):683-97. Equisetum arvense L., Bidens aurea Aiton Sherff., Micromeria fruticosa
65.  DeOliveira JR, Vilela PGF, DeOliveira FE, Belato KK, Carvalho CAT, Jorge AOC, L., Spergularia rubra L., Cynodon dactylon L. Anal Inst Bot Cavanilles.
et al. Antifungal effect of plant extracts on Candida albicans biofilm on acrylic 1978;34(2):703-14.
resin. Braz Dent Sci. 2013;16(3):77-83. 91.  Bakke ILF, Hillestad B. The diuretic effect of Equisetum arvense in rats. Medd
66.  Garcia D, Ramos AJ, Sanchis V, Marín S. Equisetum arvense hydro‐alcoholic Norsk Farm Selsk. 1980;42:9-14.

Pharmacognosy Reviews, Vol 13, Issue 26, Jul-Dec, 2019 57


Carneiro, et al.: Equisetum arvense: New Medical Evidences

GRAPHICAL ABSTRACT

ABOUT AUTHORS

Danilo Maciel Carneiro, MD, Ph.D, Reference Ana Luiza Lima Sousa, RN, Ph.D, Hypertension
Center in Integrative and Complementary Medi- League - School of Medicine, Federal University
cine of the State Health Department of Goiás. of Goiás (UFG). Goiânia – Goiás – Brazil.
Goiânia – Goiás – Brazil.

Thiago Veiga Jardim, MD, Ph.D, Hypertension Luiz Carlos da Cunha, Pharm.D, Ph.D, NEPET-
League - School of Medicine, Federal University UFG – Toxicopharmacological Studies and Re-
of Goiás (UFG). Goiânia – Goiás – Brazil. search Group (School of Pharmacy-UFG).

Ymara Cássia Luciana Araújo, RN, Hyperten- Hérica Núbia Cardoso Cirilo, Pharm., M.Sc,
sion League - School of Medicine, Federal Uni- NEPET-UFG – Toxicopharmacological Studies and
versity of Goiás (UFG). Goiânia – Goiás – Brazil. Research Group (School of Pharmacy-UFG).

Ana Carolina Arantes, RN, M.Sc, Hypertension Maria Teresa Freitas Bara, Pharm.D, Ph.D,
League - School of Medicine, Federal University NEPET-UFG – Toxicopharmacological Studies and
of Goiás (UFG). Goiânia – Goiás – Brazil. Research Group (School of Pharmacy-UFG).

Andrea Cristina de Sousa, RN, M.Sc, Hyperten- Paulo César Brandão da Veiga Jardim, MD,
sion League - School of Medicine, Federal Uni- Ph.D, Hypertension League - School of Medi-
versity of Goiás (UFG). Goiânia – Goiás – Brazil. cine, Federal University of Goiás (UFG). Goiânia
– Goiás – Brazil.

Weimar Kunz Sebba Barroso, MD, Ph.D, Hy-


pertension League - School of Medicine, Fed-
eral University of Goiás (UFG). Goiânia – Goiás
– Brazil.

Cite this article: Carneiro DM, Jardim TV, Araújo YCL, Arantes AC, Sousa AC, et al. Equisetum arvense: New Evidences Supports
Medical use in Daily Clinic. Pharmacog Rev. 2019;13(26):50-8.

58 Pharmacognosy Reviews, Vol 13, Issue 26, Jul-Dec, 2019

You might also like